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 2024; 27:100575. [PMID: 39729771 DOI: 10.1016/j.ijpddr.2024.100575] [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: 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
|
Zhang Y, Li S, Chu H, Li J, Lu S, Zheng B. A novel mRNA vaccine, TGGT1_278620 mRNA-LNP, prolongs the survival time in BALB/c mice with acute toxoplasmosis. Microbiol Spectr 2024; 12:e0286623. [PMID: 38038457 PMCID: PMC10783036 DOI: 10.1128/spectrum.02866-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023] Open
Abstract
IMPORTANCE Toxoplasma gondii, an obligate intracellular eukaryotic parasite, can infect about one-third of the world's population. One vaccine, Toxovax, has been developed and licensed commercially; however, it is only used in the sheep industry to reduce the losses caused by congenital toxoplasmosis. Various other vaccine approaches have been explored, including excretory secretion antigen vaccines, subunit vaccines, epitope vaccines, and DNA vaccines. However, current research has not yet developed a safe and effective vaccine for T. gondii. Here, we generated an mRNA vaccine candidate against T. gondii. We investigated the efficacy of vaccination with a novel identified candidate, TGGT1_278620, in a mouse infection model. We screened T. gondii-derived protective antigens at the genome-wide level, combined them with mRNA-lipid nanoparticle vaccine technology against T. gondii, and investigated immune-related factors and mechanisms. Our findings might contribute to developing vaccines for immunizing humans and animals against T. gondii.
Collapse
Affiliation(s)
- Yizhuo Zhang
- Laboratory of Pathogen Biology, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Shiyu Li
- Laboratory of Pathogen Biology, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Hongkun Chu
- Laboratory of Pathogen Biology, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Jing Li
- Laboratory of Pathogen Biology, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Shaohong Lu
- Laboratory of Pathogen Biology, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Bio-Tech Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Bin Zheng
- Laboratory of Pathogen Biology, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Bio-Tech Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
3
|
Muslimov A, Tereshchenko V, Shevyrev D, Rogova A, Lepik K, Reshetnikov V, Ivanov R. The Dual Role of the Innate Immune System in the Effectiveness of mRNA Therapeutics. Int J Mol Sci 2023; 24:14820. [PMID: 37834268 PMCID: PMC10573212 DOI: 10.3390/ijms241914820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Advances in molecular biology have revolutionized the use of messenger RNA (mRNA) as a therapeutic. The concept of nucleic acid therapy with mRNA originated in 1990 when Wolff et al. reported successful expression of proteins in target organs by direct injection of either plasmid DNA or mRNA. It took decades to bring the transfection efficiency of mRNA closer to that of DNA. The next few decades were dedicated to turning in vitro-transcribed (IVT) mRNA from a promising delivery tool for gene therapy into a full-blown therapeutic modality, which changed the biotech market rapidly. Hundreds of clinical trials are currently underway using mRNA for prophylaxis and therapy of infectious diseases and cancers, in regenerative medicine, and genome editing. The potential of IVT mRNA to induce an innate immune response favors its use for vaccination and immunotherapy. Nonetheless, in non-immunotherapy applications, the intrinsic immunostimulatory activity of mRNA directly hinders the desired therapeutic effect since it can seriously impair the target protein expression. Targeting the same innate immune factors can increase the effectiveness of mRNA therapeutics for some indications and decrease it for others, and vice versa. The review aims to present the innate immunity-related 'barriers' or 'springboards' that may affect the development of immunotherapies and non-immunotherapy applications of mRNA medicines.
