1
|
Lu JM, Jin GN, Xin Y, Ma JW, Shen XY, Quan YZ, Liu YM, Zhou JY, Wang BZ, Li YB, Xu X, Piao LX. Lactoferrin-modified nanoemulsions enhance brain-targeting and therapeutic efficacy of arctigenin against Toxoplasma gondii-induced neuronal injury. Int J Parasitol Drugs Drug Resist 2025; 27:100575. [PMID: 39729771 PMCID: PMC11733198 DOI: 10.1016/j.ijpddr.2024.100575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 12/29/2024]
Abstract
Toxoplasma gondii, a neurotropic protozoan parasite, affects the central nervous system and causes various neurological disorders. Previous studies have demonstrated that Arctigenin (AG) exhibits anti-T. gondii activity and reduces depression-like behaviors induced by T. gondii infection. This study aimed to enhance AG's brain-targeting and therapeutic efficacy by developing lactoferrin-modified nanoemulsions loaded with AG (Lf-AG-NEs). Lf-modified nanoemulsions were prepared and assessed using in vivo and in vitro infection models with the T. gondii RH strain, and a co-culture system of BV2 microglia and primary neuron cells. The effects of Lf-AG-NEs on T. gondii-induced neuronal injury were examined, and potential molecular mechanisms were elucidated through real-time quantitative PCR, western blotting, immunofluorescence, flow cytometry, immunohistochemistry, and Nissl staining. In vitro assessments showed significant increases in cellular uptake and blood-brain barrier penetration by Lf-AG-NEs. These nanoemulsions notably inhibited T. gondii proliferation in brain tissue and BV2 cells, surpassing the effects of free AG or AG-NEs alone. Additionally, Lf-AG-NEs substantially alleviated neuropathological changes and reduced microglial activation and neuroinflammation by downregulating the TLR4/NF-κB and TNFR1/NF-κB signaling pathways. Co-culturing BV2 cells with primary cortical neurons indicated that Lf-AG-NEs, similarly to CLI-095 and R7050, attenuated T. gondii-induced microglial activation and subsequent neuronal injury. In conclusion, the successfully prepared Lf-AG-NEs not only enhanced the anti-T. gondii effect but also strengthened the protective impact against neuronal injury induced by T. gondii, through the modulation of microglial signaling pathways.
Collapse
Affiliation(s)
- Jing-Mei Lu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Guang-Nan Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yan Xin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Jing-Wen Ma
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Xin-Yu Shen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yan-Zhu Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yi-Ming Liu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Jin-Yi Zhou
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Bing-Zhe Wang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Ying-Biao Li
- Department of Neurology, Yanbian University Hospital, Yanbian University, Yanji, 133000, Jilin Province, China.
| | - Xiang Xu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
| | - Lian-Xun Piao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
| |
Collapse
|
2
|
El-Kady AM, Elshazly H, Alsulami MN, Albohiri HH, Alshehri EA, Alfaifi MS, Mohamed K, Wakid MH, Gattan HS, Altwaim SA, Al-Megrin WAI, Almalki GH, Abdel-Rahman IAM, Elshabrawy HA, Younis S. Zingiber officinale Ameliorates Acute Toxoplasmosis-Induced Pathology in Mice. Acta Parasitol 2024; 69:1785-1800. [PMID: 39225734 DOI: 10.1007/s11686-024-00884-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Toxoplasma gondii (T. gondii) infects one third of the world's population with significant illness, mainly among immunocompromised individuals and pregnant women. Treatment options for toxoplasmosis are limited which signifies the need for novel, potent, and safe therapeutic options. The goal of this study was to assess the effectiveness of the ethanolic extract of Zingiber officinale (Z. officinale) in treating mice infected with the RH T. gondii strain. MATERIALS AND METHODS Gas Chromatography/Mass Spectrometry (GC/MS) was used to identify components of ethanolic extract of Z. officinale. A total of 80 mice were randomly allocated into four experimental groups that contained 20 mice each. The first group was left uninfected (uninfected control), while three groups were infected with T. gondii RH virulent strain tachyzoites at 2500 tachyzoites/mouse. One infected group was left untreated (infected, untreated), whereas the other two groups were treated orally with either spiramycin (positive control) or Z. officinale ethanolic extract at doses of 200 mg/kg and 500 mg/kg, respectively for 5 days, starting the day of infection. Ten mice from each group were used to assess mice survival in different groups, whereas the other ten mice in each group were sacrificed on the 5th day post-infectin (dpi) to estimate the treatment efficacy by quantifying liver parasite load, liver function, nitric oxide (NO) production, and levels of antioxidant enzymes. Additionally, histopathological studies were performed to evaluate the therapeutic effect of Z. officinale treatment on toxoplasmosis-induced pathological alterations in liver, brain, and spleen. RESULTS Treatment with Z. officinale ethanolic extract extended the survival of mice till 9th dpi compared to 7th dpi in infected untreated mice. Higher percentage of mice survived in Z. officinale-treated group compared to spiramycin-treatment group at different time points. Liver parasite loads were significantly lower in Z. officinale extract-treated mice and spiramycin-treated mice compared to infected untreated mice which correlated with significantly lower levels of serum liver enzymes (ALT, AST) and nitric oxide (NO), as well as significantly higher catalase (CAT) antioxidant enzyme activity. Scanning electron microscopy (SEM) examination of tachyzoites from the peritoneal fluid revealed marked damage in tachyzoites from Z. officinale-treated group compared to that from infected untreated mice. Moreover, treatment with Z. officinale ethanolic extract alleviated infection-induced pathological alterations and restored normal tissue morphology of liver, brain, and spleen. CONCLUSION Our results demonstrated that Z. officinale treatment reduced parasite burden and reversed histopathological and biochemical alterations in acute murine toxoplasmosis. These findings support the potential utility of Z. officinale as a future effective natural therapeutic for toxoplasmosis. Further studies are needed to determine the effective active ingredient in Z. officinale extract that can be further optimized for treatment of toxoplasmosis.
Collapse
Affiliation(s)
- Asmaa M El-Kady
- Department of Medical Parasitology, Faculty of Medicine, South Valley University, Qena, 83523, Egypt.
| | - Hayam Elshazly
- Department of Biology, Faculty of Sciences-Scientific Departments, Qassim University, Bu-raidah, Qassim, 52571, Saudi Arabia
| | - Muslimah N Alsulami
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Haleema H Albohiri
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Eman Abdullah Alshehri
- Department of Zoology, College of Science, King Saud University, Riyadh, 11362, Saudi Arabia
| | - Mashael S Alfaifi
- Department of Epidemiology and Medical Statistics, Faculty of Public Health and Health Informatics, Umm Al-Qura 21 University, Mecca, 21961, Saudi Arabia
| | - Khalil Mohamed
- Department of Epidemiology and Medical Statistics, Faculty of Public Health and Health Informatics, Umm Al-Qura 21 University, Mecca, 21961, Saudi Arabia
| | - 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, Jeddah, 21589, Saudi Arabia.
| | - Hattan S Gattan
- 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, Jeddah, 21589, Saudi Arabia
| | - Sarah A Altwaim
- Special Infectious Agents unit, King Fahd Medical Research Center, Jeddah, 21589, Saudi Arabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Wafa Abdullah I Al-Megrin
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Ghaliah H Almalki
- Department of Biology, College of Science, Jazan University, Jazan, 45142, Saudi Arabia
| | - Iman A M Abdel-Rahman
- Department of Pharmacognosy, Faculty of Pharmacy, South Valley University, Qena, 83523, Egypt
| | - Hatem A Elshabrawy
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX, 77304, USA.
| | - Salwa Younis
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, 21526, Egypt
| |
Collapse
|
3
|
Diniz DG, de Oliveira JHP, Guerreiro LCF, de Menezes GC, de Assis ACL, Duarte TQ, dos Santos IBD, Maciel FD, Soares GLDS, Araújo SC, Franco FTDC, do Carmo EL, Morais RDAB, de Lima CM, Brites D, Anthony DC, Diniz JAP, Diniz CWP. Contrasting Disease Progression, Microglia Reactivity, Tolerance, and Resistance to Toxoplasma gondii Infection in Two Mouse Strains. Biomedicines 2024; 12:1420. [PMID: 39061995 PMCID: PMC11274029 DOI: 10.3390/biomedicines12071420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/02/2024] [Accepted: 06/08/2024] [Indexed: 07/28/2024] Open
Abstract
Our study investigated the innate immune response to Toxoplasma gondii infection by assessing microglial phenotypic changes and sickness behavior as inflammatory response markers post-ocular tachyzoite instillation. Disease progression in Swiss albino mice was compared with the previously documented outcomes in BALB/c mice using an identical ocular route and parasite burden (2 × 105 tachyzoites), with saline as the control. Contrary to expectations, the Swiss albino mice displayed rapid, lethal disease progression, marked by pronounced sickness behaviors and mortality within 11-12 days post-infection, while the survivors exhibited no apparent signs of infection. Comparative analysis revealed the T. gondii-infected BALB/c mice exhibited reduced avoidance of feline odors, while the infected Swiss albino mice showed enhanced avoidance responses. There was an important increase in microglial cells in the dentate gyrus molecular layer of the infected Swiss albino mice compared to the BALB/c mice and their respective controls. Hierarchical cluster and discriminant analyses identified three microglial morphological clusters, differentially affected by T. gondii infection across strains. The BALB/c mice exhibited increased microglial branching and complexity, while the Swiss albino mice showed reduced shrunken microglial arbors, diminishing their morphological complexity. These findings highlight strain-specific differences in disease progression and inflammatory regulation, indicating lineage-specific mechanisms in inflammatory responses, tolerance, and resistance. Understanding these elements is critical in devising control measures for toxoplasmosis.
Collapse
Affiliation(s)
- Daniel G. Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | - Jhonnathan H. P. de Oliveira
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Luma C. F. Guerreiro
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal do Pará, Campus Bragança, Bragança 68600-000, Pará, Brazil
| | - Gabriel C. de Menezes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Alexa C. L. de Assis
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Tainá Q. Duarte
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Izabelly B. D. dos Santos
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Flávia D. Maciel
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Gabrielly L. da S. Soares
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Sanderson C. Araújo
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Felipe T. de C. Franco
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Ediclei L. do Carmo
- Seção de Parasitologia, Instituto Evandro Chagas, Belém 67030-000, Pará, Brazil; (E.L.d.C.); (R.d.A.B.M.)
| | - Rafaela dos A. B. Morais
- Seção de Parasitologia, Instituto Evandro Chagas, Belém 67030-000, Pará, Brazil; (E.L.d.C.); (R.d.A.B.M.)
| | - Camila M. de Lima
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal;
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Daniel C. Anthony
- Laboratory of Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford OX1 2JD, UK;
| | - José A. P. Diniz
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Cristovam W. P. Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| |
Collapse
|
4
|
Itani S, Hamie M, El Jammal R, Abdine W, Doumit M, Charafeddine A, El-Sabban M, Patinote C, Masquefa C, Bonnet PA, Obeid M, El Hajj H. Imiquimod Reverses Chronic Toxoplasmosis-Associated Behavioral and Neurocognitive Anomalies in a Rat Model. Biomedicines 2024; 12:1295. [PMID: 38927503 PMCID: PMC11202296 DOI: 10.3390/biomedicines12061295] [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: 05/14/2024] [Revised: 06/04/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Toxoplasma gondii is the etiologic agent of toxoplasmosis, a highly prevalent parasitosis. Toxoplasma gondii (T. gondii) transits in the brain from acute (AT) to chronic toxoplasmosis (CT), under host immune control. In immunocompromised patients, reactivation of CT is potentially life-threatening. Behavioral and neurological complications have been associated with CT. Furthermore, an effective treatment targeting CT is still lacking. We previously reported the efficacy of imiquimod against CT. Here, we demonstrate the molecular effects of imiquimod or imiquimod followed by the clinically used combination of sulfadiazine and pyrimethamine (SDZ + PYR) on CT-associated behavior in a rat model. Imiquimod decreased the number of cysts in the brains of chronically infected rats due to an induced reactivation of bradyzoites into tachyzoites. Importantly, this decrease was more pronounced in rats treated with imiquimod followed by SDZ + PYR. Rats chronically infected with T. gondii exhibited an anxiety-like behavior. Notably, treatment with imiquimod reversed this behavior aberrancy, with even a more pronounced effect with imiquimod followed by SDZ/PYR. Similarly, rats chronically infected with T. gondii exhibited learning deficits, and imiquimod alone or followed by SDZ/PYR reversed this behavior. Our results enhance our knowledge of the implications of CT on behavioral aberrancies and highlight the potency of imiquimod followed by SDZ + PYR on these CT-associated complications.
