1
|
Abramova O, Zorkina Y, Goncharov D, Abbazova E, Baranets M, Berdalin A, Ushakova V, Zakurazhnaya V, Morozova I, Ochneva A, Pavlov K, Andreyuk D, Kostyuk G, Morozova A. Association of neurobiological and immune serum biomarkers with Toxoplasma gondii infection in patients with schizophrenia. Parasitol Res 2025; 124:53. [PMID: 40397143 PMCID: PMC12095432 DOI: 10.1007/s00436-025-08498-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/28/2025] [Indexed: 05/22/2025]
Abstract
Some studies suggest that Toxoplasma gondii infection may increase the risk of developing schizophrenia. Determining changes in blood biomarker concentrations may provide new insights into the underlying mechanisms associated with Toxoplasma gondii infection in schizophrenia. The aim of the study was to evaluate the concentrations of several serum neurobiological and immune parameters and to identify changes in their concentrations that could potentially be indicators of psychopathologic changes in infection. The concentration of biomarkers was determined in serum from patients with schizophrenia (uninfected n = 50, infected n = 30) and from mentally healthy volunteers (uninfected n = 51, infected n = 29) using multiplex analysis. A number of psychometric scales have been applied to assess the cognitive functioning. No significant associations were between schizophrenia and Toxoplasma gondii infection (p = 0.54; OR = 1.18; 95% CI = 0.69-2.01). However, infected patients with schizophrenia had more severe cognitive impairment compared to uninfected schizophrenia patients (PDQ-20). The group of biomarkers has been identified whose concentration changes were observed only between Toxoplasma gondii-infected healthy individuals and individuals with schizophrenia (neurobiological indicators KLK6, UCHL1, Amyloid beta 1-42 and neurogranin; anti-inflammatory cytokine IL-10; chemokines IL-8 and MIP-1 beta), but not between uninfected groups. The hypothesis was proposed that it is possible to use these indices as indicators of the development of schizophrenic psychopathology in Toxoplasma gondii infection. The associations of blood biomarker concentrations with IgA and IgM antibody levels (chemokine RANTES) and with schizophrenia symptoms (hormone-like messenger KLK6; chemokines IP-10 and GRO alpha) were found. Toxoplasma gondii reactivation leads to a decrease in negative symptomatology and reduced FGF-21 levels in patients with schizophrenia, and increased CNTF and NGF beta levels compared to the latent form.
Collapse
Affiliation(s)
- Olga Abramova
- Mental-Health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Zagorodnoe Highway 2, 115191, Moscow, Russia.
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky Per. 23, 119034, Moscow, Russia.
| | - Yana Zorkina
- Mental-Health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Zagorodnoe Highway 2, 115191, Moscow, Russia
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky Per. 23, 119034, Moscow, Russia
| | - Dmitry Goncharov
- "National Research Center for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya" of the Ministry of Health of the Russian Federation, Gamaleyi Street, 18, 123098, Moscow, Russia
| | - Evgenia Abbazova
- "National Research Center for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya" of the Ministry of Health of the Russian Federation, Gamaleyi Street, 18, 123098, Moscow, Russia
| | - Marina Baranets
- "National Research Center for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya" of the Ministry of Health of the Russian Federation, Gamaleyi Street, 18, 123098, Moscow, Russia
| | - Alexander Berdalin
- Mental-Health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Zagorodnoe Highway 2, 115191, Moscow, Russia
| | - Valeriya Ushakova
- Mental-Health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Zagorodnoe Highway 2, 115191, Moscow, Russia
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky Per. 23, 119034, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Valeria Zakurazhnaya
- Mental-Health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Zagorodnoe Highway 2, 115191, Moscow, Russia
| | - Irina Morozova
- Mental-Health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Zagorodnoe Highway 2, 115191, Moscow, Russia
| | - Alexandra Ochneva
- Mental-Health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Zagorodnoe Highway 2, 115191, Moscow, Russia
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky Per. 23, 119034, Moscow, Russia
| | - Konstantin Pavlov
- Mental-Health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Zagorodnoe Highway 2, 115191, Moscow, Russia
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky Per. 23, 119034, Moscow, Russia
| | - Denis Andreyuk
- Mental-Health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Zagorodnoe Highway 2, 115191, Moscow, Russia
| | - Georgy Kostyuk
- Mental-Health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Zagorodnoe Highway 2, 115191, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
- Russian Biotechnological University (ROSBIOTECH), Moscow, Russia
- Department of Psychiatry and Psychosomatics, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119435, Moscow, Russia
| | - Anna Morozova
- Mental-Health Clinic No. 1 Named After N.A. Alexeev of Moscow Healthcare Department, Zagorodnoe Highway 2, 115191, Moscow, Russia
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky Per. 23, 119034, Moscow, Russia
| |
Collapse
|
2
|
Luo X, Yang X, Tan S, Zhang Y, Liu Y, Tian X, Huang Y, Zhou Y, He C, Yin K, Xu D, Li X, Sun F, Tang R, Cao J, Zheng K, Yu Y, Pan W. Gut microbiota mediates anxiety-like behaviors induced by chronic infection of Toxoplasma gondii in mice. Gut Microbes 2024; 16:2391535. [PMID: 39182245 PMCID: PMC11346544 DOI: 10.1080/19490976.2024.2391535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/05/2024] [Accepted: 08/08/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Chronic infection with the neurotropic parasite Toxoplasma gondii (T. gondii) can cause anxiety and gut microbiota dysbiosis in hosts. However, the potential role of gut microbiota in anxiety induced by the parasite remains unclear. METHODS C57BL/6J mice were infected with 10 cysts of T. gondii. Antibiotic depletion of gut microbiota and fecal microbiota transplantation experiments were utilized to investigate the causal relationship between gut microbiota and anxiety. Anxiety-like behaviors were examined by the elevated plus maze test and the open field test; blood, feces, colon and amygdala were collected to evaluate the profiles of serum endotoxin (Lipopolysaccharide, LPS) and serotonin (5-hydroxytryptamine, 5-HT), gut microbiota composition, metabolomics, global transcriptome and neuroinflammation in the amygdala. Furthermore, the effects of Diethyl butylmalonate (DBM, an inhibitor of mitochondrial succinate transporter, which causes the accumulation of endogenous succinate) on the disorders of the gut-brain axis were evaluated. RESULTS Here, we found that T. gondii chronic infection induced anxiety-like behaviors and disturbed the composition of the gut microbiota in mice. In the amygdala, T. gondii infection triggered the microglial activation and neuroinflammation. In the colon, T. gondii infection caused the intestinal dyshomeostasis including elevated colonic inflammation, enhanced bacterial endotoxin translocation to blood and compromised intestinal barrier. In the serum, T. gondii infection increased the LPS levels and decreased the 5-HT levels. Interestingly, antibiotics ablation of gut microbiota alleviated the anxiety-like behaviors induced by T. gondii infection. More importantly, transplantation of the fecal microbiota from T. gondii-infected mice resulted in anxiety and the transcriptomic alteration in the amygdala of the antibiotic-pretreated mice. Notably, the decreased abundance of succinate-producing bacteria and the decreased production of succinate were observed in the feces of the T. gondii-infected mice. Moreover, DBM administration ameliorated the anxiety and gut barrier impairment induced by T. gondii infection. CONCLUSIONS The present study uncovers a novel role of gut microbiota in mediating the anxiety-like behaviors induced by chronic T. gondii infection. Moreover, we show that DBM supplementation has a beneficial effect on anxiety. Overall, these findings provide new insights into the treatment of T. gondii-related mental disorders.