Collapse
Affiliation(s)
- Albert Muslimov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Valeriy Tereshchenko
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Daniil Shevyrev
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Anna Rogova
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- Saint-Petersburg Chemical-Pharmaceutical University, Professora Popova 14, 197376 St. Petersburg, Russia
- School of Physics and Engineering, ITMO University, Lomonosova 9, 191002 St. Petersburg, Russia
| | - Kirill Lepik
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Vasiliy Reshetnikov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Roman Ivanov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| |
Collapse
|
4
|
Coixol ameliorates Toxoplasma gondii infection-induced lung injury by interfering with T. gondii HSP70/TLR4/NF-κB signaling pathway. Int Immunopharmacol 2023; 118:110031. [PMID: 36933491 DOI: 10.1016/j.intimp.2023.110031] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023]
Abstract
Toxoplasma gondii (T. gondii) is an obligate intracellular protozoan parasite that causes pulmonary toxoplasmosis, although its pathogenesis is incompletely understood. There is no cure for toxoplasmosis. Coixol, a plant polyphenol extracted from coix seeds, has a variety of biological activities. However, the effects of coixol on T. gondii infection have not been clarified. In this study, we infected a mouse macrophage cell line (RAW 264.7) and BALB/c mice with the T. gondii RH strain to establish infection models in vitro and in vivo, respectively, to explore protective effects and potential mechanisms of coixol on lung injury caused by T. gondii infection. Anti-T. gondii effects and underlying anti-inflammatory mechanisms of coixol were investigated by real-time quantitative PCR, molecular docking, localized surface plasmon resonance, co-immunoprecipitation, enzyme-linked immunosorbent assay, western blotting, and immunofluorescence microscopy. The results show that coixol inhibits T. gondii loads and T. gondii-derived heat shock protein 70 (T.g.HSP70) expression. Moreover, coixol reduced inflammatory cell recruitment and infiltration, and ameliorated pathological lung injury induced by T. gondii infection. Coixol can directly bind T.g.HSP70 or Toll-like receptor 4 (TLR4) to disrupt their interaction. Coixol prevented overexpression of inducible nitric oxide synthase, tumor necrosis factor-α, and high mobility group box 1 by inhibiting activation of the TLR4/nuclear factor (NF)-κB signaling pathway, consistent with effects of the TLR4 inhibitor CLI-095. These results indicate that coixol improves T. gondii infection-induced lung injury by interfering with T.g.HSP70-mediated TLR4/NF-κB signaling. Altogether, these findings suggest that coixol is a promising effective lead compound for the treatment of toxoplasmosis.
Collapse
|
5
|
Lu YN, Shen XY, Lu JM, Jin GN, Lan HW, Xu X, Piao LX. Resveratrol inhibits Toxoplasma gondii-induced lung injury, inflammatory cascade and evidences of its mechanism of action. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154522. [PMID: 36332392 DOI: 10.1016/j.phymed.2022.154522] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 10/03/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Toxoplasma gondii is an opportunistic protozoan that can infect host to cause toxoplasmosis. We have previously reported that resveratrol (RSV) has protective effects against liver damage in T. gondii infected mice. However, the effect of RSV on lung injury caused by T. gondii infection and its mechanism of action remain unclear. PURPOSE In this work, we studied the protective effects of RSV on lung injury caused by T. gondii infection and explored the underlying mechanism. METHODS Molecular docking and localized surface plasmon resonance assay were used to detect the molecular interactions between RSV and target proteins. In vitro, the anti-T. gondii effects and potential anti-inflammatory mechanisms of RSV were investigated by quantitative competitive-PCR, RT-PCR, ELISA, Western blotting and immunofluorescence using RAW 264.7 cells infected with tachyzoites of T. gondii RH strain. In vivo, the effects of RSV on lung injury caused by T. gondii infection were assessed by observing pathological changes and the expression of inflammatory factors of lung. RESULTS RSV inhibited T. gondii loads and T. gondii-derived heat shock protein 70 (T.g.HSP70) expression in RAW 264.7 cells and lung tissues. Moreover, RSV interacts with T.g.HSP70 and toll-like receptor 4 (TLR4), respectively, and interferes with the interaction between T.g.HSP70 and TLR4. It also inhibited the overproduction of inducible nitric oxide synthase, TNF-α and high mobility group protein 1 (HMGB1) by down-regulating TLR4/nuclear factor kappa B (NF-κB) signaling pathway, which is consistent with the effect of TLR4 inhibitor CLI-095. In vivo, RSV improved the pathological lung damage produced by T. gondii infection, as well as decreased the number of inflammatory cells in bronchoalveolar lavage fluid and the release of HMGB1 and TNF-α. CONCLUSION These findings indicate that RSV can inhibit the proliferation of T. gondii and T.g.HSP70 expression both in vitro and in vivo. RSV can inhibit excessive inflammatory response by intervening T.g.HSP70 and HMGB1 mediated TLR4/NF-κB signaling pathway activation, thereby ameliorating lung injury caused by T. gondii infection. The present study provides new data that may be useful for the development of RSV as a new agent for the treatment of lung damage caused by T. gondii infection.
