Open Access
Issue
Parasite
Volume 28, 2021
Article Number 83
Number of page(s) 9
DOI https://doi.org/10.1051/parasite/2021082
Published online 15 December 2021

© Y. Xu et al., published by EDP Sciences, 2021

Licence Creative CommonsThis is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Introduction

As the causative agent of toxoplasmosis, Toxoplasma gondii can infect all warm-blooded animals, affecting approximately 30% of the world’s population [12, 25]. Primary infection in immune-potent individuals is usually asymptomatic or presents as a mild, flu-like illness, concomitant with parasite conversion to dormant bradyzoites within a tissue cyst [20]. However, severe toxoplasmosis in immunocompromised individuals can develop following reactivation of bradyzoites into disseminating tachyzoites, leading to toxoplasmic encephalitis (TE), eye disease, neurological problems and even death [12, 20]. Congenital infection can occur in newborns resulting from the infection of pregnant women, and is associated with dysplasia, hydrocephaly and chorioretinitis [11]. Toxoplasma gondii infection can also cause considerable economic losses to the stock-raising industry [12]. Although acute T. gondii infections can be controlled by medications, there are still no effective drugs that can be used to completely eliminate chronic infection, and more effective therapeutic agents are needed for this organism [9]. Additionally, no approved clinical vaccines are available for human infections [32]. Determining the immune response to T. gondii is vital for the design of effective vaccines and also to find drug targets.

It is well documented that the immune response induced by T. gondii infection is complex, including innate and adaptive immune response, which involves various immune cells, such as CD4+ and CD8+ T cells, natural killer (NK) cells, dendritic cells (DCs), macrophages and neutrophils [22, 36]. CD8+ T cells and their responses are essential for the control of infection by acting synergistically with CD4+ T cells [10]. Primarily, T helper 1 (Th-1) cell-mediated protective immunity to T. gondii drives the production of high levels of interleukin-12 (IL-12) and interferon-γ (IFN-γ) followed by the migration of innate immune cells (e.g., DCs, macrophages and neutrophils) to the site of infection, which are indispensable for host resistance against T. gondii, and limiting the parasite’s proliferation and the progression of infection via multiple intracellular mechanisms with the production of various antiparasitic factors [13, 23]. The expansion of Th-2 cells and anti-inflammatory cytokine production of IL-4, IL-10, IL-13, IL-27, and transforming growth factor-β (TGF-β) are responsible for controlling the effects of excessive immune activation and preventing immunologic pathology [23]. Collectively, these innate and adaptive immune responses, mainly Th-1 and Th-2-associated cytokines contribute to protective immunity against T. gondii infection.

MicroRNAs (miRNAs), as class of non-coding RNAs, are involved in gene regulation at both transcriptional and post-transcriptional levels [2]. Through several mechanisms, including translational repression and messenger RNA (mRNA) degradation, miRNAs have been shown to mediate and regulate some physiological processes, and also to be related to human disease, such as carcinogenesis, and even extended to the immune system [18, 19]. It has been revealed that miRNAs can regulate immune system biology, including both lymphocyte development and function and host immune responses, and also they have been shown to be involved in inflammation associated with CD4+ T cell differentiation and CD8+ T cell responses [19]. In particularly, miRNA-155 is a well-characterized miRNA known to be a key regulator of monocytes, macrophages, T-cells and B-cells by interference in the expression of pro-inflammatory and anti-inflammatory cytokines [1, 31]. miRNA-155 has a regulatory role in visceral leishmaniasis by up-regulating both Th-1 and Th-2 immune responses, which are together to be contributed to the control of the infection [30]. miRNA-155 has been demonstrated to be an important immune regulatory molecule for the control of Trypanosoma cruzi infection [16]. Likewise, miR-155 is known to play a critical role in maintaining the survival of Mtb-infected macrophages and the function of Mtb-specific T-cells during Mycobacterium infection [21].

Despite the fact that miR-155 is elevated in chronic T. gondii infection [34], its specific role in regulating innate and adaptive immune responses against this infection has not been elucidated. To determine whether miRNA-155 influences the progression of T. gondii infection and immune responses in mice models, we investigated the role of miRNA-155 during immunity to T. gondii infection in miR-155 gene-deficient mice and those of their age- and sex-matched wild-type (WT) counterparts. Our results show that miR-155 is required for control of T. gondii infection via mediating a wide spectrum of immune compartments.

Materials and methods

Ethics

All mice were maintained and bred in strict accordance with the Animal Ethics Procedures and Guidelines of the People’s Republic of China. Animal experiments were approved by the ethics committee of Ningbo University (permission: SYXK(ZHE)2019-0005).