Collapse
Affiliation(s)
- Shaymaa Itani
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (S.I.); (M.H.); (W.A.)
| | - Maguy Hamie
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (S.I.); (M.H.); (W.A.)
| | - Reem El Jammal
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (R.E.J.); (M.D.); (M.E.-S.); (M.O.)
| | - Wassim Abdine
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (S.I.); (M.H.); (W.A.)
| | - Mark Doumit
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (R.E.J.); (M.D.); (M.E.-S.); (M.O.)
| | - Adib Charafeddine
- College of Pharmacy, American University of Iraq-Baghdad, Baghdad 10071, Iraq;
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (R.E.J.); (M.D.); (M.E.-S.); (M.O.)
| | - Cindy Patinote
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, ENSCM, Université de Montpellier, 34090 Montpellier, France; (C.P.); (C.M.); (P.-A.B.)
| | - Carine Masquefa
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, ENSCM, Université de Montpellier, 34090 Montpellier, France; (C.P.); (C.M.); (P.-A.B.)
| | - Pierre-Antoine Bonnet
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, ENSCM, Université de Montpellier, 34090 Montpellier, France; (C.P.); (C.M.); (P.-A.B.)
| | - Makram Obeid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (R.E.J.); (M.D.); (M.E.-S.); (M.O.)
| | - Hiba El Hajj
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (S.I.); (M.H.); (W.A.)
| |
Collapse
|
5
|
Bergersen KV, Ramirez AD, Kavvathas B, Mercer F, Wilson EH. Human neutrophil-like cells demonstrate antimicrobial responses to the chronic cyst form of Toxoplasma gondii. Parasite Immunol 2023; 45:e13011. [PMID: 37776091 PMCID: PMC11246559 DOI: 10.1111/pim.13011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 10/01/2023]
Abstract
The protozoan parasite Toxoplasma gondii infects approximately 2.5 billion people worldwide. Infection induces a rapid dissemination of parasites throughout the body followed by the formation of lifelong cysts within neurons of the host brain. Both stages require a dynamic immune response comprised of both innate and adaptive cells. Neutrophils are a primary responding cell to acute infection and have been observed in the brain during murine chronic infection. Previous studies investigating human neutrophils found that invasion by Toxoplasma tachyzoites inhibits apoptosis of neutrophils, prolonging their survival under inflammatory conditions. Here, we demonstrate the differentiation of two distinct subsets following exposure of human neutrophil-like-cells (HNLC) to Toxoplasma cysts. In vitro stimulation and imaging studies show cyst-specific induction of cytokines and cyst clearance by HNLCs. Further testing demonstrates that aged HNLCs perform less phagocytosis of cysts compared to non-aged HNLCs. In conclusion, this study identifies a novel response of HNLCs to Toxoplasma cysts and may indicate a role for neutrophils in the clearance of cysts during human infection with Toxoplasma.
Collapse
Affiliation(s)
- Kristina V. Bergersen
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States of America
| | - Ashley D. Ramirez
- Department of Biological Sciences, California State Polytechnic University, Pomona
| | - Bill Kavvathas
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States of America
| | - Frances Mercer
- Department of Biological Sciences, California State Polytechnic University, Pomona
| | - Emma H. Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States of America
| |
Collapse
|
6
|
Mostafa E, Ahmed FASMS, Yahia SH, Ibrahim AIM, Elbahaie ES. The effects of intracellular iron availability on the outcome of Toxoplasma gondii infection in mice. J Parasit Dis 2023; 47:608-618. [PMID: 37520204 PMCID: PMC10382456 DOI: 10.1007/s12639-023-01603-9] [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: 02/16/2023] [Accepted: 05/26/2023] [Indexed: 08/01/2023] Open
Abstract
Toxoplasma gondii (T. gondii) is a parasite that obtains the iron it needs for its own metabolism from the host-cell iron pool. In this work, we aimed to investigate if iron supplementation or deficiency affected the course of T. gondii infection. Eighty mice were divided into four groups, each with 20 animals: Group (I): Uninfected control group. Group (II): Infected control group: injected with Phosphate buffered saline. Group (III): Infected group: received iron sucrose treatment. Group (IV): Infected group: treated with deferoxamine. Quantitative PCR studies were performed on days 3 and 8 post-infection to detect the expression of iron metabolism genes (hamp and ferroprotin) and immune-histochemical analysis to study the percentage of TNF-α and TGF-β tissue expression. Iron supplementation induced progressions of infection evident by increased tissue expression of pro-inflammatory cytokine TNF-α and downregulation of TGF-β which is mostly linked to suppression of the inflammatory process caused by T. gondii. Increased expression of TGF-β and decreased expression of TNF-α was noticed when iron deprivation occurred. On day 3, we noticed increased expression in the hamp gene with iron supplementation while it decreases when the iron supply is low. On the contrary, iron deficiency increased ferroprotin gene expression whereas supplementing decreased it. On day 8, the level of expression of these genes returned to normal levels. These observations document the potential role of iron in controlling toxoplasmosis infection and indicate that the transcription of hamp and ferroprotin in T. gondii-infected cells appears to be regulated by a sophisticated indirect mechanism.
Collapse
Affiliation(s)
- Eman Mostafa
- Medical Parasitology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | - Samah Hassan Yahia
- Medical Parasitology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | - Enas Saed Elbahaie
- Medical Parasitology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
7
|
Xie H, Sun H, Dong H, Dai L, Xu H, Zhang L, Wang Q, Zhang J, Zhao G, Xu C, Yin K. Label-free quantitative proteomic analyses of mouse astrocytes provides insight into the host response mechanism at different developmental stages of Toxoplasma gondii. PLoS Negl Trop Dis 2023; 17:e0011102. [PMID: 37721957 PMCID: PMC10538781 DOI: 10.1371/journal.pntd.0011102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 09/28/2023] [Accepted: 08/07/2023] [Indexed: 09/20/2023] Open
Abstract
Toxoplasma gondii (T. gondii) is an opportunistic parasite that can infect the central nervous system (CNS), causing severe toxoplasmosis and behavioral cognitive impairment. Mortality is high in immunocompromised individuals with toxoplasmosis, most commonly due to reactivation of infection in the CNS. There are still no effective vaccines and drugs for the prevention and treatment of toxoplasmosis. There are five developmental stages for T. gondii to complete life cycle, of which the tachyzoite and bradyzoite stages are the key to the acute and chronic infection. In this study, to better understanding of how T. gondii interacts with the host CNS at different stages of infection, we constructed acute and chronic infection models of T. gondii in astrocytes, and used label-free proteomics to detect the proteome changes before and after infection, respectively. A total of 4676 proteins were identified, among which 163 differentially expressed proteins (fold change ≥ 1.5 or ≤ 0.67 and p-value ≤ 0.05) including 109 up-regulated proteins and 54 down-regulated proteins in C8-TA vs C8 group, and 719 differentially expressed proteins including 495 up-regulated proteins and 224 down-regulated proteins in C8-BR vs C8-TA group. After T. gondii tachyzoites infected astrocytes, differentially expressed proteins were enriched in immune-related biological processes to promote the formation of bradyzoites and maintain the balance of T. gondii, CNS and brain. After T. gondii bradyzoites infected astrocytes, the differentially expressed proteins up-regulated the host's glucose metabolism, and some up-regulated proteins were strongly associated with neurodegenerative diseases. These findings not only provide new insights into the psychiatric pathogenesis of T. gondii, but also provide potential targets for the treatment of acute and chronic Toxoplasmosis.
Collapse
Affiliation(s)
- Huanhuan Xie
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong, China
| | - Hang Sun
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong, China
| | - Hongjie Dong
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong, China
| | - Lisha Dai
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong, China
| | - Haozhi Xu
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong, China
| | - Lixin Zhang
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong, China
- Xingan League Center for Disease Control and Prevention, Ulanhot, Inner Mongolia, China
| | - Qi Wang
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong, China
| | - Junmei Zhang
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong, China
| | - Guihua Zhao
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong, China
| | - Chao Xu
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong, China
| | - Kun Yin
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong, China
| |
Collapse
|
8
|
Wesołowski R, Pawłowska M, Mila-Kierzenkowska C. The Medical Relevance of Toxoplasma Infections in Terms of the Safety of Blood Recipients under Immunosuppression-A Meta-Analysis. Microorganisms 2023; 11:1980. [PMID: 37630541 PMCID: PMC10458665 DOI: 10.3390/microorganisms11081980] [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: 06/29/2023] [Revised: 07/25/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
Laboratory diagnosis of Toxoplasma gondii infection plays a crucial role in ensuring the safety of blood recipients, especially in the case of immunosuppressed people, such as organ transplant patients. Toxoplasmosis, caused by the parasite Toxoplasma gondii, is a potential threat to people with weakened immune systems, and blood transfusions from infected donors can lead to severe complications. In this publication, we analyze the medical relevance of Toxoplasma infection in the context of the safety of blood recipients, focusing on the immunosuppressed patient population. We present various diagnostic methods, such as serological, molecular, and microscopic tests, which can detect the presence of Toxoplasma gondii in donors' blood. We also discuss the importance of adequately interpreting diagnostic results, considering risk factors, and detectability of the infection. We pay special attention to high-sensitivity and -specificity diagnostic techniques, which allow us to minimize the risk of Toxoplasma gondii transmission to blood recipients. Our findings have important implications for clinical practice and organ transplantation guidelines, emphasizing the need to diagnose and monitor Toxoplasma infections in blood donors and recipients.
Collapse
Affiliation(s)
| | | | - Celestyna Mila-Kierzenkowska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland; (R.W.); (M.P.)
| |
Collapse
|
9
|
Qamar W, Alsayeqh AF. A review of foodborne Toxoplasma gondii with a special focus on its prevalence in Pakistan from 2000 to 2022. Front Vet Sci 2023; 9:1080139. [PMID: 36744224 PMCID: PMC9890071 DOI: 10.3389/fvets.2022.1080139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/07/2022] [Indexed: 01/19/2023] Open
Abstract
Third-world countries have a higher prevalence of food-related disorders than developed nations. Millions of people in underdeveloped countries are seriously at risk from the potential water supply contamination with protozoan diseases. Toxoplasma gondii is one of the important protozoans causing diseases in livestock and humans. Despite the standard tests for diagnosing this parasite and different treatment methods, the spread of these parasites is uncontrollable and rising every year due to other management disorders. In this review, we summarize etiopathogenesis and prevalence in Pakistan. We looked for papers reporting the seroprevalence of T. gondii in people and animals between 2000 and 2022 in different databases: PubMed, Google Scholar, ScienceDirect, Scopus, and Web of Science. Data on the seroprevalence of T. gondii in Pakistan's domestic animals (sheep and goats, horses, donkeys, mules, cattle, and buffaloes), domestic pets (cats and dogs), poultry and rodents, and humans were gathered. According to the findings, sheep had an estimated pooled seroprevalence of T. gondii that varied from 11.20 to 26.50 %, and goats from 24.50 to 38.40%. Whereas in buffalo the opposite trend was followed, and the prevalence was observed is 0% in 2022, in horses, donkeys, and mules, only one study was reported according to which a high prevalence was observed in mules (28.60%) followed by donkeys (23.50%) and horses (23.50%), in cats 38.5% prevalence was observed in a recent study and in dogs 28.43% observed, and in humans from 22 to 60%. Human beings are found to be the most affected species showing high prevalence among all. According to our findings, animals and pets not only serve as a reservoir for the parasite but also serve as a direct route for human infection with T. gondii. The diagnostic techniques used in the observed studies were mostly serological testing whereas only a few studies have only been observed with molecular testing. To know the exact pattern of the disease for its control, the trend of molecular and advanced testing should be adopted as it is more reliable. Moreover, to decrease the transmission chances of T. gondii to humans, it is crucial to manage T. gondii infections in non-human species.
Collapse
Affiliation(s)
- Warda Qamar
- Department of Parasitology, University of Agriculture, Faisalabad, Pakistan
| | - Abdullah F. Alsayeqh
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraidah, Saudi Arabia,*Correspondence: Abdullah F. Alsayeqh ✉
| |
Collapse
|
10
|
Figueiredo CA, Düsedau HP, Steffen J, Ehrentraut S, Dunay MP, Toth G, Reglödi D, Heimesaat MM, Dunay IR. The neuropeptide PACAP alleviates T. gondii infection-induced neuroinflammation and neuronal impairment. J Neuroinflammation 2022; 19:274. [PMCID: PMC9675261 DOI: 10.1186/s12974-022-02639-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 10/23/2022] [Indexed: 11/21/2022] Open
Abstract
Background Cerebral infection with the protozoan Toxoplasma gondii (T. gondii) is responsible for inflammation of the central nervous system (CNS) contributing to subtle neuronal alterations. Albeit essential for brain parasite control, continuous microglia activation and recruitment of peripheral immune cells entail distinct neuronal impairment upon infection-induced neuroinflammation. PACAP is an endogenous neuropeptide known to inhibit inflammation and promote neuronal survival. Since PACAP is actively transported into the CNS, we aimed to assess the impact of PACAP on the T. gondii-induced neuroinflammation and subsequent effects on neuronal homeostasis. Methods Exogenous PACAP was administered intraperitoneally in the chronic stage of T. gondii infection, and brains were isolated for histopathological analysis and determination of pathogen levels. Immune cells from the brain, blood, and spleen were analyzed by flow cytometry, and the further production of inflammatory mediators was investigated by intracellular protein staining as well as expression levels by RT-qPCR. Neuronal and synaptic alterations were assessed on the transcriptional and protein level, focusing on neurotrophins, neurotrophin-receptors and signature synaptic markers. Results Here, we reveal that PACAP administration reduced the inflammatory foci and the number of apoptotic cells in the brain parenchyma and restrained the activation of microglia and recruitment of monocytes. The neuropeptide reduced the expression of inflammatory mediators such as IFN-γ, IL-6, iNOS, and IL-1β. Moreover, PACAP diminished IFN-γ production by recruited CD4+ T cells in the CNS. Importantly, PACAP promoted neuronal health via increased expression of the neurotrophin BDNF and reduction of p75NTR, a receptor related to neuronal cell death. In addition, PACAP administration was associated with increased expression of transporters involved in glutamatergic and GABAergic signaling that are particularly affected during cerebral toxoplasmosis. Conclusions Together, our findings unravel the beneficial effects of exogenous PACAP treatment upon infection-induced neuroinflammation, highlighting the potential implication of neuropeptides to promote neuronal survival and minimize synaptic prejudice. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02639-z.