Collapse
Affiliation(s)
- Xiaotong Luo
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shimin Tan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yongsheng Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yunqiu Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaokang Tian
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yingting Huang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuying Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Cheng He
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kun Yin
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong, China
| | - Daxiang Xu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiangyang Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fenfen Sun
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, Shanghai, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, Shanghai, China
| |
Collapse
|
3
|
Ju S, Shin Y, Han S, Kwon J, Choi TG, Kang I, Kim SS. The Gut-Brain Axis in Schizophrenia: The Implications of the Gut Microbiome and SCFA Production. Nutrients 2023; 15:4391. [PMID: 37892465 PMCID: PMC10610543 DOI: 10.3390/nu15204391] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Schizophrenia, a severe mental illness affecting about 1% of the population, manifests during young adulthood, leading to abnormal mental function and behavior. Its multifactorial etiology involves genetic factors, experiences of adversity, infection, and gene-environment interactions. Emerging research indicates that maternal infection or stress during pregnancy may also increase schizophrenia risk in offspring. Recent research on the gut-brain axis highlights the gut microbiome's potential influence on central nervous system (CNS) function and mental health, including schizophrenia. The gut microbiota, located in the digestive system, has a significant role to play in human physiology, affecting immune system development, vitamin synthesis, and protection against pathogenic bacteria. Disruptions to the gut microbiota, caused by diet, medication use, environmental pollutants, and stress, may lead to imbalances with far-reaching effects on CNS function and mental health. Of interest are short-chain fatty acids (SCFAs), metabolic byproducts produced by gut microbes during fermentation. SCFAs can cross the blood-brain barrier, influencing CNS activity, including microglia and cytokine modulation. The dysregulation of neurotransmitters produced by gut microbes may contribute to CNS disorders, including schizophrenia. This review explores the potential relationship between SCFAs, the gut microbiome, and schizophrenia. Our aim is to deepen the understanding of the gut-brain axis in schizophrenia and to elucidate its implications for future research and therapeutic approaches.
Collapse
Affiliation(s)
- Songhyun Ju
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoonhwa Shin
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Juhui Kwon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Tae Gyu Choi
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Insug Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
4
|
Naufel MF, Truzzi GDM, Ferreira CM, Coelho FMS. The brain-gut-microbiota axis in the treatment of neurologic and psychiatric disorders. ARQUIVOS DE NEURO-PSIQUIATRIA 2023. [PMID: 37402401 PMCID: PMC10371417 DOI: 10.1055/s-0043-1767818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
The human gut microbiota is a complex ecosystem made of trillions of microorganisms. The composition can be affected by diet, metabolism, age, geography, stress, seasons, temperature, sleep, and medications. The increasing evidence about the existence of a close and bi-directional correlation between the gut microbiota and the brain indicates that intestinal imbalance may play a vital role in the development, function, and disorders of the central nervous system. The mechanisms of interaction between the gut-microbiota on neuronal activity are widely discussed. Several potential pathways are involved with the brain-gut-microbiota axis, including the vagus nerve, endocrine, immune, and biochemical pathways. Gut dysbiosis has been linked to neurological disorders in different ways that involve activation of the hypothalamic-pituitary-adrenal axis, imbalance in neurotransmitter release, systemic inflammation, and increase in the permeability of the intestinal and the blood-brain barrier. Mental and neurological diseases have become more prevalent during the coronavirus disease 2019pandemic and are an essential issue in public health globally. Understanding the importance of diagnosing, preventing, and treating dysbiosis is critical because gut microbial imbalance is a significant risk factor for these disorders. This review summarizes evidence demonstrating the influence of gut dysbiosis on mental and neurological disorders.
Collapse
Affiliation(s)
| | | | | | - Fernando Morgadinho Santos Coelho
- Universidade Federal de São Paulo, Departamento de Psicobiologia, São Paulo SP, Brazil
- Universidade Federal de São Paulo, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil
| |
Collapse
|
5
|
Moradi F, Dashti N, Farahvash A, Baghaei Naeini F, Zarebavani M. Curcumin ameliorates chronic Toxoplasma gondii infection-induced affective disorders through modulation of proinflammatory cytokines and oxidative stress. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:461-467. [PMID: 37009013 PMCID: PMC10008396 DOI: 10.22038/ijbms.2023.68487.14937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/08/2023] [Indexed: 04/04/2023]
Abstract
Objectives Long-term infection with Toxoplasma gondii is associated with affective disorders (i.e., anxiety and depression) in adults. We aimed to explore the effects of curcumin (CR) on anxiety- and depressive-like behaviors in mice infected with T. gondii. Materials and Methods Animals were studied in five groups: Control, Model, Model + CR20, 40, and 80 (with IP injection of 20, 40, and 80 mg/kg CR). T. gondii infection was prolonged for four weeks. The animals were then treated with CR or vehicle for two weeks and evaluated by behavioral tests at the end of the study. Hippocampal levels of oxidative stress biomarkers (superoxide dismutase; SOD, glutathione; GSH, and malondialdehyde; MDA) and gene expression and protein levels of hippocampal proinflammatory mediators (interleukin-1β; IL-1β, IL-6, IL-18, and tumor necrosis factor- α; TNF-α) were determined. Results Behavioral tests confirmed that long-term infection with T. gondii led to anxiety- and depressive-like behaviors. Antidepressant effects of CR were linked to modulation of oxidative stress and cytokine network in the hippocampal region of infected mice. These results showed that CR reduced anxiety and depression symptoms via regulation of oxidative stress and proinflammatory cytokines in the hippocampus of T. gondii-infected mice. Conclusion Therefore, CR can be used as a potential antidepressant agent against T. gondii-induced affective disorders.