Mice and parasites

Age-matched 6–8 week old female miR-155KO C57BL/6 and WT C57BL/6 mice were purchased from Beijing Vital River Laboratory Animal Technology Co., China.

The PRU strain (Type II) of T. gondii was used for the in vivo challenge of mice, which was propagated and harvested as described in studies [35]. The tachyzoite forming of the PRU strain (Type II) of T. gondii were also used for preparation of soluble tachyzoite antigens (TLA), as previously described [33, 35].

Cytokine ELISA

According to previously described methods [33, 35], splenocytes were prepared by pushing the spleens through a wire mesh, and then purified by removing red blood cells using RBC erythrocyte lysis buffer, and then re-suspended in DMEM medium supplemented with 10% FCS, 1% penicillin – streptomycin, and 1% HEPES. The test specimen was then plated at a concentration of 5 × 106 cells/mL and stimulated for 72 h with 15 μg/mL TLA. Cell-free supernatants were collected and assayed for IL-2 and IL-4 at 24 h, for IL-22 activity at 48 h, for IL-13, IL-17A, IL-17F activity at 72 h, and for IFN-γ activity at 96 h, using commercial ELISA kits according to the manufacturer’s instructions (Biolegend, USA).

Isolation of mouse spleen lymphocytes

After reaching the experimental detection conditions, the mice were sacrificed and spleens were taken out. Red blood cell lysate was used to lyse red blood cells, and 40% and 70% percoll solutions were prepared with percoll stock solution, and lymphocytes were separated by density gradient centrifugation.

Flow cytometry

Single-cell suspensions were prepared according to the method mentioned above. Followed by blocking of FC receptors with addition of normal mouse serum, cells were stained with the following antibodies: CD3 (BV421, clone: 145-2C11, BD Biosciences), CD4 (BV510, clone: RM4-5, BD Biosciences), CD8 (PE, clone: 53-6.7, BD Biosciences), NK1.1 (PE-CY7, clone: PK136, BD Biosciences), CD11b (BV605, clone: M1/70, BD Biosciences), Ly6C (APC, clone: 560595, BD Biosciences). For intracellular cytokine staining (ICS) assays, prior to intracellular stained with IFN- and AF647 (clone: XMG1.2, BD Biosciences) for 30 min at 4° C, the cells were surface-stained CD8-PE (BD Biosciences), fixed and permeabilized with Cytofix/Cytoperm solution (BD Biosciences) for 20 min in dark, and data were then acquired by performing these cells on a Beckman CytoFLEX S and data were further analyzed by FlowJo software (Tree Star, Inc., Ashland, OR, USA).

qRT-PCR

The expression of IL-1α, IL-1 β, IL-6, SHIP-1 and SOCS1 were analysed by qRT-PCR. Total RNA was isolated from three purified splenocytes of mice in each group using Trizol reagent (Invitrogen, USA), as per the manufacturer’s instructions. RNAs were dissolved in RNase-free ddH2O (TaKaRa, China) and cDNA was synthesized using a GoScript™ Reverse Transcription System (Promega, Madison, WI, USA) and used as a template for quantitative real-time polymerase chain reaction (qRT-PCR). qRT-PCR was performed using a Light Cycler 480 SYBR Green I Master (Roche, Switzerland). The primers used for amplification are listed in Table 1. qRT-PCR analysis was performed on the Light Cycler 480 (Roche, Switzerland) and data were calculated using the comparative cycle threshold (CT) method (2−ΔCT).

Table 1

Primer sequences.

Challenge and parasite burdens

A total of five mice in each group were challenged with 10 PRU tissue cysts of T. gondii PRU strain orally, and the survival periods were recorded daily until all mice were dead. In parallel, mean brain cyst loadings were counted at 14 days after the challenge, as described in our previous studies [33, 35]. All samples were counted in triplicate. The parasite reduction rate of brain cysts is relative to that of the control.

Statistical analysis

Statistical analysis was conducted using GraphPad Prism 8. Student’s unpaired t test was used to determine statistical significance of differences among the groups. p-values < 0.05 were considered statistically significant.

Results

miR-155 deficiency enhanced the susceptibility to T. gondii

miR-155 was recently found to contribute to resistance to the experimental parasitic diseases [16, 30]. To test whether the lack of miR-155 would potentiate chronic T. gondii in mice models, miR-155−/− mice were orally inoculated with 10 PRU tissue cysts. All of the miR-155−/− infected mice died after 21 days of infection, but WT mice infected with T. gondii showed a prolonged survival time (Fig. 1A). To further confirm the T. gondii infection in WT and miR-155−/− mice, the mean brain cyst loadings were counted. Toxoplasma gondii-infected miR-155−/− mice showed more brain cysts (Fig. 1B). These data demonstrate that the absence of miR-155 enhances susceptibility to T. gondii in mice models.