Collapse
Affiliation(s)
- Caio Andreeta Figueiredo
- grid.5807.a0000 0001 1018 4307Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-Von-Guericke University, Magdeburg, Germany
| | - Henning Peter Düsedau
- grid.5807.a0000 0001 1018 4307Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-Von-Guericke University, Magdeburg, Germany
| | - Johannes Steffen
- grid.5807.a0000 0001 1018 4307Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-Von-Guericke University, Magdeburg, Germany
| | - Stefanie Ehrentraut
- grid.5807.a0000 0001 1018 4307Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-Von-Guericke University, Magdeburg, Germany
| | - Miklos P. Dunay
- grid.483037.b0000 0001 2226 5083Department and Clinic of Surgery and Ophthalmology, University of Veterinary Medicine, Budapest, Hungary
| | - Gabor Toth
- grid.9008.10000 0001 1016 9625Department of Medical Chemistry, University of Szeged, Budapest, Hungary
| | - Dora Reglödi
- grid.9679.10000 0001 0663 9479Department of Anatomy, MTA-PTE PACAP Research Team and Szentagothai Research Center, University of Pecs Medical School, Pecs, Hungary
| | - Markus M. Heimesaat
- grid.6363.00000 0001 2218 4662Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Ildiko Rita Dunay
- grid.5807.a0000 0001 1018 4307Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-Von-Guericke University, Magdeburg, Germany ,grid.418723.b0000 0001 2109 6265Center for Behavioral Brain Sciences – CBBS, Magdeburg, Germany
| |
Collapse
|
11
|
Rostami A, Riahi SM, Mollalo A, Razavian I, Akbari N, Marhoommirzabak E, Mahjour S, Sartip B, Arshadi M, Razavian E, Ardekani A. Does latent Toxoplasma infection have a protective effect against developing multiple sclerosis? Evidence from an updated meta-analysis. Trans R Soc Trop Med Hyg 2022; 116:996-1006. [PMID: 35696089 DOI: 10.1093/trstmh/trac053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/14/2022] [Accepted: 05/17/2022] [Indexed: 01/19/2023] Open
Abstract
Previous epidemiologic evidence suggests a protective effect of Toxoplasma gondii infection against multiple sclerosis (MS) development; however, inconsistent findings have been reported in this regard. Therefore, we performed an updated meta-analysis of observational studies to investigate the association of To. gondii infection with MS development. We searched all articles published in PubMed, Scopus, Embase and Web of Science databases as of 20 December 2021. A random effects meta-analysis model was used to generate the pooled OR at 95% CIs. The heterogeneity between studies was assessed using I2 and Cochran's Q statistics. Moreover, the likelihood of publication bias was determined by Egger's regression test. A total of 11 studies were eligible for meta-analysis, including 1172 MS cases and 1802 controls. Our findings indicated that 29.8% (95% CI 22.8 to 37.2%) of MS patients were seropositive for To. gondii infection, compared with 34.2% (95% CI 21.9 to 47.6%) of control subjects. The estimated pooled OR was 0.79 (95% CI 0.49 to 1.26), suggesting a non-significant negative association between To. gondii infection and MS development (p>0.05). The current study does not support the significant protective role of To. gondii infection on MS development. Our findings imply that further well-designed epidemiological and mechanistic studies are warranted to ascertain the possible association between To. gondii infection and MS and to exclude the potential confounders.
Collapse
Affiliation(s)
- Ali Rostami
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Seyed Mohammad Riahi
- Cardiovascular Diseases Research Center, Department of Epidemiology and Biostatistics, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Abolfazl Mollalo
- Department of Public Health and Prevention Science, School of Health Sciences, Baldwin Wallace University, Berea, Ohio, USA
| | - Iman Razavian
- Department of Neurosurgery, Functional Neurosurgery Research Center, Shohada Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nahid Akbari
- Department of Genetic, Faculty of Basic Science, Islamic Azad University, Varamin Pishva Branch, Tehran, Iran
| | - Elika Marhoommirzabak
- Department of Neurology, University of Visayas, Gullas College of Medicine, Cebu city, 600 Cebu, Philippines
| | - Sanaz Mahjour
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Behnam Sartip
- Department of Internal Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahdi Arshadi
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Elnaz Razavian
- Department of Neurology, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Ali Ardekani
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
French T, Steffen J, Glas A, Osbelt L, Strowig T, Schott BH, Schüler T, Dunay IR. Persisting Microbiota and Neuronal Imbalance Following T. gondii Infection Reliant on the Infection Route. Front Immunol 2022; 13:920658. [PMID: 35898505 PMCID: PMC9311312 DOI: 10.3389/fimmu.2022.920658] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/25/2022] [Indexed: 12/18/2022] Open
Abstract
Toxoplasma gondii is a highly successful parasite capable of infecting all warm-blooded animals. The natural way of infection in intermediate hosts is the oral ingestion of parasite-contaminated water or food. In murine experimental models, oral infection (p.o.) of mice with T. gondii is applied to investigate mucosal and peripheral immune cell dynamics, whereas intraperitoneal infection (i.p.) is frequently used to study peripheral inflammation as well as immune cell – neuronal interaction in the central nervous system (CNS). However, the two infection routes have not yet been systematically compared along the course of infection. Here, C57BL/6 mice were infected p.o. or i.p. with a low dose of T. gondii cysts, and the acute and chronic stages of infection were compared. A more severe course of infection was detected following i.p. challenge, characterized by an increased weight loss and marked expression of proinflammatory cytokines particularly in the CNS during the chronic stage. The elevated proinflammatory cytokine expression in the ileum was more prominent after p.o. challenge that continued following the acute phase in both i.p. or p.o. infected mice. This resulted in sustained microbial dysbiosis, especially after p.o. challenge, highlighted by increased abundance of pathobionts from the phyla proteobacteria and a reduction of beneficial commensal species. Further, we revealed that in the CNS of i.p. infected mice CD4 and CD8 T cells displayed higher IFNγ production in the chronic stage. This corresponded with an increased expression of C1q and CD68 in the CNS and reduced expression of genes involved in neuronal signal transmission. Neuroinflammation-associated synaptic alterations, especially PSD-95, VGLUT, and EAAT2 expression, were more pronounced in the cortex upon i.p. infection highlighting the profound interplay between peripheral inflammation and CNS homeostasis.
Collapse
Affiliation(s)
- Timothy French
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
| | - Johannes Steffen
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
| | - Albert Glas
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
| | - Lisa Osbelt
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Björn H. Schott
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences, Magdeburg, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences, Magdeburg, Germany
- *Correspondence: Ildiko Rita Dunay,
| |
Collapse
|
13
|
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
|
14
|
Yin K, Xu C, Zhao G, Xie H. Epigenetic Manipulation of Psychiatric Behavioral Disorders Induced by Toxoplasma gondii. Front Cell Infect Microbiol 2022; 12:803502. [PMID: 35237531 PMCID: PMC8882818 DOI: 10.3389/fcimb.2022.803502] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/17/2022] [Indexed: 11/21/2022] Open
Abstract
Toxoplasma gondii is known to have a complex life cycle and infect almost all kinds of warm-blooded animals around the world. The brain of the host could be persistently infected by cerebral cysts, and a variety of psychiatric disorders such as schizophrenia and suicide have been reported to be related with latent toxoplasmosis. The infected animals showed fear reduction and a tendency to be preyed upon. However, the mechanism of this “parasites manipulation” effects have not been elucidated. Here, we reviewed the recent infection prevalence of toxoplasmosis and the evidence of mental and behavioral disorders induced by T. gondii and discussed the related physiological basis including dopamine dysregulation and gamma-aminobutyric acid (GABA) pathway and the controversial opinion of the necessity for cerebral cysts existence. Based on the recent advances, we speculated that the neuroendocrine programs and neurotransmitter imbalance may play a key role in this process. Simultaneously, studies in the evaluation of the expression pattern of related genes, long noncoding RNAs (lncRNAs), and mRNAs of the host provides a new point for understanding the mechanism of neurotransmitter dysfunction induced by parasite manipulation. Therefore, we summarized the animal models, T. gondii strains, and behavioral tests used in the related epigenetic studies and the responsible epigenetic processes; pinpointed opportunities and challenges in future research including the causality evidence of human psychiatric disorders, the statistical analysis for rodent-infected host to be more vulnerable preyed upon; and identified responsible genes and drug targets through epigenetics.
Collapse
|
15
|
Figueiredo CA, Steffen J, Morton L, Arumugam S, Liesenfeld O, Deli MA, Kröger A, Schüler T, Dunay IR. Immune response and pathogen invasion at the choroid plexus in the onset of cerebral toxoplasmosis. J Neuroinflammation 2022; 19:17. [PMID: 35027063 PMCID: PMC8759173 DOI: 10.1186/s12974-021-02370-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/22/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Toxoplasma gondii (T. gondii) is a highly successful parasite being able to cross all biological barriers of the body, finally reaching the central nervous system (CNS). Previous studies have highlighted the critical involvement of the blood-brain barrier (BBB) during T. gondii invasion and development of subsequent neuroinflammation. Still, the potential contribution of the choroid plexus (CP), the main structure forming the blood-cerebrospinal fluid (CSF) barrier (BCSFB) have not been addressed. METHODS To investigate T. gondii invasion at the onset of neuroinflammation, the CP and brain microvessels (BMV) were isolated and analyzed for parasite burden. Additionally, immuno-stained brain sections and three-dimensional whole mount preparations were evaluated for parasite localization and morphological alterations. Activation of choroidal and brain endothelial cells were characterized by flow cytometry. To evaluate the impact of early immune responses on CP and BMV, expression levels of inflammatory mediators, tight junctions (TJ) and matrix metalloproteinases (MMPs) were quantified. Additionally, FITC-dextran was applied to determine infection-related changes in BCSFB permeability. Finally, the response of primary CP epithelial cells to T. gondii parasites was tested in vitro. RESULTS Here we revealed that endothelial cells in the CP are initially infected by T. gondii, and become activated prior to BBB endothelial cells indicated by MHCII upregulation. Additionally, CP elicited early local immune response with upregulation of IFN-γ, TNF, IL-6, host-defence factors as well as swift expression of CXCL9 chemokine, when compared to the BMV. Consequently, we uncovered distinct TJ disturbances of claudins, associated with upregulation of MMP-8 and MMP-13 expression in infected CP in vivo, which was confirmed by in vitro infection of primary CP epithelial cells. Notably, we detected early barrier damage and functional loss by increased BCSFB permeability to FITC-dextran in vivo, which was extended over the infection course. CONCLUSIONS Altogether, our data reveal a close interaction between T. gondii infection at the CP and the impairment of the BCSFB function indicating that infection-related neuroinflammation is initiated in the CP.
Collapse
Affiliation(s)
- Caio Andreeta Figueiredo
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
| | - Johannes Steffen
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
| | - Lorena Morton
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
| | - Sushmitha Arumugam
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
| | - Oliver Liesenfeld
- Institute for Microbiology and Hygiene, Charité Medical School, Berlin, Germany
| | - Mária A Deli
- Institute of Biophysics, Biological Research Centre, 6726, Szeged, Hungary
| | - Andrea Kröger
- Institute for Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, CBBS, Magdeburg, Germany.
| |
Collapse
|
16
|
Abstract
Toxoplasma gondii is a parasitic protist infecting a wide group of warm-blooded animals, ranging from birds to humans. While this infection is usually asymptomatic in healthy individuals, it can also lead to severe ocular or neurological outcomes in immunocompromised individuals or in developing fetuses. This obligate intracellular parasite has the ability to infect a considerable range of nucleated cells and can propagate in the intermediate host. Yet, under the pressure of the immune system it transforms into an encysted persistent form residing primarily in the brain and muscle tissues. Encysted parasites, which are resistant to current medication, may reactivate and give rise to an acute infection. The clinical outcome of toxoplasmosis depends on a complex balance between the host immune response and parasite virulence factors. Susceptibility to the disease is thus determined by both parasite strains and host species. Recent advances on our understanding of host cell-parasite interactions and parasite virulence have brought new insights into the pathophysiology of T. gondii infection and are summarized here.