Collapse
Affiliation(s)
- Yu Nan 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, PR. 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, PR. China
| | - 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, PR. 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, PR. China
| | - Hui Wen Lan
- 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, PR. 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, PR. 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, PR. China.
| |
Collapse
|
6
|
Zhang Y, Li D, Shen Y, Li S, Lu S, Zheng B. Immunization with a novel mRNA vaccine, TGGT1_216200 mRNA-LNP, prolongs survival time in BALB/c mice against acute toxoplasmosis. Front Immunol 2023; 14:1161507. [PMID: 37122740 PMCID: PMC10140528 DOI: 10.3389/fimmu.2023.1161507] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Toxoplasma gondii, a specialized intracellular parasite, causes a widespread zoonotic disease and is a severe threat to social and economic development. There is a lack of effective drugs and vaccines against T. gondii infection. Recently, mRNA vaccines have been rapidly developed, and their packaging materials and technologies are well established. In this study, TGGT1_216200 (TG_200), a novel molecule from T. gondii, was identified using bioinformatic screening analysis. TG_200 was purified and encapsulated with a lipid nanoparticle (LNP) to produce the TG_200 mRNA-LNP vaccine. The immune protection provided by the new vaccine and its mechanisms after immunizing BABL/C mice via intramuscular injection were investigated. There was a strong immune response when mice were vaccinated with TG_200 mRNA-LNP. Elevated levels of anti-T. gondii-specific immunoglobulin G (IgG), and a higher IgG2a-to-IgG1 ratio was observed. The levels of interleukin-12 (IL-12), interferon-γ (IFN-γ), IL-4, and IL-10 were also elevated. The result showed that the vaccine induced a mixture of Th1 and Th2 cells, and Th1-dominated humoral immune response. Significantly increased antigen-specific splenocyte proliferation was induced by TG_200 mRNA-LNP immunization. The vaccine could also induce T. gondii-specific cytotoxic T lymphocytes (CTLs). The expression levels of interferon regulatory factor 8 (IRF8), T-Box 21 (T-bet), and nuclear factor kappa B (NF-κB) were significantly elevated after TG_200 mRNA-LNP immunization. The levels of CD83, CD86, MHC-I, MHC-II, CD8, and CD4 molecules were also higher. The results indicated that TG_200 mRNA-LNP produced specific cellular and humoral immune responses. Most importantly, TG_200 mRNA-LNP immunized mice survived significantly longer (19.27 ± 3.438 days) than the control mice, which died within eight days after T. gondii challenge (P< 0.001). The protective effect of adoptive transfer was also assessed, and mice receiving serum and splenocytes from mice immunized with TG_200 mRNA-LNP showed improved survival rates of 9.70 ± 1.64 days and, 13.40 ± 2.32 days, respectively (P< 0.001). The results suggested that TG_200 mRNA-LNP is a safe and promising vaccine against T. gondii infection.
Collapse
Affiliation(s)
- Yizhuo Zhang
- Institute of Parasitic Diseases, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Dan Li
- Institute of Parasitic Diseases, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Yu Shen
- Institute of Parasitic Diseases, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Shiyu Li
- Institute of Parasitic Diseases, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Shaohong Lu
- Institute of Parasitic Diseases, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Bio-tech Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Shaohong Lu, ; Bin Zheng,
| | - Bin Zheng
- Institute of Parasitic Diseases, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Bio-tech Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Shaohong Lu, ; Bin Zheng,
| |
Collapse
|
7
|
Genetic immunization against toxoplasmosis: A review article. Microb Pathog 2021; 155:104888. [PMID: 33930415 DOI: 10.1016/j.micpath.2021.104888] [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: 01/18/2021] [Revised: 04/13/2021] [Accepted: 04/13/2021] [Indexed: 11/21/2022]
Abstract
Toxoplasma gondii is a protozoan coccidian parasite belonging to Phylum Apicomplexa and is the causative agent of toxoplasmosis as a zoonotic disease around the world. It is one of the most important protozoa which is transmitted via various routes and infects several warm-blooded animals. The seroprevalence of T. gondii infection is high worldwide and leads to clinical, psychological, and economic problems. At present, available drug therapy for toxoplasmosis has severe side effects, so the development of new anti-toxoplasma drugs or effective vaccines is mandatory. Therefore, different measures have been taken for the development of anti-toxoplasmosis vaccines, and various studies have shown that DNA vaccines could be one of the most successful approaches against the intracellular parasite, T. gondii. Many of these studies have evaluated the efficacy of immunogenicity and different aspects of the DNA vaccines for toxoplasmosis including single genes or multi-gene plasmids with or without adjuvants. Most of the literature confirms that DNA vaccines containing different antigens of the toxoplasma parasite can induce suitable immune response and protection in acute or chronic toxoplasmosis. Therefore, in this review article, we aimed to discuss the current status of DNA vaccines as a new immunization method against toxoplasmosis.