thumbnail Figure 1

miR-155 deficiency enhanced cyst burden and reduced survival time in mice models. (A) Survival curves of mice after challenge of cysts of the PRU strain. miR-155−/− mice had 0% survival at day 17. (B) Cyst loads were counted from whole brain homogenates of mice 4 weeks after challenge. The bars represented the mean cyst burden per mouse brain after challenge orally with a dose of 10 cysts of the PRU strain. Data are mean ± SD (representative of three experiments). **p < 0.01; *p < 0.05, compared with the WT groups.

miR-155 deficiency impairs both Th-1 and Th-2 immune responses in the spleens of T. gondii-infected mice

As miR-155 has been shown to be critical for regulating T-cell responses as well as inflammatory responses and cytokine signals, which have well-documented roles in the control of intracellular pathogens, including T. cruzi and Leishmania donovani [16, 30], we further analysed the production of cytokines Th-1-associated IFN-γ, IL-2, Th-2-associated IL-4 and IL-13, Th-17-associated IL-17A and IL-17F by splenic cells harvested from WT and miR-155KO (miR-155−/−) mice after infection with T. gondii PRU cysts.

After ELISA analysis of TLA-stimulated spleen cells supernatants, it was shown that the production of IFN-γ and IL-2 was significantly reduced in T. gondii-infected miR-155−/− mice compared to WT controls (Fig. 2). Likewise, IL-13 production by splenic cells from T. gondii-infected miR-155−/− mice was dramatically decreased in contrast to WT controls (Fig. 2). However, there were no significant changes in the levels of IL-4, IL-17A and IL-17F in these T. gondii-infected miR-155−/− mice and WT controls (Fig. 2). Taken together, these data suggest that this T. gondii infection in miR-155−/− is due to lower protective cytokine production.

thumbnail Figure 2

Evaluation of Th-1\Th-2\Th-17 immune responses in WT and miR-155−/− mice. The levels of IFN-γ, IL-2, IL-13, IL-4, IL-17A and IL-17F were measured from splenic lymphocytes of WT- and miR-155KO-infected mice. Significant decreases in IFN-γ, IL-2, IL-13 were observed in miR-155−/− mice. Data are shown from one representative experiment of three independent experiments and presented as a mean ± SD. ***p < 0.001; N/S = not significant.

Lack of miR-155 contributes to impaired recruitment of CD4+ T, CD8+ T, NK and NK-T cells in the spleens of T. gondii-infected mice

NK cells, NK-T, and T cells are usually recruited to kill T. gondii by hosts through the production of IFN-γ and a perforin-independent mechanism [13, 23]. To investigate whether miR-155 deficiency has an effect in recruitment of these immune cells, the splenic cell populations of CD4+ T cells, CD8+ T cells, NK cells and NK-T cells were analysed from T. gondii-infected miR-155−/− and WT mice by flow cytometry. We found that these splenic cell populations of T. gondii-infected miR-155−/− mice were significantly decreased in contrast to WT counterparts (Fig. 3). These results suggest that miR-155 plays a role in mediating recruitment of NK, NK-T cells, CD4+ T cells and CD8+ T cells, which control T. gondii infection, while these decreased recruited cells in miR-155−/− mice could not activate protective immunity effectively, leading to subsequent aggravated parasitic infection and even death.

thumbnail Figure 3

miR-155 deficiency impairs the recruitment of CD4+ T cells, CD8+ T cells, NK and NKT cell. Spleens were collected from each mouse in the WT and miR-155−/− groups. The splenocytes labeled with cell-specific fluorescent labeled antibodies were analysed by flow cytometry. Results were calculated and expressed as a mean ± SD of three independent experiments. ***p < 0.001; **p < 0.01.

miR-155 deficiency exacerbated CD8+ T cell exhaustion

miR-155 has recently been found to restrain CD8+ T cell functional exhaustion in chronic virus infection and tumors, by targeting several inhibitors of cytokine signaling, including SOCS1 and SHIP1 [8, 24]. Thus, to better understand the role of miR-155 on CD8+ T cell exhaustion during chronic infection with T. gondii, we analysed the expression of IFN-γ, TFN-α and PD-1 in CD8+ T cells and the number of regulatory cells (Tregs) by flow cytometry (Fig. 4). As we expected, in miR-155KO mice, there were significantly decreased numbers of IFN-γ CD8+ and TNF-α CD8+ T cells in contrast to WT mice, suggesting decreased effector T cells in miR-155KO mice (Fig. 4). Similarly, the number of Tregs were also up-regulated in T. gondii-infected miR-155KO mice, suggesting enhanced immune-inhibitory effects followed by the deficient expression of miR-155.

thumbnail Figure 4

miR-155 deficiency exacerbated CD8+ T cell exhaustion by targeting to SHIP-1 and SOCS1. Spleens were collected from each mouse in the WT and miR-155−/− groups. The percentages of Tregs, IFN-γ CD8+ T, TNF-α CD8+ T and PD1 CD8+ T cells were analysed by flow cytometric analysis. The expressions of SHIP-1 and SOCS1 were estimated by qRT-PCR. Data are shown from one representative experiment of three independent experiments and presented as a mean ± SD. ***p < 0.001; **p < 0.01.