Collapse
|
17
|
Hajj RE, Tawk L, Itani S, Hamie M, Ezzeddine J, El Sabban M, El Hajj H. Toxoplasmosis: Current and Emerging Parasite Druggable Targets. Microorganisms 2021; 9:microorganisms9122531. [PMID: 34946133 PMCID: PMC8707595 DOI: 10.3390/microorganisms9122531] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022] Open
Abstract
Toxoplasmosis is a prevalent disease affecting a wide range of hosts including approximately one-third of the human population. It is caused by the sporozoan parasite Toxoplasma gondii (T. gondii), which instigates a range of symptoms, manifesting as acute and chronic forms and varying from ocular to deleterious congenital or neuro-toxoplasmosis. Toxoplasmosis may cause serious health problems in fetuses, newborns, and immunocompromised patients. Recently, associations between toxoplasmosis and various neuropathies and different types of cancer were documented. In the veterinary sector, toxoplasmosis results in recurring abortions, leading to significant economic losses. Treatment of toxoplasmosis remains intricate and encompasses general antiparasitic and antibacterial drugs. The efficacy of these drugs is hindered by intolerance, side effects, and emergence of parasite resistance. Furthermore, all currently used drugs in the clinic target acute toxoplasmosis, with no or little effect on the chronic form. In this review, we will provide a comprehensive overview on the currently used and emergent drugs and their respective parasitic targets to combat toxoplasmosis. We will also abridge the repurposing of certain drugs, their targets, and highlight future druggable targets to enhance the therapeutic efficacy against toxoplasmosis, hence lessening its burden and potentially alleviating the complications of its associated diseases.
Collapse
Affiliation(s)
- Rana El Hajj
- Department of Biological Sciences, Beirut Arab University, P.O. Box 11-5020, Riad El Solh, Beirut 1107 2809, Lebanon;
| | - Lina Tawk
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Balamand, Beirut 1100 2807, Lebanon; (L.T.); (J.E.)
| | - Shaymaa Itani
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon; (S.I.); (M.H.)
| | - Maguy Hamie
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon; (S.I.); (M.H.)
| | - Jana Ezzeddine
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Balamand, Beirut 1100 2807, Lebanon; (L.T.); (J.E.)
| | - Marwan El Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon;
| | - Hiba El Hajj
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon; (S.I.); (M.H.)
- Correspondence: ; Tel.: +961–1-350000 (ext. 4897)
| |
Collapse
|
18
|
Daher D, Shaghlil A, Sobh E, Hamie M, Hassan ME, Moumneh MB, Itani S, El Hajj R, Tawk L, El Sabban M, El Hajj H. Comprehensive Overview of Toxoplasma gondii-Induced and Associated Diseases. Pathogens 2021; 10:pathogens10111351. [PMID: 34832507 PMCID: PMC8625914 DOI: 10.3390/pathogens10111351] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/07/2021] [Accepted: 10/14/2021] [Indexed: 12/03/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is a prevalent protozoan parasite of medical and veterinary significance. It is the etiologic agent of toxoplasmosis, a neglected disease in which incidence and symptoms differ between patients and regions. In immunocompetent patients, toxoplasmosis manifests as acute and chronic forms. Acute toxoplasmosis presents as mild or asymptomatic disease that evolves, under the host immune response, into a persistent chronic disease in healthy individuals. Chronic toxoplasmosis establishes as latent tissue cysts in the brain and skeletal muscles. In immunocompromised patients, chronic toxoplasmosis may reactivate, leading to a potentially life-threatening condition. Recently, the association between toxoplasmosis and various diseases has been shown. These span primary neuropathies, behavioral and psychiatric disorders, and different types of cancer. Currently, a direct pre-clinical or clinical molecular connotation between toxoplasmosis and most of its associated diseases remains poorly understood. In this review, we provide a comprehensive overview on Toxoplasma-induced and associated diseases with a focus on available knowledge of the molecular players dictating these associations. We will also abridge the existing therapeutic options of toxoplasmosis and highlight the current gaps to explore the implications of toxoplasmosis on its associated diseases to advance treatment modalities.
Collapse
Affiliation(s)
- Darine Daher
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Ahmad Shaghlil
- Department of Biology, Faculty of Sciences, R. Hariri Campus, Lebanese University, Beirut 1107 2020, Lebanon; (A.S.); (E.S.)
| | - Eyad Sobh
- Department of Biology, Faculty of Sciences, R. Hariri Campus, Lebanese University, Beirut 1107 2020, Lebanon; (A.S.); (E.S.)
| | - Maguy Hamie
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Malika Elhage Hassan
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Mohamad Bahij Moumneh
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Shaymaa Itani
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Rana El Hajj
- Department of Biological Sciences, Beirut Arab University, Beirut 1107 2809, Lebanon;
| | - Lina Tawk
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Balamand, Beirut 1100 2807, Lebanon;
| | - Marwan El Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon;
| | - Hiba El Hajj
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
- Correspondence: ; Tel.: +961–1-350000 (ext. 4897)
| |
Collapse
|
19
|
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
|
20
|
Maruszewska-Cheruiyot M, Stear M, Donskow-Łysoniewska K. Galectins - Important players of the immune response to CNS parasitic infection. Brain Behav Immun Health 2021; 13:100221. [PMID: 34589740 PMCID: PMC8474370 DOI: 10.1016/j.bbih.2021.100221] [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: 10/06/2020] [Revised: 01/18/2021] [Accepted: 01/30/2021] [Indexed: 11/18/2022] Open
Abstract
Galectins are a family of proteins that bind β-galactosides and play key roles in a variety of cellular processes including host defense and entry of parasites into the host cells. They have been well studied in hosts but less so in parasites. As both host and parasite galectins are highly upregulated proteins following infection, galectins are an area of increasing interest and their role in immune modulation has only recently become clear. Correlation of CNS parasitic diseases with mental disorders as a result of direct or indirect interaction has been observed. Therefore, galectins produced by the parasite should be taken into consideration as potential therapeutic agents.
Collapse
Affiliation(s)
- Marta Maruszewska-Cheruiyot
- Laboratory of Parasitology, General Karol Kaczkowski Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163, Warsaw, Poland
- Corresponding author.
| | - Michael Stear
- Department of Animal, Plant and Soil Science, Agribio, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Katarzyna Donskow-Łysoniewska
- Laboratory of Parasitology, General Karol Kaczkowski Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163, Warsaw, Poland
| |
Collapse
|
21
|
Hamie M, Tawil N, El Hajj R, Najm R, Moodad S, Hleihel R, Karam M, El Sayyed S, Besteiro S, El-Sabban M, Dubremetz JF, Lebrun M, El Hajj H. P18 (SRS35/TgSAG4) Plays a Role in the Invasion and Virulence of Toxoplasma gondii. Front Immunol 2021; 12:643292. [PMID: 34262559 PMCID: PMC8273438 DOI: 10.3389/fimmu.2021.643292] [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: 12/17/2020] [Accepted: 06/10/2021] [Indexed: 11/13/2022] Open
Abstract
Toxoplasmosis is a prevalent parasitic disease caused by Toxoplasma gondii (T. gondii). Under the control of the host immune system, T. gondii persists as latent bradyzoite cysts. Immunosuppression leads to their reactivation, a potentially life-threatening condition. Interferon-gamma (IFN-γ) controls the different stages of toxoplasmosis. Here, we addressed the role of the parasite surface antigen P18, belonging to the Surface-Antigen 1 (SAG-1) Related Sequence (SRS) family, in a cyst-forming strain. Deletion of P18 gene (KO P18) impaired the invasion of parasites in macrophages and IFN-γ-mediated activation of macrophages further reduced the invasion capacity of this KO, as compared to WT strain. Mice infected by KO P18, showed a marked decrease in virulence during acute toxoplasmosis. This was consequent to less parasitemia, accompanied by a substantial recruitment of dendritic cells, macrophages and natural killer cells (NK). Furthermore, KO P18 resulted in a higher number of bradyzoite cysts, and a stronger inflammatory response. A prolonged survival of mice was observed upon immunosuppression of KO P18 infected BALB/c mice or upon oral infection of Severe Combined Immunodeficiency (SCID) mice, with intact macrophages and natural killer (NK) cells. In stark contrast, oral infection of NSG (NOD/Shi-scid/IL-2Rγnull) mice, defective in macrophages and NK cells, with KO P18, was as lethal as that of the control strain showing that the conversion from bradyzoites to tachyzoites is intact and, suggesting a role of P18 in the response to host IFN-γ. Collectively, these data demonstrate a role for P18 surface antigen in the invasion of macrophages and in the virulence of the parasite, during acute and chronic toxoplasmosis.
Collapse
Affiliation(s)
- Maguy Hamie
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nadim Tawil
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rana El Hajj
- Department of Biological Sciences, Beirut Arab University, Beirut, Lebanon
| | - Rania Najm
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sara Moodad
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rita Hleihel
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Martin Karam
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sana El Sayyed
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Maryse Lebrun
- LPHI UMR5235, Univ Montpellier, CNRS, Montpellier, France
| | - Hiba El Hajj
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
22
|
Nayeri T, Sarvi S, Sharif M, Daryani A. Toxoplasma gondii: A possible etiologic agent for Alzheimer's disease. Heliyon 2021; 7:e07151. [PMID: 34141920 PMCID: PMC8187970 DOI: 10.1016/j.heliyon.2021.e07151] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/03/2021] [Accepted: 05/24/2021] [Indexed: 01/03/2023] Open
Abstract
Toxoplasma gondii (T. gondii) is one of the most pervasive neurotropic pathogens causing different lesions in a wide variety of mammals as intermediate hosts, including humans. It is estimated that one-third of the world population is infected with T. gondii; however, for a long time, there has been much interest in the examination of the possible role of this parasite in the development of mental disorders, such as Alzheimer's disease (AD). T. gondii may play a role in the progression of AD using mechanisms, such as the induction of the host's immune responses, inflammation of the central nervous system (CNS), alteration in the levels of neurotransmitters, and activation of indoleamine-2,3-dyoxigenase. This paper presents an appraisal of the literature, reports, and studies that seek to the possible role of T. gondii in the development of AD. For achieving the purpose of the current study, a search of six English databases (PubMed, ScienceDirect, Web of Science, Scopus, ProQuest, and Google Scholar) was performed. The results support the involvement of T. gondii in the induction and development of AD. Indeed, T. gondii can be considered a risk factor for the development of AD and requires the special attention of specialists and patients. Furthermore, the results of this study may contribute to prevent or delay the progress of AD worldwide. Therefore, it is required to carry out further studies in order to better perceive the parasitic mechanisms in the progression of AD.
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
| | - Mehdi Sharif
- 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
|
23
|
Poncet AF, Bosteels V, Hoffmann E, Chehade S, Rennen S, Huot L, Peucelle V, Maréchal S, Khalife J, Blanchard N, Janssens S, Marion S. The UPR sensor IRE1α promotes dendritic cell responses to control Toxoplasma gondii infection. EMBO Rep 2021; 22:e49617. [PMID: 33586853 DOI: 10.15252/embr.201949617] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 01/23/2023] Open
Abstract
The unfolded protein response (UPR) has emerged as a central regulator of immune cell responses in several pathologic contexts including infections. However, how intracellular residing pathogens modulate the UPR in dendritic cells (DCs) and thereby affect T cell-mediated immunity remains uncharacterized. Here, we demonstrate that infection of DCs with Toxoplasma gondii (T. gondii) triggers a unique UPR signature hallmarked by the MyD88-dependent activation of the IRE1α pathway and the inhibition of the ATF6 pathway. Induction of XBP1s controls pro-inflammatory cytokine secretion in infected DCs, while IRE1α promotes MHCI antigen presentation of secreted parasite antigens. In mice, infection leads to a specific activation of the IRE1α pathway, which is restricted to the cDC1 subset. Mice deficient for IRE1α and XBP1 in DCs display a severe susceptibility to T. gondii and succumb during the acute phase of the infection. This early mortality is correlated with increased parasite burden and a defect in splenic T-cell responses. Thus, we identify the IRE1α/XBP1s branch of the UPR as a key regulator of host defense upon T. gondii infection.