Collapse
Affiliation(s)
- Fatemeh Moradi
- School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasrin Dashti
- Department of Clinical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Mitra Zarebavani
- Department of Clinical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Matt SM. Targeting neurotransmitter-mediated inflammatory mechanisms of psychiatric drugs to mitigate the double burden of multimorbidity and polypharmacy. Brain Behav Immun Health 2021; 18:100353. [PMID: 34647105 PMCID: PMC8495104 DOI: 10.1016/j.bbih.2021.100353] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 09/16/2021] [Accepted: 09/18/2021] [Indexed: 12/12/2022] Open
Abstract
The increased incidence of multimorbidities and polypharmacy is a major concern, particularly in the growing aging population. While polypharmacy can be beneficial, in many cases it can be more harmful than no treatment, especially in individuals suffering from psychiatric disorders, who have elevated risks of multimorbidity and polypharmacy. Age-related chronic inflammation and immunopathologies might contribute to these increased risks in this population, but the optimal clinical management of drug-drug interactions and the neuro-immune mechanisms that are involved warrants further investigation. Given that neurotransmitter systems, which psychiatric medications predominantly act on, can influence the development of inflammation and the regulation of immune function, it is important to better understand these interactions to develop more successful strategies to manage these comorbidities and complicated polypharmacy. I propose that expanding upon research in translationally relevant human in vitro models, in tandem with other preclinical models, is critical to defining the neurotransmitter-mediated mechanisms by which psychiatric drugs alter immune function. This will define more precisely the interactions of psychiatric drugs and other immunomodulatory drugs, used in combination, enabling identification of novel targets to be translated into more efficacious diagnostic, preventive, and therapeutic interventions. This interdisciplinary approach will aid in better precision polypharmacy for combating adverse events associated with multimorbidity and polypharmacy in the future.
Collapse
Affiliation(s)
- Stephanie M. Matt
- Drexel University College of Medicine, Department of Pharmacology and Physiology, Philadelphia, PA, USA
| |
Collapse
|
7
|
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: 20] [Impact Index Per Article: 5.0] [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
|
8
|
Kelly JR, Minuto C, Cryan JF, Clarke G, Dinan TG. The role of the gut microbiome in the development of schizophrenia. Schizophr Res 2021; 234:4-23. [PMID: 32336581 DOI: 10.1016/j.schres.2020.02.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 02/22/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a heterogeneous neurodevelopmental disorder involving the convergence of a complex and dynamic bidirectional interaction of genetic expression and the accumulation of prenatal and postnatal environmental risk factors. The development of the neural circuitry underlying social, cognitive and emotional domains requires precise regulation from molecular signalling pathways, especially during critical periods or "windows", when the brain is particularly sensitive to the influence of environmental input signalling. Many of the brain regions involved, and the molecular substrates sub-serving these domains are responsive to life-long microbiota-gut-brain (MGB) axis signalling. This intricate microbial signalling system communicates with the brain via the vagus nerve, immune system, enteric nervous system, enteroendocrine signalling and production of microbial metabolites, such as short-chain fatty acids. Preclinical data has demonstrated that MGB axis signalling influences neurotransmission, neurogenesis, myelination, dendrite formation and blood brain barrier development, and modulates cognitive function and behaviour patterns, such as, social interaction, stress management and locomotor activity. Furthermore, preliminary clinical studies suggest altered gut microbiota profiles in schizophrenia. Unravelling MGB axis signalling in the context of an evolving dimensional framework in schizophrenia may provide a more complete understanding of the neurobiological architecture of this complex condition and offers the possibility of translational interventions.
Collapse
Affiliation(s)
- John R Kelly
- Department of Psychiatry, Trinity College Dublin, Ireland
| | - Chiara Minuto
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Timothy G Dinan
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland.
| |
Collapse
|
9
|
Rosuvastatin revert memory impairment and anxiogenic-like effect in mice infected with the chronic ME-49 strain of Toxoplasma gondii. PLoS One 2021; 16:e0250079. [PMID: 33857221 PMCID: PMC8049280 DOI: 10.1371/journal.pone.0250079] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/30/2021] [Indexed: 11/19/2022] Open
Abstract
The aim of this study was to investigate the effect of rosuvastatin treatment on memory impairment, and anxiogenic-like effects in mice chronically infected with Toxoplasma gondii. For this, Balb/c mice were infected orally with chronic ME-49 strain of Toxoplasma gondii. Oral treatment with rosuvastatin (40mg/kg/day) started on the 51st day post-infection and was performed daily for 21 days. After completion of treatment, anxiety-like effects and locomotion were investigated in the open field (OF) test, whereas novel object recognition (NOR) test was used for evaluation of short- and long-term memory. At the end of the experiments, the brain was collected for Toxoplasma gondii DNA quantification and histopathological analysis. Infection with ME-49 strain decreased the time spent in the center of OF, indicating an anxiogenic effect, without affecting total and peripheral locomotion. Rosuvastatin treatment inhibited the change in the center time. Besides, pharmacological treatment increased total and central locomotion in both non-infected and infected animals. Infection also impaired both short- and long-term memory in the NOR test, and these effects were reverted by rosuvastatin treatment. In addition to effects in behavioral changes, rosuvastatin also reduced parasite load in the brain and attenuated signs of brain inflammation such as perivascular cuffs, inflammatory cell infiltration and tissue damage. These findings indicate for the first time the efficacy of rosuvastatin in treatment of memory impairment and anxiogenic effect evoked by infection with Toxoplasma gondii. These effects might be mediated by reduced cyst load, which in turn decrease inflammation and damage in the brain.