It is known that PD-1 expression by the myeloid cell population plays an important role in immune regulation in various infectious diseases as well as in several cancer models [15, 17, 29]. Since PD-1 has been implicated in suppressing T cell response in T. gondii [4], flow cytometric analysis of the expression PD-1 in CD8+ T cell revealed that T. gondii-infected miR-155KO mice up-regulated the expression PD-1 in CD8+ T cell in contrast to T. gondii-infected WT mice. Furthermore, miR-155KO mice showed increased expression of both SHIP-1 and SOCS1 by qRT-PCR (Fig. 4), which are known to be direct targets of miR-155 [8, 24]. Together, these findings indicate that miR-155 deficiency contribute to increased T cell exhaustion by suppressing T cell responses by targeting SHIP-1 and SOCS1.

miR-155 deficiency decreased accumulation of splenic inflammatory monocytes and DCs and expression of pro-inflammatory mediators

Recent studies have established the critical role of CD11b+Ly6C+ cells and DCs in T. gondii infection [5, 26]. Flow cytometric analysis of phagocyte populations has revealed that T. gondii-infected miR-155−/− mice contained significantly lower numbers of CD11b+Ly6C+ cells and DCs than their WT counterparts (Fig. 5). We further determined whether decreased numbers of CD11b+Ly6C+ cells and DCs in miR-155-KO mice consequently impaired the expression of pro-inflammatory mediators by flow cytometric analysis. Consistent with decreased numbers of CD11b+Ly6C+ cells and DCs, the expression of IL-1α, IL-1β and IL-6 was significantly reduced in contrast to their T. gondii-infected WT counterparts (Fig. 5). These data suggest that miR-155 deficiency leads to decreased accumulation of DCs and Ly6C+ inflammatory monocytes, in combination with reduced production of pro-inflammatory mediators that could contribute to high parasitic burdens in miR-155KO mice.

thumbnail Figure 5

Decreased accumulation of splenic inflammatory monocytes and DCs and expression of pro-inflammatory mediators in miR-155KO-infected mice. Flow cytometric analysis of splenic inflammatory monocytes and DCs of parasite infected-WT and miR-155−/− mice. The expression of pro-inflammatory mediators was analysed by qRT-PCR. Data are presented as a mean ± SD of three independent experiments. The asterisks indicate the statistical significance of difference between WT and miR-155−/− mice. ***p < 0.001; **p < 0.01.

Discussion

It is well known that both IFN-γ and IL-4-associated signaling pathways are desired immunological responses to defend against T. gondii infection [22]. In particular, releasing of Th-1 type cytokines, IL-12, IL-2 and IFN-γ are essential for the development of T cell immunity against T. gondii infection [13]. Likewise, T helper type 2 (Th-2) cell-associated cytokines, such as IL-4, IL-10 and IL-13, also play an important role in coordinating the immune response by dampening systemic Th-1 type cytokine production and thus prevent lethal immunopathology [10]. Th-17-associated immune responses are also known to provide significant protective immunity against T. gondii infection [14]. Although miR-155 has been shown to be able to regulate the development and activity of Th-1, Th-12, and CD8+ T cells, its specific role in immunity against T. gondii infection has yet to be well studied. In our study, we established that miR-155 contributes to host immunity against T. gondii infection through the regulation of Th-1 and Th-2 immune responses, but it showed no any effect in Th-17-associated immune responses.

Some recent studies have shown the important role of microRNAs during infection [6, 8, 24]. In our study, it was found that a lack of miR-155 led to a significant increase in parasitic brain cysts and decreases in survival times. Since miR-155 has been shown to be the key regulator of IFN-γ production via the targeting of SOCS1 [3], it is not surprising that this regulative effect should be ascribed to the inability of miR-155−/− mice to mount an efficient Th-1 immune response, with the reduced production of IFN-γ and IL-2. Additionally, we observed a decrease in Th2 immune responses in miR-155−/− mice, which is similar to a previous study in vitro in L. donovani-infected miR-155−/− mice [16], indicating that miR-155 is potentially beneficial for the generation of optimal Th-2 immune responses against T. gondii infection. These data demonstrate that miR-155 plays a significant role in the regulation of both Th-1 and Th-2 immune responses during T. gondii infection.

miR-155 regulates the activation of several immune subpopulations including CD8+T cells, as well as NK and NKT cells [27, 28], which are critical for protective immunity against T. gondii infection [14]. Previous studies have found that miR-155 is important in the control of Leishmania donovani and T. cruzi infection by mediating the regulation of T-cell proliferation and thus the activation of CD8+ T cells, NK and NKT cells [16, 30]. In support of these findings, our data indicate that decreased recruitment of CD8+ T cells, NK and NKT cells resulting from the absence of miR-155 expression, can lead to the lack of control of parasite infection in the miR-155−/− mice.