Collapse
Affiliation(s)
- Anaïs F Poncet
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Victor Bosteels
- Laboratory for ER stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Eik Hoffmann
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Sylia Chehade
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Sofie Rennen
- Laboratory for ER stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Ludovic Huot
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Véronique Peucelle
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Sandra Maréchal
- Laboratory for ER stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Jamal Khalife
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Nicolas Blanchard
- Centre de Physiopathologie Toulouse Purpan (CPTP), INSERM, CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Sophie Janssens
- Laboratory for ER stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sabrina Marion
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
24
|
Ham DW, Kim SG, Seo SH, Shin JH, Lee SH, Shin EH. Chronic Toxoplasma gondii Infection Alleviates Experimental Autoimmune Encephalomyelitis by the Immune Regulation Inducing Reduction in IL-17A/Th17 Via Upregulation of SOCS3. Neurotherapeutics 2021; 18:430-447. [PMID: 33205383 PMCID: PMC8116467 DOI: 10.1007/s13311-020-00957-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2020] [Indexed: 12/13/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a mouse model of multiple sclerosis (MS), a demyelinating autoimmune disease caused by the infiltration of a harmful autoreactive Th1 and Th17 cells. To mitigate MS, which is impossible to cure with medication only, immunomodulatory interventions that prevent Th17 cell activation are ideal. The objective of the present study was to analyze the effect of Toxoplasma gondii infection on the onset of EAE. Our results found that Toxoplasma gondii infection in the brain increases SOCS3 expression and decreases the phosphorylation of STAT3, resulting in reducing IL-17A and IL-23, which suppress the differentiation and expansion of pathogenic Th17 cells, an important factor in MS development. These immune responses resulted in a reduction in the clinical scoring of EAE induced by myelin oligodendrocyte glycoprotein 35-55 immunization. In the EAE group with T. gondii infection (Tg + EAE group), Th17-related immune responses that exacerbate the onset of EAE were reduced compared to those in the EAE group. This study suggests that the alleviation of EAE after T. gondii infection is regulated in a SOCS3/STAT3/IL-17A/blood-brain barrier integrity-dependent manner. Although parasite infection would not be permitted for MS treatment, this study using T. gondii infection identified potential targets that contribute to disease attenuation.
Collapse
Affiliation(s)
- Do-Won Ham
- Department of Tropical Medicine and Parasitology, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul, 03080, Republic of Korea
| | - Sang-Gyun Kim
- Department of Tropical Medicine and Parasitology, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul, 03080, Republic of Korea
| | - Seung-Hwan Seo
- Department of Tropical Medicine and Parasitology, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul, 03080, Republic of Korea
| | - Ji-Hun Shin
- Department of Tropical Medicine and Parasitology, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul, 03080, Republic of Korea
| | - Sang Hyung Lee
- Department of Neurosurgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, 07061, Republic of Korea.
| | - Eun-Hee Shin
- Department of Tropical Medicine and Parasitology, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul, 03080, Republic of Korea.
- Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea.
| |
Collapse
|
25
|
Mukhopadhyay D, Arranz-Solís D, Saeij JPJ. Influence of the Host and Parasite Strain on the Immune Response During Toxoplasma Infection. Front Cell Infect Microbiol 2020; 10:580425. [PMID: 33178630 PMCID: PMC7593385 DOI: 10.3389/fcimb.2020.580425] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/11/2020] [Indexed: 01/02/2023] Open
Abstract
Toxoplasma gondii is an exceptionally successful parasite that infects a very broad host range, including humans, across the globe. The outcome of infection differs remarkably between hosts, ranging from acute death to sterile infection. These differential disease patterns are strongly influenced by both host- and parasite-specific genetic factors. In this review, we discuss how the clinical outcome of toxoplasmosis varies between hosts and the role of different immune genes and parasite virulence factors, with a special emphasis on Toxoplasma-induced ileitis and encephalitis.
Collapse
Affiliation(s)
| | | | - Jeroen P. J. Saeij
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
26
|
Salvioni A, Belloy M, Lebourg A, Bassot E, Cantaloube-Ferrieu V, Vasseur V, Blanié S, Liblau RS, Suberbielle E, Robey EA, Blanchard N. Robust Control of a Brain-Persisting Parasite through MHC I Presentation by Infected Neurons. Cell Rep 2020; 27:3254-3268.e8. [PMID: 31189109 DOI: 10.1016/j.celrep.2019.05.051] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 03/03/2019] [Accepted: 05/15/2019] [Indexed: 12/27/2022] Open
Abstract
Control of CNS pathogens by CD8 T cells is key to avoid fatal neuroinflammation. Yet, the modalities of MHC I presentation in the brain are poorly understood. Here, we analyze the antigen presentation mechanisms underlying CD8 T cell-mediated control of the Toxoplasma gondii parasite in the CNS. We show that MHC I presentation of an efficiently processed model antigen (GRA6-OVA), even when not expressed in the bradyzoite stage, reduces cyst burden and dampens encephalitis in C57BL/6 mice. Antigen presentation assays with infected primary neurons reveal a correlation between lower MHC I presentation of tachyzoite antigens by neurons and poor parasite control in vivo. Using conditional MHC I-deficient mice, we find that neuronal MHC I presentation is required for robust restriction of T. gondii in the CNS during chronic phase, showing the importance of MHC I presentation by CNS neurons in the control of a prevalent brain pathogen.
Collapse
Affiliation(s)
- Anna Salvioni
- Center for Pathophysiology Toulouse-Purpan (CPTP), INSERM, CNRS, University of Toulouse, 31024 Toulouse, France
| | - Marcy Belloy
- Center for Pathophysiology Toulouse-Purpan (CPTP), INSERM, CNRS, University of Toulouse, 31024 Toulouse, France
| | - Aurore Lebourg
- Center for Pathophysiology Toulouse-Purpan (CPTP), INSERM, CNRS, University of Toulouse, 31024 Toulouse, France
| | - Emilie Bassot
- Center for Pathophysiology Toulouse-Purpan (CPTP), INSERM, CNRS, University of Toulouse, 31024 Toulouse, France
| | - Vincent Cantaloube-Ferrieu
- Center for Pathophysiology Toulouse-Purpan (CPTP), INSERM, CNRS, University of Toulouse, 31024 Toulouse, France
| | - Virginie Vasseur
- Center for Pathophysiology Toulouse-Purpan (CPTP), INSERM, CNRS, University of Toulouse, 31024 Toulouse, France
| | - Sophie Blanié
- Center for Pathophysiology Toulouse-Purpan (CPTP), INSERM, CNRS, University of Toulouse, 31024 Toulouse, France
| | - Roland S Liblau
- Center for Pathophysiology Toulouse-Purpan (CPTP), INSERM, CNRS, University of Toulouse, 31024 Toulouse, France
| | - Elsa Suberbielle
- Center for Pathophysiology Toulouse-Purpan (CPTP), INSERM, CNRS, University of Toulouse, 31024 Toulouse, France
| | - Ellen A Robey
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Nicolas Blanchard
- Center for Pathophysiology Toulouse-Purpan (CPTP), INSERM, CNRS, University of Toulouse, 31024 Toulouse, France.
| |
Collapse
|
27
|
Batista SJ, Still KM, Johanson D, Thompson JA, OʼBrien CA, Lukens JR, Harris TH. Gasdermin-D-dependent IL-1α release from microglia promotes protective immunity during chronic Toxoplasma gondii infection. Nat Commun 2020; 11:3687. [PMID: 32703941 PMCID: PMC7378823 DOI: 10.1038/s41467-020-17491-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/29/2020] [Indexed: 12/20/2022] Open
Abstract
Microglia, resident immune cells of the CNS, are thought to defend against infections. Toxoplasma gondii is an opportunistic infection that can cause severe neurological disease. Here we report that during T. gondii infection a strong NF-κB and inflammatory cytokine transcriptional signature is overrepresented in blood-derived macrophages versus microglia. Interestingly, IL-1α is enriched in microglia and IL-1β in macrophages. We find that mice lacking IL-1R1 or IL-1α, but not IL-1β, have impaired parasite control and immune cell infiltration within the brain. Further, we show that microglia, not peripheral myeloid cells, release IL-1α ex vivo. Finally, we show that ex vivo IL-1α release is gasdermin-D dependent, and that gasdermin-D and caspase-1/11 deficient mice show deficits in brain inflammation and parasite control. These results demonstrate that microglia and macrophages are differently equipped to propagate inflammation, and that in chronic T. gondii infection, microglia can release the alarmin IL-1α, promoting neuroinflammation and parasite control. Control over T. gondii infection in the brain involves microglial cells, but how these cells execute this control is not clear. Here the authors show that unlike IL-1β dominant macrophages, microglia are primed for gasdermin-D-dependent IL-1α production that is critical for protection against T. gondii infection.
Collapse
Affiliation(s)
- Samantha J Batista
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, 22908, USA
| | - Katherine M Still
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, 22908, USA
| | - David Johanson
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jeremy A Thompson
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, 22908, USA
| | - Carleigh A OʼBrien
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, 22908, USA
| | - John R Lukens
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, 22908, USA
| | - Tajie H Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
28
|
Chronic toxoplasmosis and sleepiness in obstructive sleep apnea: Is there a link? PLoS One 2020; 15:e0235463. [PMID: 32609758 PMCID: PMC7329112 DOI: 10.1371/journal.pone.0235463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 06/16/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Sleepiness is the main clinical expression of obstructive sleep apnea (OSA) syndrome resulting from upper airway collapse. Recent studies have discussed the fact that the presence of T. gondii cysts in the brain and the resulting biochemical and immunological mechanisms could be linked to neurobehavioral disorders. The aim of the present study was to explore the potential impact of chronic toxoplasmosis on sleepiness and on obstructive sleep apnea (OSA) severity in OSA obese patients. MATERIALS AND METHODS A case control study on obese patients screened for OSA was performed. According to the sleep disorder and matched based on gender, age and body mass index (BMI), two groups of obese patients were selected from our sample collection database. All patients were tested for toxoplasmosis serological status measuring anti-Toxoplasma IgG and IgM levels. Univariable and multivariable logistic regression models were performed to assess the impact of chronic toxoplasmosis on sleepiness and OSA severity. RESULTS 107 obese patients suffering from OSA were included in the study (median age: 53.3 years Interquartile range (IQR): [41.9-59.9]; median BMI: 39.4 kg/m2 IQR: [35.5-44.1], apnea-hypopnea index = 27.5 events/h [10.7-49.9]). Chronic toxoplasmosis was present in 63.4% and 70.7% of patients with or without sleepiness (p = 0.48), respectively and was not associated either to sleepiness (OR: 0.76, 95% CI: [0.52; 2.33], p = 0.64) or OSA severity (OR = 1.75, 95% CI: [0.51; 5.98] p = 0.37). CONCLUSION Although chronic Toxoplasma infection in immunocompetent humans has been associated to several behavioral disorders or pathologies in recent literature, we demonstrate here that chronic toxoplasmosis is not associated to sleepiness and to sleep apnea syndrome severity in obese patients suspected of sleep apnea syndrome.
Collapse
|
29
|
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]
|
30
|
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
|
31
|
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: 12] [Impact Index Per Article: 2.4] [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
|
32
|
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
|
33
|
Brüne M. Schizophrenia as parasitic behavior manipulation: can we put together the pieces of an evolutionary puzzle? World Psychiatry 2020; 19:119-120. [PMID: 31922668 PMCID: PMC6953588 DOI: 10.1002/wps.20637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Martin Brüne
- Department of Psychiatry, Psychotherapy and Preventive Medicine, Ruhr-University Bochum, LWL University Hospital, Bochum, Germany
| |
Collapse
|
34
|
Kano SI, Hodgkinson CA, Jones-Brando L, Eastwood S, Ishizuka K, Niwa M, Choi EY, Chang DJ, Chen Y, Velivela SD, Leister F, Wood J, Chowdari K, Ducci F, Caycedo DA, Heinz E, Newman ER, Cascella N, Mortensen PB, Zandi PP, Dickerson F, Nimgaonkar V, Goldman D, Harrison PJ, Yolken RH, Sawa A. Host-parasite interaction associated with major mental illness. Mol Psychiatry 2020; 25:194-205. [PMID: 30127472 PMCID: PMC6382596 DOI: 10.1038/s41380-018-0217-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 04/16/2018] [Accepted: 06/20/2018] [Indexed: 11/23/2022]
Abstract
Clinical studies frequently report that patients with major mental illness such as schizophrenia and bipolar disorder have co-morbid physical conditions, suggesting that systemic alterations affecting both brain and peripheral tissues might underlie the disorders. Numerous studies have reported elevated levels of anti-Toxoplasma gondii (T. gondii) antibodies in patients with major mental illnesses, but the underlying mechanism was unclear. Using multidisciplinary epidemiological, cell biological, and gene expression profiling approaches, we report here multiple lines of evidence suggesting that a major mental illness-related susceptibility factor, Disrupted in schizophrenia (DISC1), is involved in host immune responses against T. gondii infection. Specifically, our cell biology and gene expression studies have revealed that DISC1 Leu607Phe variation, which changes DISC1 interaction with activating transcription factor 4 (ATF4), modifies gene expression patterns upon T. gondii infection. Our epidemiological data have also shown that DISC1 607 Phe/Phe genotype was associated with higher T. gondii antibody levels in sera. Although further studies are required, our study provides mechanistic insight into one of the few well-replicated serological observations in major mental illness.