Collapse
|
10
|
Khan IA, Hwang S, Moretto M. Toxoplasma gondii: CD8 T Cells Cry for CD4 Help. Front Cell Infect Microbiol 2019; 9:136. [PMID: 31119107 PMCID: PMC6504686 DOI: 10.3389/fcimb.2019.00136] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/15/2019] [Indexed: 11/13/2022] Open
Abstract
Toxoplasma gondii, an apicomplexan parasite, is a pathogenic protozoan that can infect the central nervous system. In pregnant women, infection can result in congenital problems of the fetus, while in immunocompromised individual it can lead to severe neurological consequences. Although CD8 T cells play an important effector role in controlling the chronic infection, their maintenance is dependent on the critical help provided by CD4 T cells. In a recent study, we demonstrated that reactivation of the infection in chronically infected host is a consequence of CD8 T dysfunction caused by CD4 T cell exhaustion. Furthermore, treatment of chronically infected host with antigen-specific non-exhausted CD4 T cells can restore CD8 T cell functionality and prevent reactivation of the latent infection. The exhaustion status of CD4 T cells is mediated by the increased expression of the transcription factor BLIMP-1, and deletion of this molecule led to the restoration of CD4 T cell function, reversal of CD8 exhaustion and prevention of reactivation of the latent infection. In a recent study from our laboratory, we also observed an increased expression of miR146a levels by CD4 T cells from the chronically infected animals. Recent reports have demonstrated that microRNAs (especially miR146a) has a strong impact on the immune system of T. gondii infected host. Whether these molecules have any role in the BLIMP-1 up-regulation and dysfunctionality of these cells needs to be investigated.
Collapse
Affiliation(s)
- Imtiaz A. Khan
- Department Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, DC, United States
| | | | | |
Collapse
|
11
|
Chronic Toxoplasma gondii Infection Induces Anti- N-Methyl-d-Aspartate Receptor Autoantibodies and Associated Behavioral Changes and Neuropathology. Infect Immun 2018; 86:IAI.00398-18. [PMID: 30037790 DOI: 10.1128/iai.00398-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/13/2018] [Indexed: 12/27/2022] Open
Abstract
Anti-NMDA receptor (NMDAR) autoantibodies have been postulated to play a role in the pathogenesis of NMDAR hypofunction, which contributes to the etiology of psychotic symptoms. Toxoplasma gondii is a pathogen implicated in psychiatric disorders and associated with elevation of NMDAR autoantibodies. However, it remains unclear whether parasite infection is the cause of NMDAR autoantibodies. By using mouse models, we found that NMDAR autoantibody generation had a strong temporal association with tissue cyst formation, as determined by MAG1 antibody seroreactivity (r = 0.96; P < 0.0001), which is a serologic marker for the cyst burden. The presence of MAG1 antibody response, but not T. gondii IgG response, was required for NMDAR autoantibody production. The pathogenic relevance of NMDAR autoantibodies to behavioral abnormalities (blunted response to amphetamine-triggered activity and decreased locomotor activity and exploration) and reduced expression of synaptic proteins (the GLUN2B subtype of NMDAR and PSD-95) has been demonstrated in infected mice. Our study suggests that NMDAR autoantibodies are specifically induced by persistent T. gondii infection and are most likely triggered by tissue cysts. NMDAR autoantibody seroreactivity may be a novel pathological hallmark of chronic toxoplasmosis, which raises questions about NMDAR hypofunction and neurodegeneration in the infected brain.
Collapse
|
12
|
Xiao J, Prandovszky E, Kannan G, Pletnikov MV, Dickerson F, Severance EG, Yolken RH. Toxoplasma gondii: Biological Parameters of the Connection to Schizophrenia. Schizophr Bull 2018; 44:983-992. [PMID: 29889280 PMCID: PMC6101499 DOI: 10.1093/schbul/sby082] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
It is increasingly evident that the brain is not truly an immune privileged site and that cells of the central nervous system are sensitive to the inflammation generated when the brain is fighting off infection. Among the many microorganisms that have access to the brain, the apicomplexan protozoan Toxoplasma gondii has been one of the most studied. This parasite has been associated with many neuropsychiatric disorders including schizophrenia. This article provides a comprehensive review of the status of Toxoplasma research in schizophrenia. Areas of interest include (1) the limitations and improvements of immune-based assays to detect these infections in humans, (2) recent discoveries concerning the schizophrenia-Toxoplasma association, (3) findings of Toxoplasma neuropathology in animal models related to schizophrenia pathogenesis, (4) interactions of Toxoplasma with the host genome, (5) gastrointestinal effects of Toxoplasma infections, and (6) therapeutic intervention of Toxoplasma infections.
Collapse
Affiliation(s)
- Jianchun Xiao
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - Emese Prandovszky
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - Geetha Kannan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Mikhail V Pletnikov
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Faith Dickerson
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD
| | - Emily G Severance
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD,To whom correspondence should be addressed; Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21287 USA; tel: +1-410-614-0004, fax: +1-410-955-3723, e-mail:
| |
Collapse
|
13
|
Abstract
OBJECTIVE The brain-gut-microbiota axis has been put forward as a new paradigm in neuroscience, which may be of relevance to mental illness. The mechanisms of signal transmission in the brain-gut-microbiota axis are complex and involve bidirectional communications that enable gut microbes to communicate with the brain and the brain to communicate with the microbes. This review assesses the potential usefulness and limitations of the paradigm. METHODS A selective literature review was conducted to evaluate the current knowledge in clinical and preclinical brain-gut-microbiota interactions as related to psychiatric disorders. RESULTS Most published studies in the field are preclinical, and there is so far a lack of clinical studies. Preliminary studies in psychiatric populations support the view of a dysbiosis in some conditions, but studies are often small scale and marred by potential confounding variables. Preclinical studies support the view that psychobiotics ("bacteria which when ingested in adequate amounts have a positive mental health benefit") might be of use in treating some patients with mental health difficulties. To date, we have no well-conducted studies in clinical populations, although there are some studies in healthy volunteers. A cocktail of probiotics has been shown to alter brain activity as monitored by functional magnetic resonance imaging, and Bifidobacterium longum was reported to alter brain electrical activity. CONCLUSIONS It has yet to be convincingly demonstrated that the exciting findings of psychobiotic efficacy demonstrated in preclinical models of psychiatric illness will translate to patients.