As the first line cell types in the initial stages of infection, inflammatory monocytes, especially Ly6C+ monocytes are considered to be necessary to govern the control of chronic infection with T. gondii in mice [5], through the inductive production of proinflammatory mediators, such as IL-1α, IL-1β, IL-6, inducible NO synthase, TNF, and reactive oxygen intermediate. Additionally, DCs are essential for the expansion and subsequent T cell priming and activation of Ag-specific CD8+ T cells during infection with T. gondii [26]. Our studies have shown decreased accumulation of Ly6C+ inflammatory monocytes and DCs in miR-155KO mice, associated with a significantly increased parasite load, together with the reduced expression of these proinflammatory mediators. However, it is contradictory that aberrant miR-155 expression has recently been shown to adversely affect Ly6C+ inflammatory monocytes migration in L. donovani and T. cruzi, which is due to a fact that inflammatory monocytes facilitate the growth of the parasites in the spleen in visceral leishmaniasis and trypanosomosis [16, 30]. These results demonstrate that miR-155 deficiency exacerbated T. gondii infection through the down-regulation of DCs and inflammatory monocyte infiltration.

Recent studies showed that chronic infection with T. gondii led to CD8+ T cell exhaustion, concomitant with decreased CD8+ T cell effector response, which is characterized as up-regulated expression of inhibitory receptor PD-1 on these CD8+ T cells [4]. miRNA-155 expression is essential for optimal CD8+ T cell responses toward chronic infection with LCMV, and cancer, involved in regulation of CD8+ T cell exhaustion [8, 24]. In this study, our analysis of the effects of miR-155 on cellular immune responses showed that the deficiency of miR-155 ablated CD8 T cell responses during chronic T. gondii infection, along with up-regulation of PD-1 and down-regulation of IFN-γ and TNF-α in CD8+ T cells. In the meanwhile, Tregs were also augmented in miR-155−/− mice, which have been shown to act as immune-counter in cancer [7], indicating a possible immune-inhibitor effect in the infection with T. gondii, but its specific role needs further confirmation. As the direct target of miR-155, SHIP-1 and SOCS1 are indicated as negative regulators of IFN-γ production and T cell exhaustion [8, 24]. The higher SHIP-1 and SOCS1 levels in infected miR-155KO mice, together with the disrupted responses of CD8+ T cells suggest that T cell exhaustion during chronic T. gondii infection is mediated by miR-155-dependent mechanisms.

Conclusions

We have demonstrated that the absence of miR-155 could enhance susceptibility to chronic T. gondii infection in mice models. It is clear that this effect of miR-155 is ascribed to its contribution to host immunity via the regulation of both Th-1 and Th-2 protective immune responses, as well as inflammatory monocyte and DC infiltration. It should be noted that miR-155 was essential to mediate T cell exhaustion in this model. Therefore, miR-155 can be exploited as a potential target for reinforcing host immunity against T. gondii.

Funding information

Public Welfare Science and Technology Plan Project of Ningbo (202002N3153), Ningbo medical and health brand discipline (PPXK2018-06), Children’s Health and Disease Clinical Medicine Research Center of Ningbo (2019A21002).

Conflict of interest

The authors declare that they have no conflict of interest.

Acknowledgments

The project was supported by the Public Welfare Science and Technology Plan Project of Ningbo (202002N3153), Ningbo medical and health brand discipline (PPXK2018-06), Children’s Health and Disease Clinical Medicine Research Center of Ningbo (2019A21002). Thanks are due to Associate Professor Jia Chen (Ningbo University School of Medicine, Ninbgo, Zhejiang, China) for providing the strain and guidance on the experimental program and technology.