Collapse
Affiliation(s)
- Shin-Ichi Kano
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Colin A Hodgkinson
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Lorraine Jones-Brando
- Stanley Division of Developmental Neurovirology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sharon Eastwood
- Department of Psychiatry, University of Oxford, Oxford, OX3 7JX, United Kingdom
| | - Koko Ishizuka
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Minae Niwa
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Eric Y Choi
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Daniel J Chang
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Yian Chen
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Swetha D Velivela
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Flora Leister
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Joel Wood
- Departments of Psychiatry and Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Kodavali Chowdari
- Departments of Psychiatry and Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Francesca Ducci
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Daniel A Caycedo
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Elizabeth Heinz
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Emily R Newman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Nicola Cascella
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Preben B Mortensen
- National Centre for Register-Based Research, University of Aarhus, Aarhus, 8000, Denmark
| | - Peter P Zandi
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Faith Dickerson
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, 21204, USA
| | - Vishwajit Nimgaonkar
- Departments of Psychiatry and Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, OX3 7JX, United Kingdom
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Akira Sawa
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
35
|
Imaging the dynamic recruitment of monocytes to the blood-brain barrier and specific brain regions during Toxoplasma gondii infection. Proc Natl Acad Sci U S A 2019; 116:24796-24807. [PMID: 31727842 DOI: 10.1073/pnas.1915778116] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Brain infection by the parasite Toxoplasma gondii in mice is thought to generate vulnerability to predation by mechanisms that remain elusive. Monocytes play a key role in host defense and inflammation and are critical for controlling T. gondii However, the dynamic and regional relationship between brain-infiltrating monocytes and parasites is unknown. We report the mobilization of inflammatory (CCR2+Ly6Chi) and patrolling (CX3CR1+Ly6Clo) monocytes into the blood and brain during T. gondii infection of C57BL/6J and CCR2RFP/+CX3CR1GFP/+ mice. Longitudinal analysis of mice using 2-photon intravital imaging of the brain through cranial windows revealed that CCR2-RFP monocytes were recruited to the blood-brain barrier (BBB) within 2 wk of T. gondii infection, exhibited distinct rolling and crawling behavior, and accumulated within the vessel lumen before entering the parenchyma. Optical clearing of intact T. gondii-infected brains using iDISCO+ and light-sheet microscopy enabled global 3D detection of monocytes. Clusters of T. gondii and individual monocytes across the brain were identified using an automated cell segmentation pipeline, and monocytes were found to be significantly correlated with sites of T. gondii clusters. Computational alignment of brains to the Allen annotated reference atlas [E. S. Lein et al., Nature 445:168-176 (2007)] indicated a consistent pattern of monocyte infiltration during T. gondii infection to the olfactory tubercle, in contrast to LPS treatment of mice, which resulted in a diffuse distribution of monocytes across multiple brain regions. These data provide insights into the dynamics of monocyte recruitment to the BBB and the highly regionalized localization of monocytes in the brain during T. gondii CNS infection.
Collapse
|
36
|
Benevides L, Saltarelli VM, Pioto F, Sacramento LA, Dias MS, Rodríguez GR, Viola JPB, Carregaro V, Silva JS. NFAT1 Regulates Ly6C hi Monocyte Recruitment to the CNS and Plays an Essential Role in Resistance to Toxoplasma gondii Infection. Front Immunol 2019; 10:2105. [PMID: 31555297 PMCID: PMC6742953 DOI: 10.3389/fimmu.2019.02105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/21/2019] [Indexed: 12/24/2022] Open
Abstract
Monocytes play key roles in the maintenance of homeostasis and in the control of the infection. Monocytes are recruited from the bone marrow to inflammatory sites and are essential for antimicrobial activity to limit tissue damage and promote adaptive T cell responses. Here, we investigated the role of Nuclear Factor of Activated T cells 1 (NFAT1) in the regulation of Ly6Chi inflammatory monocyte recruitment to the CNS upon T. gondii infection. We show that NFAT-1-deficient monocytes are unable to migrate to the CNS of T. gondii-infected mice. Moreover, NFAT1−/− mice are highly susceptible to chronic T. gondii infection due to a failure to control parasite replication in the CNS. The inhibition of Ly6Chi inflammatory monocyte recruitment to the CNS severely blocked CXCL10 production and consequently the migration of IFN-γ-producing CD4+ T cells. Moreover, the transfer of Ly6Chi monocytes to infected NFAT1−/− mice favored CD4+ T cell migration to the CNS and resulted in the inhibition of parasite replication and host defense. Together, these results demonstrated for the first time the contribution of NFAT1 to the regulation of Ly6Chi monocyte recruitment to the CNS and to resistance during chronic T. gondii infection.
Collapse
Affiliation(s)
- Luciana Benevides
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Fiocruz-Bi-Institutional Translational Medicine Plataform, Ribeirão Preto, Brazil
| | - Verônica M Saltarelli
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Franciele Pioto
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Fiocruz-Bi-Institutional Translational Medicine Plataform, Ribeirão Preto, Brazil
| | - Laís A Sacramento
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Murilo S Dias
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gretel R Rodríguez
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João P B Viola
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Vanessa Carregaro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João S Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Fiocruz-Bi-Institutional Translational Medicine Plataform, Ribeirão Preto, Brazil
| |
Collapse
|
37
|
French T, Düsedau HP, Steffen J, Biswas A, Ahmed N, Hartmann S, Schüler T, Schott BH, Dunay IR. Neuronal impairment following chronic Toxoplasma gondii infection is aggravated by intestinal nematode challenge in an IFN-γ-dependent manner. J Neuroinflammation 2019; 16:159. [PMID: 31352901 PMCID: PMC6661741 DOI: 10.1186/s12974-019-1539-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/09/2019] [Indexed: 02/06/2023] Open
Abstract
Background It has become increasingly evident that the immune and nervous systems are closely intertwined, relying on one another during regular homeostatic conditions. Prolonged states of imbalance between neural and immune homeostasis, such as chronic neuroinflammation, are associated with a higher risk for neural damage. Toxoplasma gondii is a highly successful neurotropic parasite causing persistent subclinical neuroinflammation, which is associated with psychiatric and neurodegenerative disorders. Little is known, however, by what means neuroinflammation and the associated neural impairment can be modulated by peripheral inflammatory processes. Methods Expression of immune and synapse-associated genes was assessed via quantitative real-time PCR to investigate how T. gondii infection-induced chronic neuroinflammation and associated neuronal alterations can be reshaped by a subsequent acute intestinal nematode co-infection. Immune cell subsets were characterized via flow cytometry in the brain of infected mice. Sulfadiazine and interferon-γ-neutralizing antibody were applied to subdue neuroinflammation. Results Neuroinflammation induced by T. gondii infection of mice was associated with increased microglia activation, recruitment of immune cells into the brain exhibiting Th1 effector functions, and enhanced production of Th1 and pro-inflammatory molecules (IFN-γ, iNOS, IL-12, TNF, IL-6, and IL-1β) following co-infection with Heligmosomoides polygyrus. The accelerated cerebral Th1 immune response resulted in enhanced T. gondii removal but exacerbated the inflammation-related decrease of synapse-associated gene expression. Synaptic proteins EAAT2 and GABAAα1, which are involved in the excitation/inhibition balance in the CNS, were affected in particular. These synaptic alterations were partially recovered by reducing neuroinflammation indirectly via antiparasitic treatment and especially by application of IFN-γ-neutralizing antibody. Impaired iNOS expression following IFN-γ neutralization directly affected EAAT2 and GABAAα1 signaling, thus contributing to the microglial regulation of neurons. Besides, reduced CD36, TREM2, and C1qa gene expression points toward inflammation induced synaptic pruning as a fundamental mechanism. Conclusion Our results suggest that neuroimmune responses following chronic T. gondii infection can be modulated by acute enteric nematode co-infection. While consecutive co-infection promotes parasite elimination in the CNS, it also adversely affects gene expression of synaptic proteins, via an IFN-γ-dependent manner. Electronic supplementary material The online version of this article (10.1186/s12974-019-1539-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Timothy French
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Henning Peter Düsedau
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Johannes Steffen
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Aindrila Biswas
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Norus Ahmed
- Department of Veterinary Medicine, Institute of Immunology, Free University Berlin, Berlin, Germany
| | - Susanne Hartmann
- Department of Veterinary Medicine, Institute of Immunology, Free University Berlin, Berlin, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Björn H Schott
- Leibniz Institute of Neurobiology, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany.,Department of Psychiatry and Psychotherapy, University Medicine Göttingen, Göttingen, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|
38
|
Figueiredo CA, Düsedau HP, Steffen J, Gupta N, Dunay MP, Toth GK, Reglodi D, Heimesaat MM, Dunay IR. Immunomodulatory Effects of the Neuropeptide Pituitary Adenylate Cyclase-Activating Polypeptide in Acute Toxoplasmosis. Front Cell Infect Microbiol 2019; 9:154. [PMID: 31192159 PMCID: PMC6546896 DOI: 10.3389/fcimb.2019.00154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/26/2019] [Indexed: 12/21/2022] Open
Abstract
Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) is an endogenous neuropeptide with distinct functions including the regulation of inflammatory processes. PACAP is able to modify the immune response by directly regulating macrophages and monocytes inhibiting the production of inflammatory cytokines, chemokines and free radicals. Here, we analyzed the effect of exogenous PACAP on peripheral immune cell subsets upon acute infection with the parasite Toxoplasma gondii (T. gondii). PACAP administration was followed by diminished innate immune cell recruitment to the peritoneal cavity of T. gondii-infected mice. PACAP did not directly interfere with parasite replication, instead, indirectly reduced parasite burden in mononuclear cell populations by enhancing their phagocytic capacity. Although proinflammatory cytokine levels were attenuated in the periphery upon PACAP treatment, interleukin (IL)-10 and Transforming growth factor beta (TGF-β) remained stable. While PACAP modulated VPAC1 and VPAC2 receptors in immune cells upon binding, it also increased their expression of brain-derived neurotrophic factor (BDNF). In addition, the expression of p75 neurotrophin receptor (p75NTR) on Ly6Chi inflammatory monocytes was diminished upon PACAP administration. Our findings highlight the immunomodulatory effect of PACAP on peripheral immune cell subsets during acute Toxoplasmosis, providing new insights about host-pathogen interaction and the effects of neuropeptides during inflammation.
Collapse
Affiliation(s)
- Caio Andreeta Figueiredo
- Medical Faculty, Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Henning Peter Düsedau
- Medical Faculty, Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Johannes Steffen
- Medical Faculty, Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Nishith Gupta
- Faculty of Life Sciences, Institute of Biology, Humboldt University, Berlin, Germany
| | - Miklos Pal Dunay
- Department and Clinic of Surgery and Ophthalmology, University of Veterinary Medicine, Budapest, Hungary
| | - Gabor K Toth
- Department of Medical Chemistry, University of Szeged, Szeged, Hungary
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Team, University of Pecs Medical School, Pecs, Hungary
| | - Markus M Heimesaat
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Ildiko Rita Dunay
- Medical Faculty, Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences - CBBS, Magdeburg, Germany
| |
Collapse
|
39
|
Düsedau HP, Kleveman J, Figueiredo CA, Biswas A, Steffen J, Kliche S, Haak S, Zagrebelsky M, Korte M, Dunay IR. p75 NTR regulates brain mononuclear cell function and neuronal structure in Toxoplasma infection-induced neuroinflammation. Glia 2019; 67:193-211. [PMID: 30597659 PMCID: PMC6590406 DOI: 10.1002/glia.23553] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 09/28/2018] [Accepted: 10/05/2018] [Indexed: 12/13/2022]
Abstract
Neurotrophins mediate neuronal growth, differentiation, and survival via tropomyosin receptor kinase (Trk) or p75 neurotrophin receptor (p75NTR) signaling. The p75NTR is not exclusively expressed by neurons but also by certain immune cells, implying a role for neurotrophin signaling in the immune system. In this study, we investigated the effect of p75NTR on innate immune cell behavior and on neuronal morphology upon chronic Toxoplasma gondii (T. gondii) infection‐induced neuroinflammation. Characterization of the immune cells in the periphery and central nervous system (CNS) revealed that innate immune cell subsets in the brain upregulated p75NTR upon infection in wild‐type mice. Although cell recruitment and phagocytic capacity of p75NTRexonIV knockout (p75−/−) mice were not impaired, the activation status of resident microglia and recruited myeloid cell subsets was altered. Importantly, recruited mononuclear cells in brains of infected p75−/− mice upregulated the production of the cytokines interleukin (IL)‐10, IL‐6 as well as IL‐1α. Protein levels of proBDNF, known to negatively influence neuronal morphology by binding p75NTR, were highly increased upon chronic infection in the brain of wild‐type and p75−/− mice. Moreover, upon infection the activated immune cells contributed to the proBDNF release. Notably, the neuroinflammation‐induced changes in spine density were rescued in the p75−/− mice. In conclusion, these findings indicate that neurotrophin signaling via the p75NTR affects innate immune cell behavior, thus, influencing the structural plasticity of neurons under inflammatory conditions.