Collapse
|
14
|
Kelly JR, Minuto C, Cryan JF, Clarke G, Dinan TG. Cross Talk: The Microbiota and Neurodevelopmental Disorders. Front Neurosci 2017; 11:490. [PMID: 28966571 PMCID: PMC5605633 DOI: 10.3389/fnins.2017.00490] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/17/2017] [Indexed: 12/11/2022] Open
Abstract
Humans evolved within a microbial ecosystem resulting in an interlinked physiology. The gut microbiota can signal to the brain via the immune system, the vagus nerve or other host-microbe interactions facilitated by gut hormones, regulation of tryptophan metabolism and microbial metabolites such as short chain fatty acids (SCFA), to influence brain development, function and behavior. Emerging evidence suggests that the gut microbiota may play a role in shaping cognitive networks encompassing emotional and social domains in neurodevelopmental disorders. Drawing upon pre-clinical and clinical evidence, we review the potential role of the gut microbiota in the origins and development of social and emotional domains related to Autism spectrum disorders (ASD) and schizophrenia. Small preliminary clinical studies have demonstrated gut microbiota alterations in both ASD and schizophrenia compared to healthy controls. However, we await the further development of mechanistic insights, together with large scale longitudinal clinical trials, that encompass a systems level dimensional approach, to investigate whether promising pre-clinical and initial clinical findings lead to clinical relevance.
Collapse
Affiliation(s)
- John R Kelly
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Chiara Minuto
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College CorkCork, Ireland.,Department of Anatomy and Neuroscience, University College CorkCork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Timothy G Dinan
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| |
Collapse
|
15
|
Felice VD, O'Mahony SM. The microbiome and disorders of the central nervous system. Pharmacol Biochem Behav 2017; 160:1-13. [PMID: 28666895 DOI: 10.1016/j.pbb.2017.06.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 06/12/2017] [Accepted: 06/26/2017] [Indexed: 02/07/2023]
Abstract
Alterations of the gut microbiota have been associated with stress-related disorders including depression and anxiety and irritable bowel syndrome (IBS). More recently, researchers have started investigating the implication of perturbation of the microbiota composition in neurodevelopmental disorders including autism spectrum disorders and Attention-Deficit Hypersensitivity Disorder (ADHD). In this review we will discuss how the microbiota is established and its functions in maintaining health. We also summarize both pre and post-natal factors that shape the developing neonatal microbiota and how they may impact on health outcomes with relevance to disorders of the central nervous system. Finally, we discuss potential therapeutic approaches based on the manipulation of the gut bacterial composition.
Collapse
Affiliation(s)
- Valeria D Felice
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Siobhain M O'Mahony
- APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
16
|
Sherwin E, Sandhu KV, Dinan TG, Cryan JF. May the Force Be With You: The Light and Dark Sides of the Microbiota-Gut-Brain Axis in Neuropsychiatry. CNS Drugs 2016; 30:1019-1041. [PMID: 27417321 PMCID: PMC5078156 DOI: 10.1007/s40263-016-0370-3] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The role of the gut microbiota in health and disease is becoming increasingly recognized. The microbiota-gut-brain axis is a bi-directional pathway between the brain and the gastrointestinal system. The bacterial commensals in our gut can signal to the brain through a variety of mechanisms, which are slowly being resolved. These include the vagus nerve, immune mediators and microbial metabolites, which influence central processes such as neurotransmission and behaviour. Dysregulation in the composition of the gut microbiota has been identified in several neuropsychiatric disorders, such as autism, schizophrenia and depression. Moreover, preclinical studies suggest that they may be the driving force behind the behavioural abnormalities observed in these conditions. Understanding how bacterial commensals are involved in regulating brain function may lead to novel strategies for development of microbiota-based therapies for these neuropsychiatric disorders.
Collapse
Affiliation(s)
- Eoin Sherwin
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Kiran V Sandhu
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland.
- Department of Anatomy and Neuroscience, University College Cork, Western Gateway Building, Cork, Ireland.
| |
Collapse
|
17
|
Impaired health status and increased incidence of diseases in Toxoplasma-seropositive subjects – an explorative cross-sectional study. Parasitology 2016; 143:1974-1989. [DOI: 10.1017/s0031182016001785] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SUMMARYThe global seroprevalence of latent toxoplasmosis is estimated to be higher than 30%. The presence of slowly dividing parasites in tissue cysts located mainly in immunoprivileged organs was long considered asymptomatic. Recently, many studies have shown that latent Toxoplasma infections could have serious impacts on human health. Here we ran a cross-sectional study in a population of 1486 volunteers. The results showed that 333 infected subjects scored worse than 1153 controls in 28 of 29 health-related variables. Similarly, they reported higher rates of 77 of a list of 134 disorders reported by at least 10 participants of the study. Toxoplasmosis was associated most strongly with musculoskeletal (τ = 0·107, P < 0·0005), followed by neurological (τ = 0·088, P < 0·0005), immune (τ = 0·085, p < 0·0005), metabolic (τ = 0·079, P < 0·0005), respiratory (τ = 0·068, P = 0·0001), allergic (τ = 0·053, P = 0·004), digestive system (τ = 0·052, P = 0·004) and mental health disorders (τ = 0·050, P = 0·008). Results of the present cohort study, along with the previous data from many case-control studies or ecological studies suggest that latent toxoplasmosis represents a large and so far underrated public health problem.
Collapse
|
18
|
Elsheikha HM, Büsselberg D, Zhu XQ. The known and missing links between Toxoplasma gondii and schizophrenia. Metab Brain Dis 2016; 31:749-59. [PMID: 27041387 DOI: 10.1007/s11011-016-9822-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/20/2016] [Indexed: 12/14/2022]
Abstract
Toxoplasma gondii, an intracellular protozoan parasite, has a striking predilection for infecting the Central Nervous System and has been linked to an increased incidence of a number of psychiatric diseases. Several in vitro and in vivo studies have shown that T. gondii infection can affect the structure, bioenergetics and function of brain cells, and alters several host cell processes, including dopaminergic, tryptophan-kynurenine, GABAergic, AKT1, Jak/STAT, and vasopressinergic pathways. These mechanisms underlying the neuropathology of latent toxoplasmosis seem to operate also in schizophrenia, supporting the link between the two disorders. Better understanding of the intricate parasite-neuroglial communications holds the key to unlocking the mystery of T. gondii-mediated schizophrenia and offers substantial prospects for the development of disease-modifying therapies.
