Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Rescue of maternal immune activation-induced behavioral abnormalities in adult mouse offspring by pathogen-activated maternal Treg cells

Abstract

Maternal immune activation (MIA) induced by lipopolysaccharides or polyinosinic:polycytidylic acid injections can induce behavioral abnormalities in adult mouse offspring. Here, we used the soluble tachyzoite antigen from Toxoplasma gondii, a parasite that infects approximately two billion people, to induce MIA in mice. The adult male offspring showed autism-relevant behaviors and abnormal brain microstructure, along with a pro-inflammatory T-cell immune profile in the periphery and upregulation of interleukin-6 in brain astrocytes. We show that adoptive transfer of regulatory T (Treg) cells largely reversed these MIA-induced phenotypes. Notably, pathogen-activated maternal Treg cells showed greater rescue efficacy than those from control donors. Single-cell RNA sequencing identified and characterized a unique group of pathogen-activated Treg cells that constitute 32.6% of the pathogen-activated maternal Treg population. Our study establishes a new preclinical parasite-mimicking MIA model and suggests therapeutic potential of adoptive Treg cell transfer in neuropsychiatric disorders associated with immune alterations.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: STAg-elicited MIA leads to ASD-like behaviors in the offspring.
Fig. 2: STAg-MIA altered peripheral CD4+ T-cell differentiation in adult offspring.
Fig. 3: STAg-MIA-induced upregulation of astrocytic IL-6 in the adult offspring.
Fig. 4: Adoptive transfer of Treg cells restores CD4+ T-cell profile in MIA offspring.
Fig. 5: Therapeutic effects of adoptive transfer of Treg cells in adult MIA offspring.
Fig. 6: Adoptive transfer of Treg cells suppresses pro-inflammatory IL-6 expression in ASD-like offspring brain.
Fig. 7: Identification and characterization of unique subsets of MIATreg cells.
Fig. 8: Enhanced MIATreg cell infiltration into the brain parenchyma in adult MIA offspring.

Similar content being viewed by others

Data availability

The scRNA-sequencing dataset is available in the Gene Expression Omnibus database (https://www.ncbi.nlm.nih.gov/) under accession code GSE132803. Source data are provided with this paper. All other data presented in this study are available from the corresponding authors upon request.

Code availability

The R code for scRNA-seq analysis can be found in the Supplementary Software.

References

  1. Abdallah, M. W. et al. Amniotic fluid chemokines and autism spectrum disorders: an exploratory study utilizing a Danish Historic Birth Cohort. Brain Behav. Immun. 26, 170–176 (2012).

    Article  CAS  PubMed  Google Scholar 

  2. Brown, A. S. & Derkits, E. J. Prenatal infection and schizophrenia: a review of epidemiologic and translational studies. Am. J. Psychiatry 167, 261–280 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Atladóttir, H. Ó. et al. Association of family history of autoimmune diseases and autism spectrum disorders. Pediatrics 124, 687–694 (2009).

    Article  PubMed  Google Scholar 

  4. Atladóttir, H. Ó. et al. Maternal infection requiring hospitalization during pregnancy and autism spectrum disorders. J. Autism Dev. Disord. 40, 1423–1430 (2010).

    Article  PubMed  Google Scholar 

  5. Estes, M. L. & McAllister, A. K. Immune mediators in the brain and peripheral tissues in autism spectrum disorder. Nat. Rev. Neurosci. 16, 469–486 (2015).

    Article  PubMed Central  Google Scholar 

  6. Lee, B. K. et al. Maternal hospitalization with infection during pregnancy and risk of autism spectrum disorders. Brain Behav. Immun. 44, 100–105 (2015).

    Article  PubMed  Google Scholar 

  7. Careaga, M., Murai, T. & Bauman, M. D. Maternal immune activation and autism spectrum disorder: from rodents to nonhuman and human primates. Biol. Psychiatry 81, 391–401 (2017).