References

  1. Alivernini S, Gremese E, McSharry C, Tolusso B, Ferraccioli G, McInnes IB, Stolarska MK. 2018. MicroRNA-155-at the critical interface of innate and adaptive immunity in arthritis. Frontiers in Immunology, 8, 1932. [CrossRef] [PubMed] [Google Scholar]
  2. Bartel DP. 2009. MicroRNAs: target recognition and regulatory functions. Cell, 136, 215–233. [CrossRef] [PubMed] [Google Scholar]
  3. Baumjohann D, Ansel KM. 2013. MicroRNA-mediated regulation of T helper cell differentiation and plasticity. Nature Reviews Immunology, 13, 666–678. [CrossRef] [PubMed] [Google Scholar]
  4. Bhadra R, Gigley JP, Weiss LM, Khan IA. 2011. Control of Toxoplasma reactivation by rescue of dysfunctional CD8+ T-cell response via PD-1/PDL-1 blockade. Proceedings of the National Academy of Sciences of the United States of America, 108, 9196–9201. [CrossRef] [PubMed] [Google Scholar]
  5. Biswas A, Bruder D, Wolf SA, Jeron A, Mack M, Heimesaat MM, Dunay IR. 2015. Ly6C(high) monocytes control cerebral toxoplasmosis. Journal of Immunology, 194, 3223–3235. [CrossRef] [PubMed] [Google Scholar]
  6. Cannella D, Brenier-Pinchart MP, Braun L, van Rooyen JM, Bougdour A, Bastien O, Behnke MS, Curt RS, Curt A, Saeij JP, Sibley LD, Pelloux H, Hakimi MA. 2014. miR-146a and miR-155 delineate a MicroRNA fingerprint associated with Toxoplasma persistence in the host brain. Cell Reports, 6, 928–937. [CrossRef] [PubMed] [Google Scholar]
  7. Dees S, Ganesan R, Singh S, Grewal IS. 2021. Regulatory T cell targeting in cancer: Emerging strategies in immunotherapy. European Journal of Immunology, 51, 280–291. [CrossRef] [PubMed] [Google Scholar]
  8. Dudda JC, Salaun B, Ji Y, Palmer DC, Monnot GC, Merck E, Boudousquie C, Utzschneider DT, Escobar TM, Perret R, Muljo SA, Hebeisen M, Rufer N, Zehn D, Donda A, Restifo NP, Held W, Gattinoni L, Romero P. 2013. MicroRNA-155 is required for effector CD8+ T cell responses to virus infection and cancer. Immunity, 38, 742–753. [CrossRef] [PubMed] [Google Scholar]
  9. Dunay IR, Gajurel K, Dhakal R, Liesenfeld O, Montoya JG. 2018. Treatment of toxoplasmosis: historical perspective, animal models, and current clinical practice. Clinical Microbiology Reviews, 31, e00057-17. [CrossRef] [PubMed] [Google Scholar]
  10. Dupont CD, Christian DA, Hunter CA. 2012. Immune response and immunopathology during toxoplasmosis. Seminars in Immunopathology, 34, 793–813. [CrossRef] [PubMed] [Google Scholar]
  11. Elsheikha HM. 2008. Congenital toxoplasmosis: priorities for further health promotion action. Public Health, 122, 335–353. [CrossRef] [PubMed] [Google Scholar]
  12. Elsheikha HM, Marra CM, Zhu XQ. 2020. Epidemiology, pathophysiology, diagnosis, and management of cerebral toxoplasmosis. Clinical Microbiology Reviews, 34, e00115-19. [CrossRef] [PubMed] [Google Scholar]
  13. Gigley JP, Fox BA, Bzik DJ. 2009. Cell-mediated immunity to Toxoplasma gondii develops primarily by local Th1 host immune responses in the absence of parasite replication. Journal of Immunology, 182, 1069–1078. [CrossRef] [PubMed] [Google Scholar]
  14. Ham DW, Kim SG, Seo SH, Shin JH, Lee SH, Shin EH. 2021. Chronic Toxoplasma gondii infection alleviates experimental autoimmune encephalomyelitis by the immune regulation inducing reduction in IL-17A/Th17 via upregulation of SOCS3. Neurotherapeutics, 18, 430–447. [CrossRef] [PubMed] [Google Scholar]
  15. Im SJ, Konieczny BT, Hudson WH, Masopust D, Ahmed R. 2020. PD-1+ stemlike CD8 T cells are resident in lymphoid tissues during persistent LCMV infection. Proceedings of the National Academy of Sciences of the United States of America, 117, 4292–4299. [CrossRef] [PubMed] [Google Scholar]
  16. Jha BK, Varikuti S, Seidler GR, Volpedo G, Satoskar AR, McGwire BS. 2020. MicroRNA-155 deficiency exacerbates Trypanosoma cruzi infection. Infection and Immunity, 88, e00948-19. [PubMed] [Google Scholar]
  17. Li J, Lee Y, Li Y, Jiang Y, Lu H, Zang W, Zhao X, Liu L, Chen Y, Tan H, Yang Z, Zhang MQ, Mak TW, Ni L, Dong C. 2018. Co-inhibitory molecule B7 superfamily member 1 expressed by tumor-infiltrating myeloid cells induces dysfunction of anti-tumor CD8+ T Cells. Immunity, 48, 773–786.e5. [CrossRef] [PubMed] [Google Scholar]
  18. Macfarlane LA, Murphy PR. 2010. MicroRNA: Biogenesis, function and role in cancer. Current Genomics, 11, 537–561. [CrossRef] [PubMed] [Google Scholar]
  19. Mehta A, Baltimore D. 2016. MicroRNAs as regulatory elements in immune system logic. Nature Reviews Immunology, 16, 279–294. [CrossRef] [PubMed] [Google Scholar]