Collapse
|
8
|
Cheng JH, Xu X, Li YB, Zhao XD, Aosai F, Shi SY, Jin CH, Piao JS, Ma J, Piao HN, Jin XJ, Piao LX. Arctigenin ameliorates depression-like behaviors in Toxoplasma gondii-infected intermediate hosts via the TLR4/NF-κB and TNFR1/NF-κB signaling pathways. Int Immunopharmacol 2020; 82:106302. [PMID: 32086097 DOI: 10.1016/j.intimp.2020.106302] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/27/2020] [Accepted: 02/10/2020] [Indexed: 01/23/2023]
Abstract
Toxoplasma gondii (T. gondii) is a known neurotropic protozoan that remains in the central nervous system and induces neuropsychiatric diseases in intermediate hosts. Arctigenin (AG) is one of the major bioactive lignans of the fruit Arctium lappa L. and has a broad spectrum of pharmacological activities such as neuroprotective, anti-inflammatory and anti-T. gondii effects. However, the effect of AG against depressive behaviors observed in T. gondii-infected hosts has not yet been clarified. In the present study, we analyzed the effects of AG against T. gondii-induced depressive behaviors in intermediate hosts using a microglia cell line (BV2 cells) and brain tissues of BALB/c mice during the acute phase of infection with the RH strain of T. gondii. AG attenuated microglial activation and neuroinflammation via the Toll-like receptor/nuclear factor-kappa B (NF-κB) and tumor necrosis factor receptor 1/NF-κB signaling pathways, followed by up-regulating the dopamine and 5-hydroxytryptamine levels and inhibiting the depression-like behaviors of hosts. AG also significantly decreased the T. gondii burden in mouse brain tissues. In conclusion, we elucidated the effects and underlying molecular mechanisms of AG against depressive behaviors induced by T. gondii infection.
Collapse
Affiliation(s)
- Jia-Hui Cheng
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Xiang Xu
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Ying-Biao Li
- Department of Neurology, Affliated Hospital of Yanbian University, Yanji 133000, Jilin, China
| | - Xu-Dong Zhao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Fumie Aosai
- Department of Infection and Host Defense, Graduate School of Medicine, Shinshu University, Matsumoto, Japan
| | - Su-Yun Shi
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Cheng-Hua Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Jing-Shu Piao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Juan Ma
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Hu-Nan Piao
- Department of Neurology, Affliated Hospital of Yanbian University, Yanji 133000, Jilin, China.
| | - Xue-Jun Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China.
| | - Lian-Xun Piao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China. https://orcid.org/0000-0002-8315-5918
| |
Collapse
|
9
|
Wang J, Feng H, Li Z, Zhang X. Napabucasin prevents brain injury in neuronal neonatal rat cells through suppression of apoptosis and inflammation. Microb Pathog 2019; 128:337-341. [PMID: 30659911 DOI: 10.1016/j.micpath.2019.01.019] [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: 11/29/2018] [Revised: 12/18/2018] [Accepted: 01/16/2019] [Indexed: 11/29/2022]
Abstract
The present study investigates the protective effect of napabucasin on the expression of apoptosis markers and inflammatory factors in the neuronal rat cells with post-isolation damage. The level of ROS determined by the fluorescence measurement in the neuronal rat cells with post-isolation damage was 310.21 RFU compared to 21.45 RFU in sham cell cultures. Napabucasin treatment decreased ROS level in the neuronal rat cells with post-isolation damage in dose based manner. ROS level decreased to 278.67, 203.65, 163.32 and 26.87 RFU, respectively in 1, 2, 3 and 4 μM napabucasin treated cell cultures. Treatment with napabucasin increased GSH level significantly (P < 0.05) in the neuronal rat cells with post-isolation damage. Napabucasin treatment at with 1, 2, 3 and 4 μM concentrations increased SOD activity to 2.4, 3.6, 5.1 and 6.1 U/mg, respectively. Treatment with napabucasin increased the activity of catalase in dose based manner. Napabucasin treatment increased Gpx in injured brain cells of neonatal rats. A significant (P < 0.05) increase in the activity of AChE was observed in neuronal rat cells with post-isolation damage on treatment with napabucasin. Treatment with napabucasin reduced the level of TNF-α and IL-6 significantly (P < 0.05) compared to untreated group. Napabucasin treatment decreased the expression of Bax, caspase-3 and p53 proteins in the neuronal rat cells with post-isolation damage. Napabucasin treatment protects post-isolation damage in the neuronal cells of neonatal rats by suppression of apoptosis and oxidative stress. Therefore, napabucasin can be used for the treatment of brain injury.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pediatrics, Hanzhong Central Hospital, Hanzhong, Shaanxi, 723000, China
| | - Hang Feng
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi 'an, Shaanxi, 710068, China
| | - Zhe Li
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi 'an, Shaanxi, 710068, China
| | - Xiaoge Zhang
- Department of Pediatrics, Northwest Women's and Children's Hospital, Xi 'an, Shaanxi, 710061, China.