    Article  CAS  PubMed  Google Scholar 

  8. Knuesel, I. et al. MIA and abnormal brain development across CNS disorders. Nat. Rev. Neurol. 10, 643–660 (2014).

    Article  Google Scholar 

  9. Choi, G. B. et al. The maternal interleukin-17a pathway in mice promotes autism-like phenotypes in offspring. Science 351, 933–939 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hsiao, E. Y., McBride, S. W., Chow, J., Mazmanian, S. K. & Patterson, P. H. Modeling an autism risk factor in mice leads to permanent immune dysregulation. Proc. Natl Acad. Sci. USA 109, 12776–12781 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Parker-Athill, E. C. & Tan, J. Maternal immune activation and autism spectrum disorder: interleukin-6 signaling as a key mechanistic pathway. Neurosignals 18, 113–128 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wu, W.-L., Hsiao, E. Y., Yan, Z., Mazmanian, S. K. & Patterson, P. H. The placental interleukin-6 signaling controls fetal brain development and behavior. Brain Behav. Immun. 62, 11–23 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Smith, S. E. P., Li, J., Garbett, K., Mirnics, K. & Patterson, P. H. Maternal immune activation alters fetal brain development through interleukin-6. J. Neurosci. 27, 10695–10702 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Rudolph, M. D. et al. Maternal IL-6 during pregnancy can be estimated from newborn brain connectivity and predicts future working memory in offspring. Nat. Neurosci. 21, 765–772 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gumusoglu, S. B. & Stevens, H. E. Maternal inflammation and neurodevelopmental programming: a review of preclinical outcomes and implications for translational psychiatry. Biol. Psychiatry 85, 107–121 (2019).

    Article  PubMed  Google Scholar 

  16. Pape, K., Tamouza, R., Leboyer, M. & Zipp, F. Immunoneuropsychiatry—novel perspectives on brain disorders. Nat. Rev. Neurol. 15, 317-–328 (2019).

    Article  PubMed  Google Scholar 

  17. Mattei, D. et al. MIA results in complex microglial transcriptome signature in the adult offspring that is reversed by minocycline treatment. Transl. Psychiatry 7, e1120 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Brown, A. S. & Meyer, U. MIA and neuropsychiatric illness: a translational research perspective. Am. J. Psychiatry 175, 1073–1083 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Fatoohi, A. F. et al. Heterogeneity in cellular and humoral immune responses against Toxoplasma gondii antigen in humans. Clin. Exp. Immunol. 136, 535–541 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bluestone, J. A. & Tang, Q. Treg cells—the next frontier of cell therapy. Science 362, 154–155 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Ito, M. et al. Brain regulatory T cells suppress astrogliosis and potentiate neurological recovery. Nature 565, 246–250 (2019).

    Article  CAS  PubMed  Google Scholar 

  22. Raffin, C., Vo, L. T. & Bluestone, J. A. Treg cell-based therapies: challenges and perspectives. Nat. Rev. Immunol. 20, 158–172 (2020).

    Article  CAS  PubMed  Google Scholar 

  23. Sharabi, A. et al. Regulatory T cells in the treatment of disease. Nat. Rev. Drug Discov. 17, 823–844 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. Ferreira, L. M. R., Muller, Y. D., Bluestone, J. A. & Tang, Q. Next-generation regulatory T-cell therapy. Nat. Rev. Drug Discov. 18, 749–769 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hohlfeld, P. et al. Toxoplasma gondii infection during pregnancy: T lymphocyte subpopulations in mothers and fetuses. Pediatr. Infect. Dis. J. 9, 878–881 (1990).

    Article  CAS  PubMed  Google Scholar 

  26. Al-Ayadhi, L. Y. & Mostafa, G. A. Elevated serum levels of interleukin-17A in children with autism. J. Neuroinflammation 9, 158 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Baruch, K. et al. Breaking immune tolerance by targeting Foxp3+ regulatory T cells mitigates Alzheimer’s disease pathology. Nat. Commun. 6, 7967 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Mohammad, M. G. et al. Immune cell trafficking from the brain maintains CNS immune tolerance. J. Clin. Invest. 124, 1228–1241 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ahmad, S. F. et al. Dysregulation of TH1, TH2, TH17 and T regulatory cell-related transcription factor signaling in children with autism. Mol. Neurobiol. 54, 4390–4400 (2017).

    Article  CAS  PubMed  Google Scholar 

  30. Mostafa, G. A., Al Shehab, A. & Fouad, N. R. Frequency of CD4+CD25high regulatory T cells in the peripheral blood of egyptian children with autism. J. Child Neurol. 25, 328–335 (2010).

    Article  PubMed  Google Scholar 

  31. Moaaz, M., Youssry, S., Elfatatry, A. & El Rahman, M. A. TH17/Treg cells imbalance and their related cytokines (IL-17, IL-10 and TGF-β) in children with autism spectrum disorder. J. Neuroimmunol. 337, 577071 (2019).