Collapse
Affiliation(s)
- Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, Leicestershire, LE12 5RD, UK.
| | - Dietrich Büsselberg
- Weill Cornell Medical College in Qatar, Qatar Foundation - Education City, P.O. Box: 24144, Doha, Qatar
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
| |
Collapse
|
19
|
Sutterland AL, Fond G, Kuin A, Koeter MWJ, Lutter R, van Gool T, Yolken R, Szoke A, Leboyer M, de Haan L. Beyond the association. Toxoplasma gondii in schizophrenia, bipolar disorder, and addiction: systematic review and meta-analysis. Acta Psychiatr Scand 2015; 132:161-79. [PMID: 25877655 DOI: 10.1111/acps.12423] [Citation(s) in RCA: 308] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To perform a meta-analysis on studies reporting prevalence of Toxoplasma gondii (T. gondii) infection in any psychiatric disorder compared with healthy controls. Our secondary objective was to analyze factors possibly moderating heterogeneity. METHOD A systematic search was performed to identify studies into T. gondii infection for all major psychiatric disorders versus healthy controls. Methodological quality, publication bias, and possible moderators were assessed. RESULTS A total of 2866 citations were retrieved and 50 studies finally included. Significant odds ratios (ORs) with IgG antibodies were found in schizophrenia (OR 1.81, P < 0.00001), bipolar disorder (OR 1.52, P = 0.02), obsessive-compulsive disorder (OR 3.4, P < 0.001), and addiction (OR 1.91, P < 0.00001), but not for major depression (OR 1.21, P = 0.28). Exploration of the association between T. gondii and schizophrenia yielded a significant effect of seropositivity before onset and serointensity, but not IgM antibodies or gender. The amplitude of the OR was influenced by region and general seroprevalence. Moderators together accounted for 56% of the observed variance in study effects. After controlling for publication bias, the adjusted OR (1.43) in schizophrenia remained significant. CONCLUSION These findings suggest that T. gondii infection is associated with several psychiatric disorders and that in schizophrenia reactivation of latent T. gondii infection may occur.
Collapse
Affiliation(s)
- A L Sutterland
- Department of Psychiatry, Academic Medical Centre (AMC), Amsterdam, the Netherlands
| | - G Fond
- AP-HP, DHU Pe-PSY, Pôle de Psychiatrie et d'addictologie des Hôpitaux Universitaires H Mondor, INSERM U955, Eq 15 Psychiatrie Translationnelle, Université Paris Est-Créteil, Créteil, France.,Fondation Fondamental, Créteil, France
| | - A Kuin
- Department of Psychiatry, Academic Medical Centre (AMC), Amsterdam, the Netherlands
| | - M W J Koeter
- Department of Psychiatry, Academic Medical Centre (AMC), Amsterdam, the Netherlands
| | - R Lutter
- Departments of Experimental Immunology and Respiratory Medicine, Academic Medical Centre (AMC), Amsterdam, the Netherlands
| | - T van Gool
- Department of Parasitology, Academic Medical Centre (AMC), Amsterdam, the Netherlands
| | - R Yolken
- Stanley Neurovirology Laboratory, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - A Szoke
- AP-HP, DHU Pe-PSY, Pôle de Psychiatrie et d'addictologie des Hôpitaux Universitaires H Mondor, INSERM U955, Eq 15 Psychiatrie Translationnelle, Université Paris Est-Créteil, Créteil, France.,Fondation Fondamental, Créteil, France
| | - M Leboyer
- AP-HP, DHU Pe-PSY, Pôle de Psychiatrie et d'addictologie des Hôpitaux Universitaires H Mondor, INSERM U955, Eq 15 Psychiatrie Translationnelle, Université Paris Est-Créteil, Créteil, France.,Fondation Fondamental, Créteil, France
| | - L de Haan
- Department of Psychiatry, Academic Medical Centre (AMC), Amsterdam, the Netherlands
| |
Collapse
|
20
|
Adaptive Immunity in Schizophrenia: Functional Implications of T Cells in the Etiology, Course and Treatment. J Neuroimmune Pharmacol 2015; 10:610-9. [PMID: 26162591 DOI: 10.1007/s11481-015-9626-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/03/2015] [Indexed: 12/21/2022]
Abstract
Schizophrenia is a severe and highly complex neurodevelopmental disorder with an unknown etiopathology. Recently, immunopathogenesis has emerged as one of the most compelling etiological models of schizophrenia. Over the past few years considerable research has been devoted to the role of innate immune responses in schizophrenia. The findings of such studies have helped to conceptualize schizophrenia as a chronic low-grade inflammatory disorder. Although the contribution of adaptive immune responses has also been emphasized, however, the precise role of T cells in the underlying neurobiological pathways of schizophrenia is yet to be ascertained comprehensively. T cells have the ability to infiltrate brain and mediate neuro-immune cross-talk. Conversely, the central nervous system and the neurotransmitters are capable of regulating the immune system. Neurotransmitter like dopamine, implicated widely in schizophrenia risk and progression can modulate the proliferation, trafficking and functions of T cells. Within brain, T cells activate microglia, induce production of pro-inflammatory cytokines as well as reactive oxygen species and subsequently lead to neuroinflammation. Importantly, such processes contribute to neuronal injury/death and are gradually being implicated as mediators of neuroprogressive changes in schizophrenia. Antipsychotic drugs, commonly used to treat schizophrenia are also known to affect adaptive immune system; interfere with the differentiation and functions of T cells. This understanding suggests a pivotal role of T cells in the etiology, course and treatment of schizophrenia and forms the basis of this review.