Collapse
Affiliation(s)
- Henning Peter Düsedau
- Otto-von-Guericke University Magdeburg, Institute of Inflammation and Neurodegeneration, Medical Faculty, Magdeburg, Germany
| | - Jan Kleveman
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
| | - Caio Andreeta Figueiredo
- Otto-von-Guericke University Magdeburg, Institute of Inflammation and Neurodegeneration, Medical Faculty, Magdeburg, Germany
| | - Aindrila Biswas
- Otto-von-Guericke University Magdeburg, Institute of Inflammation and Neurodegeneration, Medical Faculty, Magdeburg, Germany
| | - Johannes Steffen
- Otto-von-Guericke University Magdeburg, Institute of Inflammation and Neurodegeneration, Medical Faculty, Magdeburg, Germany
| | - Stefanie Kliche
- Otto-von-Guericke University, Institute for Molecular and Clinical Immunology, Medical Faculty, Magdeburg, Germany
| | - Stefan Haak
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
| | - Marta Zagrebelsky
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
| | - Ildiko Rita Dunay
- Otto-von-Guericke University Magdeburg, Institute of Inflammation and Neurodegeneration, Medical Faculty, Magdeburg, Germany
| |
Collapse
|
40
|
Norins LC. The Beehive Theory: Role of microorganisms in late sequelae of traumatic brain injury and chronic traumatic encephalopathy. Med Hypotheses 2019; 128:1-5. [PMID: 31203899 DOI: 10.1016/j.mehy.2019.04.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022]
Abstract
Traumatic brain injury and chronic traumatic encephalopathy are both major health problems, well-publicized for the severe delayed effects attributed to them, including cognitive decline, psychiatric disorders, seizures, impaired motor function, and personality changes. For convenience, the two afflictions are considered together under the rubric traumatic brain injury. Despite the need for neuroprotective agents, no substances have shown efficacy in clinical studies. Thus, a deeper understanding of the neuropathological mechanism of such injury is still needed. Proposed here is a theory that microorganisms from within the brain and elsewhere in the body contribute to the long-term neurological deterioration characteristic of traumatic brain injury. The label, "The Beehive Theory", is drawn from the well-known fact that disturbing a tranquil beehive with a blow can cause a swarm of angry bees to exit their dwelling place and attack nearby humans. Similarly, an impact to the head can initiate dislocations and disruptions in the microbiota present in the brain and body. First, since the normal human brain is not sterile, but is host to a variety of microorganisms, blows to the skull may dislodge them from their accustomed local environments, in which they have been living in quiet equilibrium with neighboring brain cells. Deleterious substances may be released by the displaced microbes, including metabolic products and antigens. Second, upon impact commensal microbes already resident on surfaces of the nose, mouth, and eyes, and potentially harmful organisms from the environment, may gain access to the brain through the distal ends of the olfactory and optic nerves or even a disrupted blood-brain barrier. Third, microbes dwelling in more distant parts of the body may be propelled through the walls of local blood vessels into the bloodstream, and then leak out into damaged areas of the brain that have increased blood-brain barrier permeability. Fourth, the impact may cause dysbiosis in the gastrointestinal microbiome, thereby disrupting signaling via the gut-brain axis. Possible preventatives or therapeutics that would address the adverse contributions of microbes to the late sequelae of traumatic brain injury include anti-inflammatories, antibacterials, antivirals, and probiotics.
Collapse
Affiliation(s)
- Leslie C Norins
- Alzheimer's Germ Quest, Inc., 4301 Gulfshore Blvd, Suite 1404, Naples, FL 34103, USA.
| |
Collapse
|
41
|
Jesus LB, Santos AB, Jesus EEV, Santos RGD, Grangeiro MS, Bispo-da-Silva A, Arruda MR, Argolo DS, Pinheiro AM, El-Bachá RS, Costa SL, Costa MFD. IDO, COX and iNOS have an important role in the proliferation of Neospora caninum in neuron/glia co-cultures. Vet Parasitol 2019; 266:96-102. [PMID: 30736955 DOI: 10.1016/j.vetpar.2019.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 01/03/2019] [Accepted: 01/05/2019] [Indexed: 12/23/2022]
Abstract
Central nervous system (CNS) is the main site for encystment of Neospora caninum in different animal species. In this tissue, glial cells (astrocytes and microglia) modulate responses to aggression in order to preserve homeostasis and neuronal function. Previous data showed that when primary cultures of glial cells are infected with N. caninum, they develop gliosis and the immune response is characterized by the release of TNF and IL-10, followed by the control of parasite proliferation. In order to elucidate this control, three enzymatic systems involved in parasite-versus-host interactions were observed on a model of neuron/glia co/cultures obtained from rat brains. Indoleamine 2,3-dioxygenase (IDO), induced nitric oxide synthase (iNOS) responsible for the catabolism of tryptophan and arginine, respectively, and cycloxigenase (COX) were studied comparing their modulation by respective inhibitors with the number of tachyzoites or the immune response measured by the release of IL-10 and TNF. Cells were treated with the inhibitors of iNOS (1.5 mM L-NAME), IDO (1 mM 1-methyl tryptophan), COX-1 (1 μM indomethacin) and COX-2 (1 μM nimesulide) before infection with tachyzoites of N. caninum (1:1 cell: parasite). After 72 h of infection, immunocytochemistry showed astrogliosis and a significant increase in the number and length of neurites, compared with uninfected co-cultures, while an increase of IL-10 and TNF was verified. N. caninum did not change iNOS activity, but the inhibition of the basal levels of this enzyme stimulated parasite proliferation. Additionally, a significant increase of about 40% was verified in the IDO activity, whose inhibition caused 1.2-fold increase in parasitic growth. For COX-2 activity, infection of cultures stimulated a significant increase in release of PGE2 and its inhibition by nimesulide allowed the parasitic growth. These data indicate that iNOS, IDO and COX-2 control the proliferation of N. caninum in this in vitro model. On the other hand, the release of IL-10 by glia besides modulating the inflammation also allow the continuity of parasitism.
Collapse
Affiliation(s)
- L B Jesus
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - A B Santos
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - E E V Jesus
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - R G D Santos
- Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - M S Grangeiro
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - A Bispo-da-Silva
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - M R Arruda
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - D S Argolo
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - A M Pinheiro
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil; Centro de Ciências Agrárias Ambientais e Biológica, Universidade do Recôncavo da Bahia - URBA, R. Ruy Barbosa 710 Centro, CEP 44380-000, Cruz das Almas, Bahia, Brazil
| | - R S El-Bachá
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil; INCT de Neurociência Translacional (INNT)- CNPq, Brazil
| | - S L Costa
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil; INCT de Neurociência Translacional (INNT)- CNPq, Brazil.
| | - M F D Costa
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil; INCT de Neurociência Translacional (INNT)- CNPq, Brazil.
| |
Collapse
|
42
|
Huang WY, Wang YP, Mahmmod YS, Wang JJ, Liu TH, Zheng YX, Zhou X, Zhang XX, Yuan ZG. A Double-Edged Sword: Complement Component 3 in Toxoplasma gondii Infection. Proteomics 2019; 19:e1800271. [PMID: 30515942 DOI: 10.1002/pmic.201800271] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 11/01/2018] [Indexed: 12/14/2022]
Abstract
Sprague Dawley rats and Kunming (KM) mice are artificially infected with type II Toxoplasma gondii strain Prugniaud (Pru) to generate toxoplasmosis, which is a fatal disease mediated by T. gondii invasion of the central nervous system (CNS) by unknown mechanisms. The aim is to explore the mechanism of differential susceptibility of mice and rats to T. gondii infection. Therefore, a strategy of isobaric tags for relative and absolute quantitation (iTRAQ) is established to identify differentially expressed proteins (DEPs) in the rats' and the mice's brains compared to the healthy groups. In KM mice, which is susceptible to T. gondii infection, complement component 3 (C3) is upregulated and the tight junction (TJ) pathway shows a disorder. It is presumed that T. gondii-stimulated C3 disrupts the TJ of the blood-brain barrier in the CNS. This effect allows more T. gondii passing to the brain through the intercellular space.
Collapse
Affiliation(s)
- Wan-Yi Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China
| | - Ya-Pei Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China
| | - Yasser S Mahmmod
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA), Campus de la Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Infectious Diseases, Department of Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, 44511, Zagazig, Sharkia Province, Egypt
| | - Jun-Jie Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China
| | - Tang-Hui Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China
| | - Yu-Xiang Zheng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China
| | - Xue Zhou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China
| | - Xiu-Xiang Zhang
- College of Agriculture, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China
| | - Zi-Guo Yuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China
| |
Collapse
|
43
|
Impact of Engineered Expression of Mitochondrial Association Factor 1b on Toxoplasma gondii Infection and the Host Response in a Mouse Model. mSphere 2018; 3:3/5/e00471-18. [PMID: 30333181 PMCID: PMC6193605 DOI: 10.1128/msphere.00471-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The parasite Toxoplasma gondii currently infects approximately one-third of the world’s population and causes life-threatening toxoplasmosis in individuals with undeveloped or weakened immune systems. Current treatments are unable to cure T. gondii infection, leaving infected individuals with slow-growing tissue cysts for the remainder of their lives. Previous work has shown that expression of the parasite protein mitochondrial association factor 1 (MAF1b) is responsible for the association of T. gondii parasites with host mitochondria and provides a selective advantage during acute infection. Here we examine the impact of MAF1b expression during chronic T. gondii infection. We find that mice infected with MAF1b-expressing parasites have higher cyst burden and cytokine levels than their wild-type counterparts. A better understanding of the genes involved in establishing and maintaining chronic infection will aid in discovering effective therapeutics for chronically infected individuals. The opportunistic intracellular parasite Toxoplasma gondii causes a lifelong chronic infection capable of reactivating in immunocompromised individuals, which can lead to life-threatening complications. Following invasion of the host cell, host mitochondria associate with the parasitophorous vacuole membrane. This phenotype is T. gondii strain specific and is mediated by expression of a host mitochondrial association-competent (HMA+) paralog of the parasite protein mitochondrial association factor 1 (MAF1b). Previous work demonstrated that expression of MAF1b in strains that do not normally associate with host mitochondria increases their fitness during acute infection in vivo. However, the impact of MAF1b expression during chronic T. gondii infection is unclear. In this study, we assess the impact of MAF1b expression on cyst formation and cytokine production in mice. Despite generally low numbers of cysts generated by the in vitro culture-adapted strains used in this study, we find that parasites expressing MAF1b have higher numbers of cysts in the brains of chronically infected mice and that MAF1b+ cyst burden significantly increases during the course of chronic infection. Consistent with this, mice infected with MAF1b+ parasites have higher levels of the serum cytokines RANTES and VEGF (vascular endothelial growth factor) at day 57 postinfection, although this could be due to higher parasite burden at this time point rather than direct manipulation of these cytokines by MAF1b. Overall these data indicate that MAF1b expression may also be important in determining infection outcome during the chronic phase, either by directly altering the cytokine/signaling environment or by increasing proliferation during the acute and/or chronic phase. IMPORTANCE The parasite Toxoplasma gondii currently infects approximately one-third of the world’s population and causes life-threatening toxoplasmosis in individuals with undeveloped or weakened immune systems. Current treatments are unable to cure T. gondii infection, leaving infected individuals with slow-growing tissue cysts for the remainder of their lives. Previous work has shown that expression of the parasite protein mitochondrial association factor 1 (MAF1b) is responsible for the association of T. gondii parasites with host mitochondria and provides a selective advantage during acute infection. Here we examine the impact of MAF1b expression during chronic T. gondii infection. We find that mice infected with MAF1b-expressing parasites have higher cyst burden and cytokine levels than their wild-type counterparts. A better understanding of the genes involved in establishing and maintaining chronic infection will aid in discovering effective therapeutics for chronically infected individuals.
Collapse
|
44
|
Hu RS, He JJ, Elsheikha HM, Zhang FK, Zou Y, Zhao GH, Cong W, Zhu XQ. Differential Brain MicroRNA Expression Profiles After Acute and Chronic Infection of Mice With Toxoplasma gondii Oocysts. Front Microbiol 2018; 9:2316. [PMID: 30333806 PMCID: PMC6176049 DOI: 10.3389/fmicb.2018.02316] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/11/2018] [Indexed: 12/16/2022] Open
Abstract
Brain microRNAs (miRNAs) change in abundance in response to Toxoplasma gondii infection. However, their precise role in the pathogenesis of cerebral infection with T. gondii oocyst remains unclear. We studied the abundance of miRNAs in the brain of mice on days 11 and 33 post-infection (dpi) in order to identify miRNA pattern specific to early (11 dpi) and late (33 dpi) T. gondii infection. Mice were challenged with T. gondii oocysts (Type II strain) and on 11 and 33 dpi, the expression of miRNAs in mouse brain was investigated using small RNA (sRNA) sequencing. miRNA expression was confirmed by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed to identify the biological processes, molecular functions, and cellular components, as well as pathways involved in infection. More than 1,500 miRNAs (1,352 known and 150 novel miRNAs) were detected in the infected and control mice. The expression of miRNAs varied across time after infection; 3, 38, and 108 differentially expressed miRNAs (P < 0.05) were detected during acute infection, chronic infection and chronic vs. acute infection, respectively. GO analysis showed that chronically infected mice had more predicted targets of dysregulated miRNAs than acutely infected mice. KEGG analysis indicated that most predicted targets were involved in immune- or disease-related pathways. Our data indicate that T. gondii infection alters the abundance of miRNAs in mouse brain particularly at the chronic stage, probably to fine-tune conditions required for the establishment of a latent brain infection.