| |
Collapse
|
10
|
A systematic review of Toxoplasma gondii antigens to find the best vaccine candidates for immunization. Microb Pathog 2018; 126:172-184. [PMID: 30399440 DOI: 10.1016/j.micpath.2018.11.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/30/2018] [Accepted: 11/02/2018] [Indexed: 11/24/2022]
Abstract
At present, there is not any available accepted vaccine for prevention of Toxoplasma gondii (T. gondii) in human and animals. We conducted literature search through English (Google Scholar, PubMed, Science Direct, Scopus, EBSCO, ISI Web of Science) scientific paper databases to find the best vaccine candidates against toxoplasmosis among T. gondii antigens. Articles with information on infective stage, pathogenicity, immunogenicity and characterization of antigens were selected. We considered that the ideal and significant vaccines should include different antigens and been expressed in all infective stages of the parasite with a high pathogenicity and immunogenicity. Evaluation within this systematic review indicates that MIC 3, 4, 13, ROP 2, RON 5, GRA 1, 6, 8, 14 are expressed in all three infective stages and have pathogenicity and immunogenicity. MIC 5, ROM 4, GRA 2, 4, 15, ROP 5, 16, 17, 38, RON 4, MIC 1, GRA 10, 12, 16, SAG 3 are expressed in only tachyzoites and bradyzoites stages of T. gondii with pathogenicity/immunogenicity. Some antigens appeared to be expressed in a single stage (tachyzoites) but have high pathogenicity and induce immune response. They include enolase2 (ENO2), SAG 1, SAG5D, HSP 70, ROM 1, ROM 5, AMA 1, ROP 18, RON2 and GRA 24. In conclusion, current vaccination against T. gondii infection is not satisfactory, and with the increasing number of high-risk individuals, the development of an effective and safe specific vaccine is greatly valuable for toxoplasmosis prevention. This systematic review reveals prepare candidates for immunization studies.
Collapse
|
11
|
Foroutan M, Ghaffarifar F. Calcium-dependent protein kinases are potential targets for Toxoplasma gondii vaccine. Clin Exp Vaccine Res 2018; 7:24-36. [PMID: 29399577 PMCID: PMC5795042 DOI: 10.7774/cevr.2018.7.1.24] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/03/2018] [Accepted: 01/06/2018] [Indexed: 01/30/2023] Open
Abstract
Toxoplasma gondii belongs to the Apicomplexa phylum that caused a widespread zoonotic infection in wide range of intermediate hosts. Over one-third of the world's population are latently infected with T. gondii and carry it. The complex life cycle of T. gondii indicates the presence of a plurality of antigenic epitopes. During the recent years, continuous efforts of scientists have made precious advances to elucidate the different aspects of the cell and molecular biology of T. gondii. Despite of great progresses, the development of vaccine candidates for preventing of T. gondii infection in men and animals is still remains a challenge. The calcium-dependent protein kinases (CDPKs) belongs to the superfamily of kinases, which restricted to the apicomplexans, ciliates, and plants. It has been documented that they contribute several functions in the life cycle of T. gondii such as gliding motility, cell invasion, and egress as well as some other critical developmental processes. In current paper, we reviewed the recent progress concerning the development of CDPK-based vaccines against acute and chronic T. gondii.