    Article  CAS  PubMed  Google Scholar 

  32. Onore, C., Careaga, M. & Ashwood, P. The role of immune dysfunction in the pathophysiology of autism. Brain Behav. Immun. 26, 383–392 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. Estes, M. L. & McAllister, A. K. Immune mediators in the brain and peripheral tissues in autism spectrum disorder. Nat. Rev. Neurosci. 16, 469–486 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lellem, A. et al. Unique chemotactic response profile and specific expression of chemokine receptors CCR4 and CCR8 by CD4+CD25+ regulatory T cells. J. Exp. Med. 194, 847–854 (2001).

    Article  Google Scholar 

  35. Griffith, J. W., Sokol, C. L. & Luster, A. D. Chemokines and chemokine receptors: positioning cells for host defense and immunity. Annu. Rev. Immunol. 32, 659–702 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Mostowy, S. & Shenoy, A. R. The cytoskeleton in cell-autonomous immunity: structural determinants of host defence. Nat. Rev. Immunol. 15, 559–573 (2015).

    Article  PubMed Central  Google Scholar 

  37. Pollard, T. D. & Cooper, J. A. Actin, a central player in cell shape and movement. Science 326, 1208–1212 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Fletcher, D. A. & Mullins, R. D. Cell mechanics and the cytoskeleton. Nature 463, 485–495 (2010).

    Article  Google Scholar 

  39. Yang, G., Pan, F., Parkhurst, C. N., Grutzendler, J. & Gan, W.-B. Thinned-skull cranial window technique for long-term imaging of the cortex in live mice. Nat. Protoc. 5, 201–208 (2010).

    Article  PubMed Central  Google Scholar 

  40. Herz, J. et al. In vivo imaging of lymphocytes in the CNS reveals different behaviour of naïve T cells in health and autoimmunity. J. Neuroinflammation 8, 131 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Otsu, Y. et al. Control of aversion by glycine-gated GluN1/GluN3A NMDA receptors in the adult medial habenula. Science 366, 250–254 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zhang, J. et al. Presynaptic excitation via GABAB receptors in habenula cholinergic neurons regulates fear memory expression. Cell 166, 716–728 (2016).

    Article  CAS  PubMed  Google Scholar 

  43. Orefice, L. L. et al. Peripheral mechanosensory neuron dysfunction underlies tactile and behavioral deficits in mouse models of ASDs. Cell 166, 299–313 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hitti, F. L. & Siegelbaum, S. A. The hippocampal CA2 region is essential for social memory. Nature 508, 88–92 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Leroy, F. et al. A circuit from hippocampal CA2 to lateral septum disinhibits social aggression. Nature 564, 213–218 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Spann, M. N., Monk, C., Scheinost, D. & Peterson, B. S. Maternal immune activation during the third trimester is associated with neonatal functional connectivity of the salience network and fetal to toddler behavior. J. Neurosci. 38, 2877–2886 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Missault, S. et al. Hypersynchronicity in the default mode-like network in a neurodevelopmental animal model with relevance for schizophrenia. Behav. Brain Res. 364, 303–316 (2019).

    Article  PubMed  Google Scholar 

  48. Kipnis, J., Gadani, S. & Derecki, N. C. Pro-cognitive properties of T cells. Nat. Rev. Immunol. 12, 663–669 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gadani, S. P., Cronk, J. C., Norris, G. T. & Kipnis, J. IL-4 in the brain: a cytokine to remember. J. Immunol. 189, 4213–4219 (2012).

    Article  CAS  PubMed  Google Scholar 

  50. Derecki, N. C. et al. Regulation of learning and memory by meningeal immunity: a key role for IL-4. J. Exp. Med. 207, 1067–1080 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Shin Yim, Y. et al. Reversing behavioural abnormalities in mice exposed to maternal inflammation. Nature 549, 482–487 (2017).

    Article  PubMed  Google Scholar 

  52. Behrens, T. E. et al. Characterization and propagation of uncertainty in diffusion-weighted MR imaging. Magn. Reson. Med. 50, 1077–1088 (2003).

    Article  CAS  PubMed  Google Scholar 

  53. Kong, Y. et al. Variation in anisotropy and diffusivity along the medulla oblongata and the whole spinal cord in adolescent idiopathic scoliosis: a pilot study using diffusion tensor imaging. AJNR Am. J. Neuroradiol. 35, 1621–1627 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wedeen, V. J. et al. Diffusion spectrum magnetic resonance imaging tractography of crossing fibers. Neuroimage 41, 1267–1277 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Yushkevich, P. A. et al. User-guided 3D active contour segmentation of anatomical structures: significantly improved efficiency and reliability. Neuroimage 31, 1116–1128 (2006).