Collapse
Affiliation(s)
- Rui-Si Hu
- 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, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jun-Jun He
- 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, China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, The University of Nottingham, Loughborough, United Kingdom
| | - Fu-Kai Zhang
- 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, China
| | - Yang Zou
- 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, China
| | - Guang-Hui Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Wei Cong
- 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, China.,College of Marine Science, Shandong University at Weihai, Weihai, China
| | - 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, China
| |
Collapse
|
45
|
Saberi R, Sharif M, Sarvi S, Aghayan SA, Hosseini SA, Anvari D, Nayeri Chegeni T, Hosseininejad Z, Daryani A. Is Toxoplasma gondii playing a positive role in multiple sclerosis risk? A systematic review and meta-analysis. J Neuroimmunol 2018; 322:57-62. [DOI: 10.1016/j.jneuroim.2018.06.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 06/01/2018] [Accepted: 06/13/2018] [Indexed: 12/19/2022]
|
46
|
Mhlwatika Z, Aderibigbe BA. Application of Dendrimers for the Treatment of Infectious Diseases. Molecules 2018; 23:E2205. [PMID: 30200314 PMCID: PMC6225509 DOI: 10.3390/molecules23092205] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/14/2018] [Accepted: 06/15/2018] [Indexed: 01/14/2023] Open
Abstract
Dendrimers are drug delivery systems that are characterized by a three-dimensional, star-shaped, branched macromolecular network. They possess ideal properties such as low polydispersity index, biocompatibility and good water solubility. They are made up of the interior and the exterior layers. The exterior layer consists of functional groups that are useful for conjugation of drugs and targeting moieties. The interior layer exhibits improved drug encapsulation efficiency, reduced drug toxicity, and controlled release mechanisms. These unique properties make them useful for drug delivery. Dendrimers have attracted considerable attention as drug delivery system for the treatment of infectious diseases. The treatment of infectious diseases is hampered severely by drug resistance. Several properties of dendrimers such as their ability to overcome drug resistance, toxicity and control the release mechanism of the encapsulated drugs make them ideal systems for the treatment of infectious disease. The aim of this review is to discuss the potentials of dendrimers for the treatment of viral and parasitic infections.
Collapse
Affiliation(s)
- Zandile Mhlwatika
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa.
| | - Blessing Atim Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa.
| |
Collapse
|
47
|
Dunay IR, Diefenbach A. Group 1 innate lymphoid cells in Toxoplasma gondii infection. Parasite Immunol 2018; 40. [PMID: 29315653 DOI: 10.1111/pim.12516] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/04/2018] [Indexed: 12/31/2022]
Abstract
Innate lymphoid cells (ILCs) are a group of lymphocytes that carry out important functions in immunity to infections and in organ homeostasis at epithelial barrier surfaces. ILCs are innate immune cells that provide an early source of cytokines to initiate immune responses against pathogens. Cytotoxic ILCs (i.e. conventional (c)NK cells) and several subsets of helper-like ILCs are the major branches of the ILC family. Conventional NK cells and group 1 ILCs share several characteristics such as surface receptors and the ability to produce IFN-γ upon activation, but they differ in their developmental paths and in their dependence on specific transcription factors. Infection of mice with the intracellular parasite Toxoplasma gondii is followed by a strong Th1-mediated immune response. Previous studies indicate that NK1.1+ cells contribute to the production of IFN-γ and TNF and cytotoxicity during acute T. gondii infection. Upon oral infection, the parasite infects intestinal enterocytes, and within the lamina propria, innate immune responses lead to initial parasite control although the infection disseminates widely and persists long-term in immune privileged sites despite adaptive immunity. Upon parasite entry into the small intestine, during the acute stage, ILC1 produce high levels of IFN-γ and TNF protecting barrier surfaces, thus essentially contributing to early parasite control. We will discuss here the role of innate lymphocytes during T. gondii infection in the context of the only recently appreciated diversity of ILC subsets.
Collapse
Affiliation(s)
- I R Dunay
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - A Diefenbach
- Department of Microbiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
48
|
Alvarado-Esquivel C, Rico-Almochantaf YDR, Hernández-Tinoco J, Quiñones-Canales G, Sánchez-Anguiano LF, Torres-González J, Schott B, Liesenfeld O, Dunay IR. Toxoplasma Gondii Exposure and Neurological Disorders: An Age- and Gender-Matched Case-Control Pilot Study. Eur J Microbiol Immunol (Bp) 2017; 7:303-309. [PMID: 29403659 PMCID: PMC5793700 DOI: 10.1556/1886.2017.00033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 10/09/2017] [Indexed: 01/03/2023] Open
Abstract
Little is known about the association of Toxoplasma gondii infection and neurological disorders. We performed a case-control study with 344 patients with neurological diseases and 344 neurologically healthy age- and gender-matched subjects. Sera of participants were analyzed for anti-T. gondii IgG and IgM antibodies using commercially available immunoassays. Anti-T. gondii IgG antibodies were detected in 25 (7.3%) cases and in 35 (10.2%) controls (odds ratio [OR] = 0.69; 95% confidence interval [CI]: 0.40-1.18; P = 0.17). Anti-T. gondii IgM antibodies were found in 5 (14.3%) of the 25 IgG seropositive cases and in 13 (37.1°%) of the 35 IgG seropositive controls (P = 0.15). Anti-T. gondii IgG antibodies were found in 8 (3.8%) of 213 female cases and in 23 (10.8%) of 213 female controls (OR = 0.32; 95% CI: 0.14-0.73; P = 0.005); and in 17 (13.0%) of 131 male cases and in 12 (9.2%) of 131 male controls (P = 0.32). No direct association between IgG seropositivity and specific neurological disorders was detected. We found no support for a role of latent T. gondii infection in the risk for neurological disorders in this setting. With respect to specific neurological disorders, further studies using larger patient cohorts will be required.
Collapse
Affiliation(s)
- Cosme Alvarado-Esquivel
- Biomedical Research Laboratory, Faculty of Medicine and Nutrition, Juárez University of Durango State, Avenida Universidad S/N, 34000 Durango, Mexico
| | - Yazmin del Rosario Rico-Almochantaf
- Biomedical Research Laboratory, Faculty of Medicine and Nutrition, Juárez University of Durango State, Avenida Universidad S/N, 34000 Durango, Mexico
| | - Jesús Hernández-Tinoco
- Institute for Scientific Research “Dr. Roberto Rivera-Damm,” Juárez University of Durango State, Avenida Universidad S/N, 34000 Durango, Mexico
| | - Gerardo Quiñones-Canales
- Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Predio Canoas S/N, 34000 Durango, Mexico
| | - Luis Francisco Sánchez-Anguiano
- Institute for Scientific Research “Dr. Roberto Rivera-Damm,” Juárez University of Durango State, Avenida Universidad S/N, 34000 Durango, Mexico
| | - Jorge Torres-González
- Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Predio Canoas S/N, 34000 Durango, Mexico
| | - Björn Schott
- Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
- Department of Psychiatry and Psychotherapy, Campus Mitte, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Oliver Liesenfeld
- Institute for Microbiology and Hygiene, Campus Benjamin Franklin, Charité Medical School, Hindenburgdamm 27, D-12203 Berlin, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Otto von Guericke University Magdeburg, Magdeburg D-39120, Leipziger Str. 44, Germany
| |
Collapse
|
49
|
Alvarado-Esquivel C, Rico-Almochantaf YDR, Sanchez-Anguiano LF, Quinones-Canales G, Hernandez-Tinoco J, Torres-Gonzalez J, Gonzalez-Silva MF, Ramirez-Valles EG. Toxoplasma gondii Infection and Headache: A Matched Case-Control Study in a Public Hospital in Durango City, Mexico. J Clin Med Res 2017; 10:27-31. [PMID: 29238431 PMCID: PMC5722042 DOI: 10.14740/jocmr3236w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 10/31/2017] [Indexed: 11/11/2022] Open
Abstract
Background Toxoplasma gondii (T. gondii) can disseminate to brain in infected hosts. Little is known about the magnitude of the association between this infection and headache. Therefore, we sought to determine the association of T. gondii seropositivity and headache in patients attending neurological consultations in a public hospital in Durango City, Mexico. Methods Through an age- and gender-matched case-control study, 105 patients suffering from headache and 105 subjects without headache were examined for anti-T. gondii IgG and IgM antibodies using commercially available enzyme-linked immunoassays. Seropositive cases were analyzed for detection of T. gondii DNA by polymerase chain reaction. Results Anti-T. gondii IgG antibodies were found in five (4.8%) of the 105 cases and in seven (6.7%) of the 105 controls (odds ratio (OR) = 0.70; 95% confidence interval (CI): 0.21 - 2.28; P = 0.76). The frequency of high (> 150 IU/mL) levels of anti-T. gondii IgG antibodies among anti-T. gondii IgG positive individuals was significantly (P = 0.01) higher in cases (5/5) than in controls (1/7). Anti-T. gondii IgM antibodies were found in one (20.0%) of the five IgG seropositive cases, and in three (42.9%) of the seven IgG seropositive controls (P = 0.60). T. gondii DNA was not detected in any of the five anti-T. gondii IgG positive cases. No association between T. gondii infection and specific headache types was found. Conclusions This is the first matched case-control study on the association between T. gondii infection and headache. Results suggest that high anti-T. gondii IgG antibody levels, but not T. gondii seropositivity, were associated with headache in the population studied.
Collapse
Affiliation(s)
- Cosme Alvarado-Esquivel
- Biomedical Research Laboratory, Faculty of Medicine and Nutrition, Juarez University of Durango State, Avenida Universidad S/N, 34000 Durango, Mexico
| | - Yazmin Del Rosario Rico-Almochantaf
- Biomedical Research Laboratory, Faculty of Medicine and Nutrition, Juarez University of Durango State, Avenida Universidad S/N, 34000 Durango, Mexico
| | - Luis Francisco Sanchez-Anguiano
- Institute for Scientific Research "Dr. Roberto Rivera-Damm", Juarez University of Durango State, Avenida Universidad S/N, 34000 Durango, Mexico
| | - Gerardo Quinones-Canales
- Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Predio Canoas S/N, 34000 Durango, Mexico
| | - Jesus Hernandez-Tinoco
- Institute for Scientific Research "Dr. Roberto Rivera-Damm", Juarez University of Durango State, Avenida Universidad S/N, 34000 Durango, Mexico
| | - Jorge Torres-Gonzalez
- Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Predio Canoas S/N, 34000 Durango, Mexico
| | - Maria Felix Gonzalez-Silva
- Faculty of Chemical Sciences, Juarez University of Durango State, Avenida Veterinaria S/N, 34120 Durango, Mexico
| | - Eda Guadalupe Ramirez-Valles
- Faculty of Chemical Sciences, Juarez University of Durango State, Avenida Veterinaria S/N, 34120 Durango, Mexico
| |
Collapse
|
50
|
Toxoplasma gondii infection and schizophrenia: an inter-kingdom communication perspective. Curr Opin Infect Dis 2017; 29:311-8. [PMID: 27120002 DOI: 10.1097/qco.0000000000000265] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The apicomplexan protozoan Toxoplasma gondii has a striking predilection for infecting the central nervous system and has been suggested as a risk factor for schizophrenia. Here, we address some of the mechanisms by which T. gondii achieves this by manipulating signaling pathways of the host brain cells. RECENT FINDINGS Recent years have brought notable progress in the understanding of how the opportunistic parasite T. gondii establishes a successful infection in mammalian brain by secreting effector molecules that manipulate multiple cell functions. Many pathways involved in this inter-kingdom signaling, such as dopaminergic, GABAergic and kynurenine pathways, also have key roles in the development of schizophrenia. More understanding of T. gondii-brain cell interaction holds the key to unlocking the mystery of T. gondii-mediated schizophrenia pathogenesis. SUMMARY T. gondii usurps a variety of host signaling pathways to ensure physiological adaptation, evasion of host immune defense systems, and efficient replication. A detailed knowledge of T. gondii signaling molecules involved in this cross-kingdom communication with host brain cells will probably provide novel means of pharmacologically manipulating host cellular pathways to promote efficient elimination of the parasite and may permit the development of new schizophrenia-modifying therapeutics.
Collapse
|