Collapse
Affiliation(s)
- Masoud Foroutan
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Ghaffarifar
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
12
|
Lopez-Romero G, Garzon T, Rascon R, Valdez A, Quintero J, Arvizu-Flores AA, Garibay-Escobar A, Rascon L, Astiazarán-García H, Velazquez C. Characterization of BIP protein of G. lamblia as a potential immunogen in a mouse infection model. Immunobiology 2017; 222:884-891. [PMID: 28552268 DOI: 10.1016/j.imbio.2017.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/25/2017] [Accepted: 05/14/2017] [Indexed: 12/11/2022]
Abstract
Giardia lamblia is a protozoan parasite that causes one of the most common gastrointestinal diseases worldwide. To eliminate the parasite from the host intestine, it is necessary the activation of B-cell and T-cell dependent mechanisms. The knowledge about Giardia antigens that can stimulate the host immune response is limited. Recently, it has been described the Binding Immunoglobulin Protein (BIP) of G. lamblia (71kDa) as a potential immunogen. Additionally, our group has identified a highly immunogenic antigen (5G8 protein) of G. lamblia with a relative molecular mass of approximately 70kDa. There is some evidence suggesting that the 5G8 protein may activate both humoral and cellular immune responses. Based on these observations and preliminary mass spectrometry analyses, we hypothesized that the antigen 5G8 could be the BIP protein. In the present study, we characterize immunochemically the BIP protein of Giardia. Flow cytometric assays and western blotting were used to determine the expression profile of BIP and 5G8 antigens in Giardia trophozoites. The differences in expression profile indicated that BIP and 5G8 are not the same molecule. ELISA and Western blotting assays revealed that BIP protein was recognized by antibodies produced during G. lamblia infection in C3H/HeN mice. MTT assays did not reveal the activation of cellular immune response induced by BIP protein in vitro. In addition, we identified the potential B-cell and T-cell epitopes of G. lamblia BIP protein. This molecule is a conserved protein among Giardia strains and other pathogens. The complete immunological characterization of this antigen will contribute to a better understanding of the host-parasite interactions in Giardia infection.
Collapse
Affiliation(s)
- Gloria Lopez-Romero
- Coordinación de Nutrición, Centro de Investigación en Alimentación y Desarrollo A.C. Hermosillo, Sonora, Mexico
| | - Thania Garzon
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Sonora, Mexico
| | - Raul Rascon
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Sonora, Mexico
| | - Alejandra Valdez
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Sonora, Mexico
| | - Jael Quintero
- Health Science Department, University of Sonora, Blvd Bordo Nuevo s/n, Ejido Providencia, 85199 Cd. Obregon, Sonora, Mexico
| | - Aldo A Arvizu-Flores
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Sonora, Mexico
| | | | - Lucila Rascon
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Sonora, Mexico
| | - Humberto Astiazarán-García
- Coordinación de Nutrición, Centro de Investigación en Alimentación y Desarrollo A.C. Hermosillo, Sonora, Mexico
| | - Carlos Velazquez
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Sonora, Mexico.
| |
Collapse
|
13
|
Czarnewski P, Araújo ECB, Oliveira MC, Mineo TWP, Silva NM. Recombinant TgHSP70 Immunization Protects against Toxoplasma gondii Brain Cyst Formation by Enhancing Inducible Nitric Oxide Expression. Front Cell Infect Microbiol 2017; 7:142. [PMID: 28487847 PMCID: PMC5403831 DOI: 10.3389/fcimb.2017.00142] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/06/2017] [Indexed: 11/17/2022] Open
Abstract
Toxoplasma gondii is known to cause congenital infection in humans and animals and severe disease in immunocompromised individuals; consequently development of vaccines against the parasite is highly necessary. Under stress conditions, T. gondii expresses the highly immunogenic heat shock protein 70 (TgHSP70). Here, we assessed the protective efficacy of rTgHSP70 immunization combined with Alum in oral ME-49 T. gondii infection and the mechanisms involved on it. It was observed that immunized mice with rTgHSP70 or rTgHSP70 adsorbed in Alum presented a significantly reduced number of cysts in the brain that was associated with increased iNOS+ cell numbers in the organ, irrespective the use of the adjuvant. Indeed, ex vivo experiments showed that peritoneal macrophages pre-stimulated with rTgHSP70 presented increased NO production and enhanced parasite killing, and the protein was able to directly stimulate B cells toward antibody producing profile. In addition, rTgHSP70 immunization leads to high specific antibody titters systemically and a mixed IgG1/IgG2a response, with predominance of IgG1 production. Nonetheless, it was observed that the pretreatment of the parasite with rTgHSP70 immune sera was not able to control T. gondii internalization and replication by NIH fibroblast neither peritoneal murine macrophages, nor anti-rTgHSP70 antibodies were able to kill T. gondii by complement-mediated lysis, suggesting that these mechanisms are not crucial to resistance. Interestingly, when in combination with Alum, rTgHSP70 immunization was able to reduce inflammation in the brain of infected mice and in parallel anti-rTgHSP70 immune complexes in the serum. In conclusion, immunization with rTgHSP70 induces massive amounts of iNOS expression and reduced brain parasitism, suggesting that iNOS expression and consequently NO production in the brain is a protective mechanism induced by TgHSP70 immunization, therefore rTgHSP70 can be a good candidate for vaccine development against toxoplasmosis.