  20. Montoya JG, Liesenfeld O. 2004. Toxoplasmosis. Lancet, 363, 1965–1976. [CrossRef] [PubMed] [Google Scholar]
  21. Rothchild AC, Sissons JR, Shafiani S, Plaisier C, Min D, Mai D, Gilchrist M, Peschon J, Larson RP, Bergthaler A, Baliga NS, Urdahl KB, Aderem A. 2016. MiR-155-regulated molecular network orchestrates cell fate in the innate and adaptive immune response to Mycobacterium tuberculosis. Proceedings of the National Academy of Sciences of the United States of America, 113, E6172–E6181. [CrossRef] [PubMed] [Google Scholar]
  22. Sasai M, Pradipta A, Yamamoto M. 2018. Host immune responses to Toxoplasma gondii. International Immunology, 30, 113–119. [CrossRef] [PubMed] [Google Scholar]
  23. Sasai M, Yamamoto M. 2019. Innate, adaptive, and cell-autonomous immunity against Toxoplasma gondii infection. Experimental and Molecular Medicine, 51, 1–10. [CrossRef] [PubMed] [Google Scholar]
  24. Stelekati E, Chen Z, Manne S, Kurachi M, Ali MA, Lewy K, Cai Z, Nzingha K, McLane LM, Hope JL, Fike AJ, Katsikis PD, Wherry EJ. 2018. Long-term persistence of exhausted CD8 T cells in chronic infection is regulated by MicroRNA-155. Cell Reports, 23, 2142–2156. [CrossRef] [PubMed] [Google Scholar]
  25. Tenter AM, Heckeroth AR, Weiss LM. 2000. Toxoplasma gondii: from animals to humans. International Journal for Parasitology, 30, 1217–1258. [CrossRef] [PubMed] [Google Scholar]
  26. Tait ED, Jordan KA, Dupont CD, Harris TH, Gregg B, Wilson EH, Pepper M, Dzierszinski F, Roos DS, Hunteret CA. 2010. Virulence of Toxoplasma gondii is associated with distinct dendritic cell responses and reduced numbers of activated CD8+ T cells. Journal of Immunology, 185, 1502–1512. [CrossRef] [PubMed] [Google Scholar]
  27. Trotta R, Chen L, Costinean S, Josyula S, Mundy-Bosse BL, Ciarlariello D, Mao C, Briercheck EL, McConnell KK, Mishra A, Yu L, Croce CM, Caligiuri MA. 2013. Overexpression of miR-155 causes expansion, arrest in terminal differentiation and functional activation of mouse natural killer cells. Blood, 121, 3126–3134. [CrossRef] [PubMed] [Google Scholar]
  28. Trotta R, Chen L, Ciarlariello D, Josyula S, Mao C, Costinean S, Yu L, Butchar JP, Tridandapani S, Croce CM, Caligiuri MA. 2012. miR-155 regulates IFN-γ production in natural killer cells. Blood, 119, 3478–3485. [CrossRef] [PubMed] [Google Scholar]
  29. Utzschneider DT, Gabriel SS, Chisanga D, Gloury R, Gubser PM, Vasanthakumar A, Vasanthakumar A, Shi W, Kallies A. 2020. Early precursor T cells establish and propagate T cell exhaustion in chronic infection. Nature Immunology, 21, 1256–1266. [CrossRef] [PubMed] [Google Scholar]
  30. Varikuti S, Natarajan G, Volpedo G, Singh B, Hamza O, Messick GV, Arellano MG, Papenfuss TL, Oghumu S, Satoskar AR. 2019. MicroRNA 155 Contributes to host immunity against Leishmania donovani but is not essential for resolution of infection. Infection and Immunity, 87, e00307-19. [CrossRef] [PubMed] [Google Scholar]
  31. Vigorito E, Kohlhaas S, Lu D, Leyland R. 2013. miR-155: an ancient regulator of the immune system. Immunological Reviews, 253, 146–157. [CrossRef] [PubMed] [Google Scholar]
  32. Wang JL, Zhang NZ, Li TT, He JJ, Elsheikha HM, Zhu XQ. 2019. Advances in the development of anti-Toxoplasma gondii vaccines: Challenges, opportunities, and perspectives. Trends in Parasitology, 35, 239–253. [CrossRef] [PubMed] [Google Scholar]
  33. Zhang NZ, Gao Q, Wang M, Hou JL, Zhang FK, Hu LY, Zhu XQ. 2018. Protective efficacy against acute and chronic Toxoplasma gondii infection induced by immunization with the DNA vaccine TgDOC2C. Frontiers in Microbiology, 9, 2965. [CrossRef] [PubMed] [Google Scholar]
  34. Zhou CX, Ai K, Huang CQ, Guo JJ, Cong H, He SY, Zhu XQ. 2020. miRNA and circRNA expression patterns in mouse brain during toxoplasmosis development. BMC Genomics, 21, 46. [CrossRef] [PubMed] [Google Scholar]
  35. Zhu YC, He Y, Liu JF, Chen J. 2020. Adjuvantic cytokine IL-33 improves the protective immunity of cocktailed DNA vaccine of ROP5 and ROP18 against Toxoplasma gondii infection in mice. Parasite, 27, 26. [CrossRef] [EDP Sciences] [PubMed] [Google Scholar]
  36. Zhu YC, Elsheikha HM, Wang JH, Fang S, He JJ, Zhu XQ, Chen J. 2021. Synergy between Toxoplasma gondii type I Δ GRA17 immunotherapy and PD-L1 checkpoint inhibition triggers the regression of targeted and distal tumors. Journal for ImmunoTherapy of Cancer, 9, e002970. [CrossRef] [PubMed] [Google Scholar]