    Article  PubMed  Google Scholar 

  56. Zhou, Z. et al. The C-terminal tails of endogenous GluA1 and GluA2 differentially contribute to hippocampal synaptic plasticity and learning. Nat. Neurosci. 21, 50–62 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank H.T. Wu for technical assistance with the USV experiments in pups. We thank all members of the laboratories of M.J. and Z.Z. for technical assistance and discussion. This work was supported by the National Natural Science Foundation of China (81971022 to Z.Z.), Program of Shanghai Academic Research Leader (19XD1423300 to Z.Z.), Natural Science Foundation of Jiangsu Province (BK20171049 to Z.X.), Science and Technology Commission of Shanghai Municipality (201409002600 to Z.Z.), Foundation of Jiangsu Province Key Laboratory of Modern Pathogen Biology (JX218GSP20171003 to Z.Z.), Shanghai Key Laboratory of Psychotic Disorders Open Grant (19-K01 to M.J.), Shanghai Mental Health Center-Clinical Research Center (CRC2019ZD01 to Z.Z.) and Shanghai Municipal Education Commission-Gaofeng Clinical Medicine Grant Support (20191835 to Z.Z.).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: Z.Z. and M.J.; STAg preparation and injection: Z.X., R.L., Y.N. and M.H.; behavioral tests and analysis: Xiaoyun Zhang, Xiaolin Zhang, Y.K. and Z.W.; flow cytometry: Z.X., H.C., Y.N. and W.-T.L.; immunohistochemistry: Xiaoyun Zhang, Y.K., Z.X. and S.Y.; two-photon imaging: Y.K. and Xiaoyun Zhang; Treg cell preparation and adoptive transfer: Z.X., H.C. and Y.K.; scRNA-seq and analysis: Xiaolin Zhang and Xiaoyun Zhang; MRI and analysis: Y.H., Z.Y. and R.M.; western blot, qPCR and ELISA: Xiaoyun Zhang, H.C. and Y.D.; funding acquisition: Z.Z., M.J. and Z.X.; supervision: Z.Z. and M.J.; Z.Z. wrote the paper with input from the co-authors.

Corresponding authors

Correspondence to Minjun Ji or Zikai Zhou.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Neuroscience thanks Staci Bilbo, Qizhi Tang, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Pro-inflammatory CD4+ T cell profile in the pregnant mother mice indicates STAg-elicited MIA.

a, Gating schemes for analysis of the percentages of CD3+CD4+ IFN-γ+ (TH1), CD3+CD4+IL-4+(TH2), and CD3+CD4+IL-17+(TH17) cells from R1 and R2. The expression of CD4+CD25+Foxp3+ (Treg) cells were gated from R1 and R3. Single-cell suspensions of splenic cells were labeled with CD3-APC and CD4-FITC, and then intracellularly labeled with PE-conjugated antibodies against IFN-γ, IL-4 or IL-17 for flow cytometric analysis of CD3+CD4+ IFN-γ+ (TH1), CD3+CD4+IL-4+ (TH2), and CD3+CD4+IL-17+ (TH17) cells, respectively. Cells were stained with CD4-FITC and CD25-APC, and then intracellularly labeled with PE-conjugated antibodies against Foxp3 for FACS analysis of CD4+CD25+Foxp3+ Treg cells. b-d, Flow cytometric analysis of splenic lymphocytes from pregnant mother mice prepared at three days after STAg injection. b, Bar chart shows the percentages of TH1, TH2, TH17 and Treg cells in CD4+ splenic lymphocytes. c, The absolute numbers of total splenic cells and (d) CD4+ T cells in PBS- or STAg-treated pregnant mother mice. Data are presented as mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 2 Further behavioral analysis on the MIA offspring.