Collapse
|
21
|
Deleidi M, Jäggle M, Rubino G. Immune aging, dysmetabolism, and inflammation in neurological diseases. Front Neurosci 2015; 9:172. [PMID: 26089771 PMCID: PMC4453474 DOI: 10.3389/fnins.2015.00172] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 04/28/2015] [Indexed: 12/17/2022] Open
Abstract
As we age, the immune system undergoes a process of senescence accompanied by the increased production of proinflammatory cytokines, a chronic subclinical condition named as “inflammaging”. Emerging evidence from human and experimental models suggest that immune senescence also affects the central nervous system and promotes neuronal dysfunction, especially within susceptible neuronal populations. In this review we discuss the potential role of immune aging, inflammation and metabolic derangement in neurological diseases. The discovery of novel therapeutic strategies targeting age-linked inflammation may promote healthy brain aging and the treatment of neurodegenerative as well as neuropsychiatric disorders.
Collapse
Affiliation(s)
- Michela Deleidi
- Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Hertie Institute for Clinical Brain Research, University of Tübingen Tübingen, Germany
| | - Madeline Jäggle
- Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Hertie Institute for Clinical Brain Research, University of Tübingen Tübingen, Germany
| | - Graziella Rubino
- Department of Internal Medicine II, Center for Medical Research, University of Tübingen Tübingen, Germany
| |
Collapse
|
22
|
Fabiani S, Pinto B, Bonuccelli U, Bruschi F. Neurobiological studies on the relationship between toxoplasmosis and neuropsychiatric diseases. J Neurol Sci 2015; 351:3-8. [DOI: 10.1016/j.jns.2015.02.028] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 01/21/2015] [Accepted: 02/16/2015] [Indexed: 02/02/2023]
|
23
|
Xiao J, Yolken RH. Strain hypothesis of Toxoplasma gondii infection on the outcome of human diseases. Acta Physiol (Oxf) 2015; 213:828-45. [PMID: 25600911 DOI: 10.1111/apha.12458] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 10/13/2014] [Accepted: 01/12/2015] [Indexed: 12/28/2022]
Abstract
The intracellular protozoan Toxoplasma gondii is an exceptionally successful food and waterborne parasite that infects approximately 1 billion people worldwide. Genotyping of T. gondii isolates from all continents revealed a complex population structure. Recent research supports the notion that T. gondii genotype may be associated with disease severity. Here, we (1) discuss molecular and serological approaches for designation of T. gondii strain type, (2) overview the literatures on the association of T. gondii strain type and the outcome of human disease and (3) explore possible mechanisms underlying these strain-specific pathology and severity of human toxoplasmosis. Although no final conclusions can be drawn, it is clear that virulent strains (e.g. strains containing type I or atypical alleles) are significantly more often associated with increased frequency and severity of human toxoplasmosis. The significance of highly virulent strains can cause severe diseases in immunocompetent patients and might implicated in brain disorders such as schizophrenia should led to reconsideration of toxoplasmosis. Further studies that combine parasite strain typing and human factor analysis (e.g. immune status and genetic background) are required for better understanding of human susceptibility or resistance to toxoplasmosis.
Collapse
Affiliation(s)
- J. Xiao
- Stanley Division of Developmental Neurovirology; Department of Pediatrics; Johns Hopkins School of Medicine; Baltimore MD USA
| | - R. H. Yolken
- Stanley Division of Developmental Neurovirology; Department of Pediatrics; Johns Hopkins School of Medicine; Baltimore MD USA
| |
Collapse
|
24
|
Association between latent toxoplasmosis and psychiatric disorders in HIV-infected subjects. J Acquir Immune Defic Syndr 2015; 68:e8-9. [PMID: 25296099 DOI: 10.1097/qai.0000000000000384] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
25
|
Burokas A, Moloney RD, Dinan TG, Cryan JF. Microbiota regulation of the Mammalian gut-brain axis. ADVANCES IN APPLIED MICROBIOLOGY 2015; 91:1-62. [PMID: 25911232 DOI: 10.1016/bs.aambs.2015.02.001] [Citation(s) in RCA: 194] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The realization that the microbiota-gut-brain axis plays a critical role in health and disease has emerged over the past decade. The brain-gut axis is a bidirectional communication system between the central nervous system (CNS) and the gastrointestinal tract. Regulation of the microbiota-brain-gut axis is essential for maintaining homeostasis, including that of the CNS. The routes of this communication are not fully elucidated but include neural, humoral, immune, and metabolic pathways. A number of approaches have been used to interrogate this axis including the use of germ-free animals, probiotic agents, antibiotics, or animals exposed to pathogenic bacterial infections. Together, it is clear that the gut microbiota can be a key regulator of mood, cognition, pain, and obesity. Understanding microbiota-brain interactions is an exciting area of research which may contribute new insights into individual variations in cognition, personality, mood, sleep, and eating behavior, and how they contribute to a range of neuropsychiatric diseases ranging from affective disorders to autism and schizophrenia. Finally, the concept of psychobiotics, bacterial-based interventions with mental health benefit, is also emerging.
Collapse
Affiliation(s)
- Aurelijus Burokas
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - Rachel D Moloney
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Psychiatry, University College Cork, Cork, Ireland
| | - John F Cryan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
26
|
Eugene AR, Masiak J. Electrophysiological Neuroimaging using sLORETA Comparing 100 Schizophrenia Patients to 48 Patients with Major Depression. BRAIN : BROAD RESEARCH IN ARTIFICIAL INTELLIGENCE AND NEUROSCIENCE 2014; 5:16-25. [PMID: 26609423 PMCID: PMC4655883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In this retrospective analysis of electroencephalograms were to identify a surrogate biomarker for the Dopamine D2 receptors in the brain by comparing patients diagnosed with Schizophrenia taking Atypical Antipsychotics to Depressive patients medicated with Selective Serotonin Reuptake Inhibitors. To achieve this, thirty-seconds of resting EEG were spectrally transformed in sLORETA. Three-dimensional statistical non-paramentric maps (SnPM) for the sLORETA Global Field Power within each band were then computed. Our results illustrated that the Right Superior Frontal Gyrus (t=2.049, p=0.007), along the dopamine mesolimbic pathway, had higher neuronal oscillations in the delta frequency band in the 100 Schizophrenia patients as compared to the 32-depressive female patients. The comparisons with both the 48 depressive patient cohort or the sixteen male depressive patient cohort did not yield any statistically significant findings. We conclude that the Superior Frontal Gyrus should be investigated as a possible surrogate biomarker for preclinical and clinical drug discovery in neuropharmacology.
