Nature - USA (2020-05-14)

(Antfer) #1

208 | Nature | Vol 581 | 14 May 2020


Article


here, even though our T-cell ablation study does not directly prove that
such T cells relay noradrenergic signals to B cells.
Our surgical ablation of the splenic nerve, instead of catecholamin-
ergic denervation by neurotoxins, allowed us to demonstrate that
an efferent pathway that descends into the spleen is the mechanism
responsible for the immunostimulatory effects of CRH neurons. These
effects do not appear to involve acute reflexes, but rather hinge on
tonic and provoked activities in CeA and PVN CRH neurons, which are
intrinsically rhythmic^16 and may be modulated by inputs from other
brain areas^17. The CRH-controlled HPA axis can also potently suppresses
immune responses by releasing glucocorticoids. It is likely that, above
a certain threshold of activation intensity, secretion of the HPA-driving
hormone and consequent immunosuppression would dominate over
the immunostimulatory effects of CRH neurons; below such a thresh-
old, immunostimulation by neuronal signals descending into the spleen
are more dominant.
In summary, our findings demonstrate brain control of adaptive
immunity via direct neural connection. We speculate that bodily


behaviours and psychological conditioning could be used to induce
stress of an appropriate strength and form, eventually to be defined
in qualitative and quantitative terms of neuron activation in specific
brain areas, in order to enhance host adaptive immunity.

Online content
Any methods, additional references, Nature Research reporting sum-
maries, source data, extended data, supplementary information,
acknowledgements, peer review information; details of author con-
tributions and competing interests; and statements of data and code
availability are available at https://doi.org/10.1038/s41586-020-2235-7.


  1. Rosas-Ballina, M. et al. Acetylcholine-synthesizing T cells relay neural signals in a vagus
    nerve circuit. Science 334 , 98–101 (2011).

  2. Rinner, I. & Schauenstein, K. Detection of choline-acetyltransferase activity in
    lymphocytes. J. Neurosci. Res. 35 , 188–191 (1993).

  3. Sato, K. Z. et al. Diversity of mRNA expression for muscarinic acetylcholine receptor
    subtypes and neuronal nicotinic acetylcholine receptor subunits in human mononuclear
    leukocytes and leukemic cell lines. Neurosci. Lett. 266 , 17–20 (1999).

  4. Wessler, I. & Kirkpatrick, C. J. Acetylcholine beyond neurons: the non-neuronal
    cholinergic system in humans. Br. J. Pharmacol. 154 , 1558–1571 (2008).

  5. Saalmüller, A. & Mettenleiter, T. C. Rapid identification and quantitation of cells infected
    by recombinant herpesvirus (pseudorabies virus) using a fluorescence-based
    β-galactosidase assay and flow cytometry. J. Virol. Methods 44 , 99–108 (1993).

  6. Cano, G., Sved, A. F., Rinaman, L., Rabin, B. S. & Card, J. P. Characterization of the central
    nervous system innervation of the rat spleen using viral transneuronal tracing. J. Comp.
    Neurol. 439 , 1–18 (2001).

  7. Keifer, O. P. Jr, Hurt, R. C., Ressler, K. J. & Marvar, P. J. The physiology of fear:
    reconceptualizing the role of the central amygdala in fear learning. Physiology  30 ,
    389–401 (2015).

  8. Herman, J. P. & Tasker, J. G. Paraventricular hypothalamic mechanisms of chronic stress
    adaptation. Front. Endocrinol. 7 , 137 (2016).

  9. Peng, J. et al. A quantitative analysis of the distribution of CRH neurons in whole mouse
    brain. Front. Neuroanat. 11 , 63 (2017).

  10. Kadmiel, M. & Cidlowski, J. A. Glucocorticoid receptor signaling in health and disease.
    Trends Pharmacol. Sci. 34 , 518–530 (2013).

  11. Vandevyver, S., Dejager, L., Tuckermann, J. & Libert, C. New insights into the
    anti-inflammatory mechanisms of glucocorticoids: an emerging role for
    glucocorticoid-receptor-mediated transactivation. Endocrinology 154 , 993–1007
    (2013).

  12. Roth, B. L. DREADDs for neuroscientists. Neuron 89 , 683–694 (2016).

  13. Nutt, S. L., Hodgkin, P. D., Tarlinton, D. M. & Corcoran, L. M. The generation of
    antibody-secreting plasma cells. Nat. Rev. Immunol. 15 , 160–171 (2015).

  14. Ben-Shaanan, T. L. et al. Activation of the reward system boosts innate and adaptive
    immunity. Nat. Med. 22 , 940–944 (2016).

  15. Tracey, K. J. The inflammatory reflex. Nature 420 , 853–859 (2002).

  16. Lightman, S. L. & Conway-Campbell, B. L. The crucial role of pulsatile activity of the HPA
    axis for continuous dynamic equilibration. Nat. Rev. Neurosci. 11 , 710–718 (2010).

  17. Ulrich-Lai, Y. M. & Herman, J. P. Neural regulation of endocrine and autonomic stress
    responses. Nat. Rev. Neurosci. 10 , 397–409 (2009).


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

© The Author(s), under exclusive licence to Springer Nature Limited 2020

a b d f

0

1

2

3

2 –10 2 –5 20

P = 0.1 P = 0.2 P = 0.9
Untreated

c eg

P (1.73-fold)< 0.0001

Ablation control

CRH ablation

Sham-operated

Spleen-denervated μMT: Chrna9–/–

0

1

2

3

0

1

2

3

0

1

2

3

0

1

2

3

0

1

2

3

1

2

3

2 –10 2 –5 20 2 –10 2 –5 20 2 –10^2 –5 20 2 –10^2 –5^20

2 –10 2 –5 20 2 –10 2 –5 20

EPS

P (1.41-fold)= 0.003 P (1.36-fold)= 0.01 P (1.43-fold)= 0.004

μMT: Chrna9+/+

Dilution factor Dilution factor Dilution factor Dilution factor

Dilution factor Dilution factor Dilution factor

Optical density Optical density Optical density Optical density

Optical density Optical density Optical density

Fig. 5 | EPS enhances the antigen-specif ic antibody response via the PVN/
CeA–splenic nerve axis. NP-specific IgG titres of NP-KLH-immunized control
or EPS-experienced mice following manipulations of the CeA/PVN–splenic
nerve axis. a, B6 mice that were untreated (n = 17) or subjected to EPS (n = 16).
b, c, Control (b) or CeA/PVN CRH neuron-ablated (c) mice; untreated control
(n = 14), EPS control (n = 13), untreated ablation (n = 14), EPS ablation (n = 1 1).
d, e, Sham-operated (d) or spleen-denervated (e) mice; untreated
sham-operated (n = 5), EPS sham-operated (n = 6), untreated denervated (n = 6),
EPS denervated (n = 6). f, g, μMT Chrna9+/+ (d) or μMT Chrna9−/− (e) chimaeric
mice; untreated μMT Chrna9+/+ (n = 16), EPS μMT Chrna9+/+ (n = 15), untreated
μMT Chrna9−/− (n = 16), EPS μMT Chrna9−/− (n = 14). On x-axis (dilution factor), 2^0
corresponds to 1:800 of the original serum. Mean ± s.e.m. of all animals at each
dilution overlaid with dose–response regression curves (goodness of fit
R^2  > 0.9 in all cases). For each untreated-versus-EPS comparison, fold changes in
IgG titres were calculated by EC 50 of the respective curves, with between-group
P values by extra sum-of-squares F-test.

Free download pdf