Collapse
Affiliation(s)
- Paulo Czarnewski
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of UberlândiaUberlândia, Brazil
| | - Ester C B Araújo
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of UberlândiaUberlândia, Brazil
| | - Mário C Oliveira
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of UberlândiaUberlândia, Brazil
| | - Tiago W P Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Federal University of UberlândiaUberlândia, Brazil
| | - Neide M Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of UberlândiaUberlândia, Brazil
| |
Collapse
|
14
|
Abstract
In the two decades since their initial discovery, DNA vaccines technologies have come a long way. Unfortunately, when applied to human subjects inadequate immunogenicity is still the biggest challenge for practical DNA vaccine use. Many different strategies have been tested in preclinical models to address this problem, including novel plasmid vectors and codon optimization to enhance antigen expression, new gene transfection systems or electroporation to increase delivery efficiency, protein or live virus vector boosting regimens to maximise immune stimulation, and formulation of DNA vaccines with traditional or molecular adjuvants. Better understanding of the mechanisms of action of DNA vaccines has also enabled better use of the intrinsic host response to DNA to improve vaccine immunogenicity. This review summarizes recent advances in DNA vaccine technologies and related intracellular events and how these might impact on future directions of DNA vaccine development.
Collapse
Affiliation(s)
- Lei Li
- a Vaxine Pty Ltd, Bedford Park , Adelaide , Australia.,b Department of Diabetes and Endocrinology , Flinders University, Flinders Medical Centre , Adelaide , SA , Australia
| | - Nikolai Petrovsky
- a Vaxine Pty Ltd, Bedford Park , Adelaide , Australia.,b Department of Diabetes and Endocrinology , Flinders University, Flinders Medical Centre , Adelaide , SA , Australia
| |
Collapse
|
15
|
Zhang NZ, Wang M, Xu Y, Petersen E, Zhu XQ. Recent advances in developing vaccines against Toxoplasma gondii: an update. Expert Rev Vaccines 2015; 14:1609-21. [PMID: 26467840 DOI: 10.1586/14760584.2015.1098539] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Toxoplasma gondii, a significant public health risk, is able to infect almost all warm-blooded animals including humans, and it results in economic losses in production animals. In the last three years, a large number of vaccination experiments have been performed to control T. gondii infection, with the target of limiting the acute infection and reducing or eliminating tissue cysts in the intermediate hosts. In this paper, we summarize the latest results of the veterinary vaccines against T. gondii infection since 2013. Immunization with live-attenuated whole organisms of non-reverting mutants has been shown to induce remarkably potent immune responses associated with control of acute and chronic toxoplasmosis. The non-cyst-forming mutants are promising new tools for the development of veterinary vaccines against T. gondii infection.
Collapse
Affiliation(s)
- Nian-Zhang Zhang
- a 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 , PR China
| | - Meng Wang
- a 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 , PR China
| | - Ying Xu
- a 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 , PR China.,b Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Protozoa Laboratory, College of Veterinary Medicine , China Agricultural University , Beijing , PR China
| | - Eskild Petersen
- c Department of Infectious Diseases, Clinical Institute, Faculty of Health Sciences , Aarhus University , Aarhus , Denmark
| | - Xing-Quan Zhu
- a 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 , PR China
| |
Collapse
|