Cite this article as: Xu Y, Wu J, Yuan X, Liu W, Pan J & Xu B. 2021. MicroRNA-155 contributes to host immunity against Toxoplasma gondii. Parasite 28, 83.

All Tables

Table 1

Primer sequences.

All Figures

thumbnail Figure 1

miR-155 deficiency enhanced cyst burden and reduced survival time in mice models. (A) Survival curves of mice after challenge of cysts of the PRU strain. miR-155−/− mice had 0% survival at day 17. (B) Cyst loads were counted from whole brain homogenates of mice 4 weeks after challenge. The bars represented the mean cyst burden per mouse brain after challenge orally with a dose of 10 cysts of the PRU strain. Data are mean ± SD (representative of three experiments). **p < 0.01; *p < 0.05, compared with the WT groups.

In the text
thumbnail Figure 2

Evaluation of Th-1\Th-2\Th-17 immune responses in WT and miR-155−/− mice. The levels of IFN-γ, IL-2, IL-13, IL-4, IL-17A and IL-17F were measured from splenic lymphocytes of WT- and miR-155KO-infected mice. Significant decreases in IFN-γ, IL-2, IL-13 were observed in miR-155−/− mice. Data are shown from one representative experiment of three independent experiments and presented as a mean ± SD. ***p < 0.001; N/S = not significant.

In the text
thumbnail Figure 3

miR-155 deficiency impairs the recruitment of CD4+ T cells, CD8+ T cells, NK and NKT cell. Spleens were collected from each mouse in the WT and miR-155−/− groups. The splenocytes labeled with cell-specific fluorescent labeled antibodies were analysed by flow cytometry. Results were calculated and expressed as a mean ± SD of three independent experiments. ***p < 0.001; **p < 0.01.

In the text
thumbnail Figure 4

miR-155 deficiency exacerbated CD8+ T cell exhaustion by targeting to SHIP-1 and SOCS1. Spleens were collected from each mouse in the WT and miR-155−/− groups. The percentages of Tregs, IFN-γ CD8+ T, TNF-α CD8+ T and PD1 CD8+ T cells were analysed by flow cytometric analysis. The expressions of SHIP-1 and SOCS1 were estimated by qRT-PCR. Data are shown from one representative experiment of three independent experiments and presented as a mean ± SD. ***p < 0.001; **p < 0.01.

In the text
thumbnail Figure 5

Decreased accumulation of splenic inflammatory monocytes and DCs and expression of pro-inflammatory mediators in miR-155KO-infected mice. Flow cytometric analysis of splenic inflammatory monocytes and DCs of parasite infected-WT and miR-155−/− mice. The expression of pro-inflammatory mediators was analysed by qRT-PCR. Data are presented as a mean ± SD of three independent experiments. The asterisks indicate the statistical significance of difference between WT and miR-155−/− mice. ***p < 0.001; **p < 0.01.

In the text

Current usage metrics show cumulative count of Article Views (full-text article views including HTML views, PDF and ePub downloads, according to the available data) and Abstracts Views on Vision4Press platform.

Data correspond to usage on the plateform after 2015. The current usage metrics is available 48-96 hours after online publication and is updated daily on week days.

Initial download of the metrics may take a while.