a-l, Maternal isolation-induced USV profiles. (a and b) Representative USV spectrograms from PBS and STAg pups on P12 and P14. The peak frequency at start (c), end (d) and the mean peak frequency (e); the peak amplitude at start (f), end (g) and the mean peak amplitude (h) of calls made by PBS and STAg pups at P5, P7, P9, P12 and P14. i-j, The sonograms of USVs emitted by the pups are classified into ten distinct categories and analyzed for the number of calls (i) and the duration of the calls (j) during the 5-min recording period at P7. (k and l) Ten syllable categories of 5-min recording were analyzed at P14 for the number of calls (k) and the duration of the calls (l). m-n, Open filed test. Total distance traveled (m) and mean speed (n) in 10 min of open field test. o-p, Elevated plus maze test. Number of entries into open arms (o) and percentage of entries into open arms (p). q, Olfactory function test. PBS and STAg offspring displayed comparable investigation when exposed to three non-social cues (water, banana, orange) and two social cues. r-s, The social interaction ratio in reciprocal social interactions. The ratio of experimental mouse and stimulus mouse during the test with age-matched peer (r) or a juvenile target (s). t-v, Three-chamber social interaction test. (t) Time spent in the left and right chambers during the habituation stage by PBS and STAg-MIA offspring. (u) Total time spent in the empty chamber and the chamber with stranger mouse 1 (S1) at social approach stage. (v) Total time spent in the chamber with S1 and the chamber with stranger mouse 2 (S2) at social novelty stage. Data are presented as mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 3 STAg-elicited MIA did not alter learning and memory in the offspring.

a-e, Novel object recognition test. (a) Representative heatmaps of mouse movement at the training session (top) and test session (bottom) of PBS and STAg offspring. (b) Time spent exploring the identical objects A1 and A2 and discrimination index (c) in training session. (d) Time spent exploring the familiar object A and a novel object B and discrimination index (e) in test session. f-j, Barnes maze test. (f) The experimental paradigm. (g) The image of probe test on day 5 and representative heatmaps of PBS and STAg offspring movement. (h) The latency time to enter the hiding box in the Barnes maze during acquisition days 1–4. (i and j) The number of visits to the target hole (i) and time in target quadrant (j) in probe test. Data are presented as mean ± s.e.m. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 4 Immune cell profiles in MIA offspring.

a-e, Pregnant C57BL/6 mice were i.p. injected with STAg or vehicle (PBS) on E14.5. At 2 and 4 weeks of age, 2×106 bone marrow cells were stained with CD3-PerCP-Cy5.5, CD19-APC, and Gr1-FITC, respectively. a, Gating schemes for analysis of T cells (CD19CD3+), B cells (CD19+CD3) and granulocytes (Gr-1+CD19) were gated from R1. b, Flow cytometric analysis of proportion of T cells (CD19CD3+), B cells (CD19+CD3) (b and c), and granulocytes (Gr-1+CD19) (d and e) in bone marrow cells. f-j, (f) Gating schemes for analysis of the percentages of CD3+CD4+ (CD4+ T cells) and CD3+CD4+ CXCR5+PD1+ (Tfh) cells in the offspring. Single-cell suspensions of splenic cells were labeled with CD3-APC and CD4-FITC for flow cytometric analysis of CD3+CD4+ cells (g), and the percentage of CD3+CD4+ cells were analyzed (h). i, Cells were labeled with CD3-APC, CD4-FITC, CXCR5-Percp-Cy5.5, and PD1-PE for flow cytometric analysis of TFH cells (j). Data are presented as mean ± s.e.m. ****P < 0.0001. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 5 STAg-MIA specifically upregulated IL-6 expression in multiple brain regions in adult offspring.

a-c, The mRNA levels of IL-12, TNF-α, IFN-γ, IL-10, and IL-4 in the mouse hippocampi (a), striatum (b), cortices (c) were determined by qPCR in offspring from PBS and STAg treated mother mice. The gene mRNA levels were normalized to Gapdh. d, IL-6 protein level in serum measured by ELISA. Data are presented as mean ± s.e.m. *P < 0.05, **P < 0.01, ****P < 0.0001. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 6 Further analysis of therapeutic effects of adoptive transfer of Treg cells in adult MIA offspring.

a and b, Open fields test. At 8 weeks of age, 5×105 of CTreg or MIATreg cells were transferred via intravenous injection into each adult PBS and STAg-MIA offspring. One week later, these mice were subjected to behavioral tests. Total distance traveled (a) and mean speed (b) in 10 min of open field test. c-e, Elevated plus maze test. Total time (c) and the percentage of time spent (d) in open arms, and the percentage of entries into open arms (e). f-g, The social interaction ratio in reciprocal social interactions. The ratio of experimental mouse and stimulus mouse during the test with age-matched mouse (f) or a juvenile target (g) measured after adoptive transfer of CTreg or MIATreg cells in PBS or STAg-MIA offspring. h-j, Three-chamber social interaction test. (h) Time spent in the left and right chambers during the habituation stage. i, Total time spent in the empty chamber and the chamber with stranger mouse 1 (S1) in social approach stage. j, Total time spent in the chamber with stranger mouse 1 (S1) and the chamber with stranger mouse 2 (S2) in social novelty stage. Data are presented as mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 7 Long-term maintenance of transferred EGFP-Treg cells in MIA offspring.