Collapse
Affiliation(s)
- Andy R. Eugene
- Department of Molecular Pharmacology and Experimental Therapeutics, Division of Clinical Pharmacology, Gonda 19, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| | - Jolanta Masiak
- Neurophysiology Unit, Department of Psychiatry, Medical University of Lublin, ul. Gluska 1 (SPSK Nr 1), Lublin 20-439, Poland
| |
Collapse
|
27
|
Dinan TG, Borre YE, Cryan JF. Genomics of schizophrenia: time to consider the gut microbiome? Mol Psychiatry 2014; 19:1252-7. [PMID: 25288135 DOI: 10.1038/mp.2014.93] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/06/2014] [Accepted: 07/08/2014] [Indexed: 12/12/2022]
Abstract
Research into the genomics of schizophrenia promises much, but so far is resplendent with failures to replicate, and has yielded little of therapeutic potential. Within our bodies resides a dynamic population of gut microbes forming a symbiotic superorganism comprising a myriad of bacteria of approximately 10(14) cells, containing 100 times the number of genes of the human genome and weighing approximately the same as the human brain. Recent preclinical investigations indicate that these microbes majorly impact on cognitive function and fundamental behavior patterns, such as social interaction and stress management. We are pivotally dependent on the neuroactive substances produced by such bacteria. The biological diversity of this ecosystem is established in the initial months of life and is highly impacted upon by environmental factors. To date, this vast quantity of DNA has been largely ignored in schizophrenia research. Perhaps it is time to reconsider this omission.
Collapse
Affiliation(s)
- T G Dinan
- 1] Alimentary Pharmabiotic Centre, University College, Cork, Ireland [2] Department of Psychiatry, Cork University Hospital, University College Cork, Cork, Ireland
| | - Y E Borre
- Alimentary Pharmabiotic Centre, University College, Cork, Ireland
| | - J F Cryan
- 1] Alimentary Pharmabiotic Centre, University College, Cork, Ireland [2] Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
28
|
Feigenson KA, Kusnecov AW, Silverstein SM. Inflammation and the two-hit hypothesis of schizophrenia. Neurosci Biobehav Rev 2014; 38:72-93. [PMID: 24247023 PMCID: PMC3896922 DOI: 10.1016/j.neubiorev.2013.11.006] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 10/26/2013] [Accepted: 11/07/2013] [Indexed: 12/12/2022]
Abstract
The high societal and individual cost of schizophrenia necessitates finding better, more effective treatment, diagnosis, and prevention strategies. One of the obstacles in this endeavor is the diverse set of etiologies that comprises schizophrenia. A substantial body of evidence has grown over the last few decades to suggest that schizophrenia is a heterogeneous syndrome with overlapping symptoms and etiologies. At the same time, an increasing number of clinical, epidemiological, and experimental studies have shown links between schizophrenia and inflammatory conditions. In this review, we analyze the literature on inflammation and schizophrenia, with a particular focus on comorbidity, biomarkers, and environmental insults. We then identify several mechanisms by which inflammation could influence the development of schizophrenia via the two-hit hypothesis. Lastly, we note the relevance of these findings to clinical applications in the diagnosis, prevention, and treatment of schizophrenia.
Collapse
Affiliation(s)
- Keith A Feigenson
- Robert Wood Johnson Medical School at Rutgers, The State University of New Jersey, 675 Hoes Lane, Piscataway, NJ 08854, USA.
| | - Alex W Kusnecov
- Department of Psychology, Behavioral and Systems Neuroscience Program and Joint Graduate Program in Toxicology, Rutgers University, 52 Frelinghuysen Road, Piscataway, NJ 08854-8020, USA.
| | - Steven M Silverstein
- Robert Wood Johnson Medical School at Rutgers, The State University of New Jersey, 675 Hoes Lane, Piscataway, NJ 08854, USA; University Behavioral Health Care at Rutgers, The State University of New Jersey, 671 Hoes Lane, Piscataway, NJ 08855, USA.
| |
Collapse
|
29
|
Caldas LA, Seabra SH, Attias M, de Souza W. The effect of kinase, actin, myosin and dynamin inhibitors on host cell egress by Toxoplasma gondii. Parasitol Int 2013; 62:475-82. [DOI: 10.1016/j.parint.2013.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 03/26/2013] [Accepted: 04/18/2013] [Indexed: 12/21/2022]
|
30
|
Donor CD8+ T cells prevent Toxoplasma gondii de-encystation but fail to rescue the exhausted endogenous CD8+ T cell population. Infect Immun 2013; 81:3414-25. [PMID: 23817617 DOI: 10.1128/iai.00784-12] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Functional exhaustion of CD8(+) T cells due to increased expression of inhibitory molecule PD-1 (Programmed Death-1) causes reactivation of latent disease during later phases of chronic toxoplasmosis. Onset of disease recrudescence results in decreased parasite cyst burden concomitant with parasites undergoing stage conversion from a primarily encysted, quiescent bradyzoite to a fast-replicating, highly motile tachyzoite. Thus, reduced cyst burden is one of the early hallmarks of disease recrudescence. This was further validated by depleting gamma interferon (IFN-γ), a cytokine known to control latent toxoplasmosis, in chronically infected prerecrudescent mice. Since CD8(+) T cells (an important source of IFN-γ) lose their functionality during the later phases of chronic toxoplasmosis, we next examined if adoptive transfer of functional CD8(+) T cells from acutely infected donors to the chronically infected prerecrudescent hosts could impede parasite de-encystation and rescue exhausted CD8(+) T cells. While the transfer of immune CD8(+) T cells temporarily restricted the breakdown of cysts, the exhausted endogenous CD8(+) T cell population was not rescued. Over time, the donor population got deleted, resulting in parasite de-encystation and host mortality. Considering that donor CD8(+) T cells fail to become long-lived, one of the cardinal features of memory CD8(+) T cells, it bears the implication that memory CD8 differentiation is impaired during chronic toxoplasmosis. Moreover, our data strongly suggest that while adoptive immunotherapy can prevent parasite de-encystation transiently, reduced antigen burden in the chronic phase by itself is insufficient for rescue of exhausted CD8(+) T cells. The conclusions of this study have profound ramifications in designing immunotherapeutics against chronic toxoplasmosis.
Collapse
|