a, Pregnant EGFP or WT C57BL/6 mice were i.p. injected with STAg or vehicle (PBS) on E14.5. CTreg or MIATreg cells (5×105) from EGPF mice were purified and transferred via intravenous injection into each adult STAg-MIA offspring. At 3 weeks and 8 weeks after adoptive transfer, 2×106 cells were stained with CD4-BV21and CD25-APC in the spleen and blood from the STAg-MIA offspring. The EGFP+ cells were gated from CD4+CD25+ cells. Statistical results of the percentage of CD4+CD25+ cells in EGFP+ cells in the blood (b) and spleen (c). Data are presented as mean ± s.e.m. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 8 Distribution of adoptively transferred Treg cells in the brain regions in adult MIA offspring.

a, Coronal plane of the mouse left hemisphere combined with immunohistology image of the right hemisphere section with matching stereotaxic coordinates (bregma −0.82 mm and interaural 2.98 mm). Adoptively transferred Treg cells are labeled with CMTMR (red), nuclei are labeled with DAPI (blue). Scale bar = 500 μm. b, Zoom-in images of representative brain regions: medial habenular nucleus (MHb) (region 1), cingulate/retrosplenial cortex (Cg/RS) (region 2), and primary somatosensory cortex, barrel field 0 (S1BF) (region 3). Scale bars = 25 μm. c, Coronal plane of the mouse left hemisphere combined with immunohistology image of the right hemisphere section with matching stereotaxic coordinates (bregma −2.46 mm and interaural 1.34 mm). Scale bar = 500 μm. d, Zoom-in images of representative brain regions: pre-commissural nucleus (PrC) (region 4), retrosplenial granular cortex (RSG) (region 5), and field CA2 of hippocampus (CA2) (region 6). The distribution pattern of transferred Tregs is highly similar in three independently repeated experiments.

Extended Data Fig. 9 Flow cytometric analysis of cell surface proteins in Treg cells.

a-f, Pregnant C57BL/6 mice were i.p. injected with STAg or vehicle (PBS) on E14.5. Splenic cells (2×106) were stained with CD4-FITC and CD25-APC, and then labeled with PD1-PE, CCR5-Percp-cy5.5, ICOS-BV421, KLRG-1-PE, or CCR4-PE for flow cytometric analysis of cell surface protein levels of the suppressive markers in CTreg or MIATreg cells. The percentages of PD1, CCR5, ICOS, KLRG-1, and CCR4 in CTreg or MIATreg cells were gated from CD4+CD25+ cells (a). Statistical comparation of the surface levels of PD1 (b), ICOS (c), KLRG1 (d), CCR4 (e) and CCR5 (f) are presented respectively. Data are presented as mean ± s.e.m. *P < 0.05, **P < 0.01, ****P < 0.0001. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 10 MRI analysis of neuronal connectivity in the adult brain.

a, Different layers of T2 weighted whole brain images of P60 control (PBS) and STAg-MIA offspring mice. Scale bars, 5 mm. b, Summary graph showing unaltered brain area (mm2) and cortical thickness (mm) (c). d, Fiber tractography images of adult mouse brains. The color schemes are: red, left-right; green, ventral-dorsal; and blue, caudal-rostral. e, Fractional anisotropy (FA) values of major white matter tracts. f, Average axial diffusivity (AD), radial diffusivity (RD) (g), and mean diffusivity (MD) (h) of major white matter tracts. Summary results showing fiber density of whole brain (i), hippocampus (Hip) (j), corpus callosum (CC) (k) and cortex (Ctx) (l). Data are presented as mean ± s.e.m. *P < 0.05, ***P < 0.001. For detailed statistics information, see Supplementary Table 1.

Supplementary information

Reporting Summary

Supplementary Table

Supplementary Table for statistical details.

Supplementary Software

The R code for scRNA-seq analysis.

Source data

Source Data Fig. 3

Unprocessed western blot for Fig. 3.

Source Data Fig. 6

Unprocessed western blot for Fig. 6.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, Z., Zhang, X., Chang, H. et al. Rescue of maternal immune activation-induced behavioral abnormalities in adult mouse offspring by pathogen-activated maternal Treg cells. Nat Neurosci 24, 818–830 (2021). https://doi.org/10.1038/s41593-021-00837-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-021-00837-1

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing