Abstract
The peripheral immune system communicates with the brain through complex anatomical routes involving the skull, the brain borders, circumventricular organs and peripheral nerves. These immune–brain communication pathways were classically considered to be dormant under physiological conditions and active only in cases of infection or damage. Yet, peripheral immune cells and signals are key in brain development, function and maintenance. In this Perspective, we propose an alternative framework for understanding the mechanisms of immune–brain communication. During brain development and in homeostasis, these anatomical structures allow selected elements of the peripheral immune system to affect the brain directly or indirectly, within physiological limits. By contrast, in ageing and pathological settings, detrimental peripheral immune signals hijack the existing communication routes or alter their structure. We discuss why a diversity of communication channels is needed and how they work in relation to one another to maintain homeostasis of the brain.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout





Similar content being viewed by others
References
Ito, M. et al. Brain regulatory T cells suppress astrogliosis and potentiate neurological recovery. Nature 565, 246–250 (2019).
Yshii, L. et al. Astrocyte-targeted gene delivery of interleukin 2 specifically increases brain-resident regulatory T cell numbers and protects against pathological neuroinflammation. Nat. Immunol. 23, 878–891 (2022).
Pasciuto, E. et al. Microglia require CD4 T cells to complete the fetal-to-adult transition. Cell 182, 625–640.e624 (2020).
Shen, Y. et al. CCR5 closes the temporal window for memory linking. Nature 606, 146–152 (2022).
Nautiyal, K. M., Ribeiro, A. C., Pfaff, D. W. & Silver, R. Brain mast cells link the immune system to anxiety-like behavior. Proc. Natl Acad. Sci. USA 105, 18053–18057 (2008).
Gaudenzio, N. et al. Different activation signals induce distinct mast cell degranulation strategies. J. Clin. Invest. 126, 3981–3998 (2016).
Sankowski, R. et al. Multiomic spatial landscape of innate immune cells at human central nervous system borders. Nat. Med. 30, 186–198 (2024).
Villeda, S. A. et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 477, 90 (2011).
Villeda, S. A. et al. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat. Med. 20, 659 (2014).
Fernández-Castañeda, A. et al. Mild respiratory COVID can cause multi-lineage neural cell and myelin dysregulation. Cell 185, 2452–2468 (2022).
Wälchli, T. et al. Single-cell atlas of the human brain vasculature across development, adulthood and disease. Nature 632, 603–613 (2024).
Wu, L. et al. Conditional knockout of IL-1R1 in endothelial cells attenuates seizures and neurodegeneration via inhibiting neuroinflammation mediated by Nrf2/HO-1/NLRP3 signaling in status epilepticus model. Mol. Neurobiol. 61, 4289–4303 (2023).
Nehmé, A. & Edelman, J. Dexamethasone inhibits high glucose-, TNF-α-, and IL-1β-induced secretion of inflammatory and angiogenic mediators from retinal microvascular pericytes. Invest. Ophthalmol. Vis. Sci. 49, 2030–2038 (2008).
Persidsky, Y. et al. Dysfunction of brain pericytes in chronic neuroinflammation. J. Cereb. Blood Flow. Metab. 36, 794–807 (2016).
Leaf, I. A. et al. Pericyte MyD88 and IRAK4 control inflammatory and fibrotic responses to tissue injury. J. Clin. Invest. 127, 321–334 (2017).
Duan, L. et al. PDGFRβ cells rapidly relay inflammatory signal from the circulatory system to neurons via chemokine CCL2. Neuron 100, 183–200.e188 (2018).
Ji, E. et al. The chemokine CCL2 promotes excitatory synaptic transmission in hippocampal neurons via GluA1 subunit trafficking. Neurosci. Bull. 40, 1649–1666 (2024).
Guo, H. et al. CCL2 potentiates inflammation pain and related anxiety-like behavior through NMDA signaling in anterior cingulate cortex. Mol. Neurobiol. 61, 4976–4991 (2024).
Dror, E. et al. Postprandial macrophage-derived IL-1β stimulates insulin, and both synergistically promote glucose disposal and inflammation. Nat. Immunol. 18, 283–292 (2017).
Leidmaa, E., Zimmer, A., Stein, V. & Gellner, A.-K. Acute high-fat high-sugar diet rapidly increases blood-brain barrier permeability in mice. Preprint at bioRxiv https://doi.org/10.1101/2024.04.14.589405 (2024).
Gillespie, K. M. et al. The impact of free and added sugars on cognitive function: a systematic review and meta-analysis. Nutrients 16, 75 (2023).
Fetsko, A. R., Sebo, D. J., Budzynski, L. B., Scharbarth, A. & Taylor, M. R. IL-1β disrupts the initiation of blood-brain barrier development by inhibiting endothelial Wnt/β-catenin signaling. iScience 27, 109651 (2024).
Liu, X. et al. Cell-type-specific interleukin 1 receptor 1 signaling in the brain regulates distinct neuroimmune activities. Immunity 50, 317–333.e316 (2019).
Avital, A. et al. Impaired interleukin-1 signaling is associated with deficits in hippocampal memory processes and neural plasticity. Hippocampus 13, 826–834 (2003).
Drieu, A. et al. Parenchymal border macrophages regulate the flow dynamics of the cerebrospinal fluid. Nature 611, 585–593 (2022).
Kaplan, L., Chow, B. W. & Gu, C. Neuronal regulation of the blood–brain barrier and neurovascular coupling. Nat. Rev. Neurosci. 21, 416–432 (2020).
Somasundaram, V. et al. Inducible nitric oxide synthase-derived extracellular nitric oxide flux regulates proinflammatory responses at the single cell level. Redox Biol. 28, 101354 (2020).
Haruwaka, K. et al. Dual microglia effects on blood brain barrier permeability induced by systemic inflammation. Nat. Commun. 10, 5816 (2019).
Belenichev, I. et al. Modulating nitric oxide: implications for cytotoxicity and cytoprotection. Antioxidants 13, 504 (2024).
Pfau, S. J. et al. Characteristics of blood–brain barrier heterogeneity between brain regions revealed by profiling vascular and perivascular cells. Nat. Neurosci. 27, 1892–1903 (2024).
Ghersi-Egea, J. F. et al. Molecular anatomy and functions of the choroidal blood–cerebrospinal fluid barrier in health and disease. Acta Neuropathol. 135, 337–361 (2018).
Piehl, N. et al. Cerebrospinal fluid immune dysregulation during healthy brain aging and cognitive impairment. Cell 185, 5028–5039.e5013 (2022).
Gate, D. et al. CD4+ T cells contribute to neurodegeneration in Lewy body dementia. Science 374, 868–874 (2021).
He, Y. et al. Interleukin-15 receptor is essential to facilitate GABA transmission and hippocampal-dependent memory. J. Neurosci. 30, 4725–4734 (2010).
Zeisel, A. et al. Molecular architecture of the mouse nervous system. Cell 174, 999–1014.e1022 (2018).
Carloni, S. et al. Identification of a choroid plexus vascular barrier closing during intestinal inflammation. Science 374, 439–448 (2021).
Shipley, F. B. et al. Tracking calcium dynamics and immune surveillance at the choroid plexus blood-cerebrospinal fluid interface. Neuron 108, 623–639.e610 (2020).
Van Hove, H. et al. A single-cell atlas of mouse brain macrophages reveals unique transcriptional identities shaped by ontogeny and tissue environment. Nat. Neurosci. 22, 1021–1035 (2019).
Dani, N. et al. A cellular and spatial map of the choroid plexus across brain ventricles and ages. Cell 184, 3056–3074.e3021 (2021).
Althubaity, N. et al. Choroid plexus enlargement is associated with neuroinflammation and reduction of blood brain barrier permeability in depression. Neuroimage Clin. 33, 102926 (2022).
Baruch, K. et al. CNS-specific immunity at the choroid plexus shifts toward destructive TH2 inflammation in brain aging. Proc. Natl Acad. Sci. USA 110, 2264–2269 (2013).
Kunis, G. et al. IFN-γ-dependent activation of the brain’s choroid plexus for CNS immune surveillance and repair. Brain 136, 3427–3440 (2013).
Kertser, A. et al. Corticosteroid signaling at the brain–immune interface impedes coping with severe psychological stress. Sci. Adv. 5, eaav4111 (2019).
Shimada, A. & Hasegawa-Ishii, S. Increased cytokine expression in the choroid plexus stroma and epithelium in response to endotoxin-induced systemic inflammation in mice. Toxicol. Rep. 8, 520–528 (2021).
Kolabas, Z. I. et al. Distinct molecular profiles of skull bone marrow in health and neurological disorders. Cell 186, 3706–3725.e3729 (2023).
Aspelund, A. et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J. Exp. Med. 212, 991–999 (2015).
Louveau, A. et al. Structural and functional features of central nervous system lymphatic vessels. Nature 523, 337–341 (2015).
Rustenhoven, J. et al. Functional characterization of the dural sinuses as a neuroimmune interface. Cell 184, 1000–1016.e1027 (2021).
Eide, P. K. & Ringstad, G. Cerebrospinal fluid egress to human parasagittal dura and the impact of sleep deprivation. Brain Res. 1772, 147669 (2021).
Ringstad, G. & Eide, P. K. Cerebrospinal fluid tracer efflux to parasagittal dura in humans. Nat. Commun. 11, 354 (2020).
Louveau, A. et al. CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nat. Neurosci. 21, 1380–1391 (2018).
Herisson, F. et al. Direct vascular channels connect skull bone marrow and the brain surface enabling myeloid cell migration. Nat. Neurosci. 21, 1209–1217 (2018).
Cai, R. et al. Panoptic imaging of transparent mice reveals whole-body neuronal projections and skull-meninges connections. Nat. Neurosci. 22, 317–327 (2019).
Brioschi, S. et al. Heterogeneity of meningeal B cells reveals a lymphopoietic niche at the CNS borders. Science 373, eabf9277 (2021).
Cugurra, A. et al. Skull and vertebral bone marrow are myeloid cell reservoirs for the meninges and CNS parenchyma. Science 373, eabf7844 (2021).
Wang, Y. et al. Early developing B cells undergo negative selection by central nervous system-specific antigens in the meninges. Immunity 54, 2784–2794.e2786 (2021).
Mazzitelli, J. A. et al. Cerebrospinal fluid regulates skull bone marrow niches via direct access through dural channels. Nat. Neurosci. 25, 555–560 (2022).
Pulous, F. E. et al. Cerebrospinal fluid can exit into the skull bone marrow and instruct cranial hematopoiesis in mice with bacterial meningitis. Nat. Neurosci. 25, 567–576 (2022).
Zhang, Y. et al. Mucosal-associated invariant T cells restrict reactive oxidative damage and preserve meningeal barrier integrity and cognitive function. Nat. Immunol. 23, 1714–1725 (2022).
Borrell, V. & Marín, O. Meninges control tangential migration of hem-derived Cajal-Retzius cells via CXCL12/CXCR4 signaling. Nat. Neurosci. 9, 1284–1293 (2006).
Siegenthaler, J. A. & Pleasure, S. J. We have got you ‘covered’: how the meninges control brain development. Curr. Opin. Genet. Dev. 21, 249–255 (2011).
Herz, J. et al. GABAergic neuronal IL-4R mediates T cell effect on memory. Neuron 109, 3609–3618.e3609 (2021).
Garofalo, S. et al. Natural killer cells and innate lymphoid cells 1 tune anxiety-like behavior and memory in mice via interferon-γ and acetylcholine. Nat. Commun. 14, 3103 (2023).
Filiano, A. J. et al. Unexpected role of interferon-γ in regulating neuronal connectivity and social behaviour. Nature 535, 425–429 (2016).
Alves de Lima, K. et al. Meningeal γδ T cells regulate anxiety-like behavior via IL-17a signaling in neurons. Nat. Immunol. 21, 1421–1429 (2020).
Ribeiro, M. et al. Meningeal γδ T cell-derived IL-17 controls synaptic plasticity and short-term memory. Sci. Immunol. 4, eaay5199 (2019).
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).
Brombacher, T. M. et al. IL-13-mediated regulation of learning and memory. J. Immunol. 198, 2681–2688 (2017).
Li, S. et al. Interleukin-13 and its receptor are synaptic proteins involved in plasticity and neuroprotection. Nat. Commun. 14, 200 (2023).
Smyth, L. C. D. et al. Identification of direct connections between the dura and the brain. Nature 627, 165–173 (2024).
Morita, S. et al. Heterogeneous vascular permeability and alternative diffusion barrier in sensory circumventricular organs of adult mouse brain. Cell Tissue Res. 363, 497–511 (2016).
Knorr, C. et al. Macrophage-activating lipopeptide-2 (MALP-2) induces a localized inflammatory response in rats resulting in activation of brain sites implicated in fever. Brain Res. 1205, 36–46 (2008).
Wei, S. G. et al. Subfornical organ mediates sympathetic and hemodynamic responses to blood-borne proinflammatory cytokines. Hypertension 62, 118–125 (2013).
Ilanges, A. et al. Brainstem ADCYAP1+ neurons control multiple aspects of sickness behaviour. Nature 609, 761–771 (2022).
Chen, Z. H. et al. Single-cell transcriptomic profiling of the hypothalamic median eminence during aging. J. Genet. Genomics 49, 523–536 (2022).
Takagi, S., Furube, E., Nakano, Y., Morita, M. & Miyata, S. Microglia are continuously activated in the circumventricular organs of mouse brain. J. Neuroimmunol. 331, 74–86 (2019).
Willis, C. L., Garwood, C. J. & Ray, D. E. A size selective vascular barrier in the rat area postrema formed by perivascular macrophages and the extracellular matrix. Neuroscience 150, 498–509 (2007).
Loeffler, C. et al. Immune surveillance of the normal human CNS takes place in dependence of the locoregional blood–brain barrier configuration and is mainly performed by CD3+/CD8+ lymphocytes. Neuropathology 31, 230–238 (2011).
Song, C. et al. Expansion of brain T cells in homeostatic conditions in lymphopenic Rag2−/− mice. Brain Behav. Immun. 57, 161–172 (2016).
Guzik, T. J. et al. Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J. Exp. Med. 204, 2449–2460 (2007).
Marvar, P. J. et al. Central and peripheral mechanisms of T-lymphocyte activation and vascular inflammation produced by angiotensin II-induced hypertension. Circ. Res. 107, 263–270 (2010).
Murtazina, A. & Adameyko, I. The peripheral nervous system. Development 150, dev201164 (2023).
Diogenes, A., Ferraz, C. C., Akopian, A. N., Henry, M. A. & Hargreaves, K. M. LPS sensitizes TRPV1 via activation of TLR4 in trigeminal sensory neurons. J. Dent. Res. 90, 759–764 (2011).
Xu, Z. Z. et al. Inhibition of mechanical allodynia in neuropathic pain by TLR5-mediated A-fiber blockade. Nat. Med. 21, 1326–1331 (2015).
Zanos, T. P. et al. Identification of cytokine-specific sensory neural signals by decoding murine vagus nerve activity. Proc. Natl Acad. Sci. USA 115, E4843–E4852 (2018).
Jin, H., Li, M., Jeong, E., Castro-Martinez, F. & Zuker, C. S. A body–brain circuit that regulates body inflammatory responses. Nature 630, 695–703 (2024).
Needham, B. D. et al. A gut-derived metabolite alters brain activity and anxiety behaviour in mice. Nature 602, 647–653 (2022).
Grabrucker, S. et al. Microbiota from Alzheimer’s patients induce deficits in cognition and hippocampal neurogenesis. Brain 146, 4916–4934 (2023).
Liu, P. et al. Immunoregulatory role of the gut microbiota in inflammatory depression. Nat. Commun. 15, 3003 (2024).
Cox, L. M. et al. γδ T cells suppress microbial metabolites that activate striatal neurons and induce repetitive/compulsive behavior in mice. Brain Behav. Immun. 117, 242–254 (2024).
Zhang, X. et al. Brain control of humoral immune responses amenable to behavioural modulation. Nature 581, 204–208 (2020).
Zhu, Y. et al. A chemogenetic screen reveals that Trpv1-expressing neurons control regulatory T cells in the gut. Science 385, eadk1679 (2024).
Pinho-Ribeiro, F. A. et al. Bacteria hijack a meningeal neuroimmune axis to facilitate brain invasion. Nature 615, 472–481 (2023).
Ono, H. K. et al. Histamine release from intestinal mast cells induced by staphylococcal enterotoxin A (SEA) evokes vomiting reflex in common marmoset. PLoS Pathog. 15, e1007803 (2019).
Shechter, R., London, A. & Schwartz, M. Orchestrated leukocyte recruitment to immune-privileged sites: absolute barriers versus educational gates. Nat. Rev. Immunol. 13, 206–218 (2013).
Betsholtz, C. et al. Advances and controversies in meningeal biology. Nat. Neurosci. 27, 2056–2072 (2024).
Ringstad, G. & Eide, P. K. Molecular trans-dural efflux to skull bone marrow in humans with CSF disorders. Brain 145, 1464–1472 (2022).
Xu, H. et al. The choroid plexus synergizes with immune cells during neuroinflammation. Cell 187, 4946–4963.e4917 (2024).
Ben-Zvi, A. et al. Mfsd2a is critical for the formation and function of the blood–brain barrier. Nature 509, 507–511 (2014).
Derk, J. et al. Formation and function of the meningeal arachnoid barrier around the developing mouse brain. Dev. Cell 58, 635–644.e634 (2023).
Travier, L. et al. Neonatal susceptibility to meningitis results from the immaturity of epithelial barriers and gut microbiota. Cell Rep. 35, 109319 (2021).
Liddelow, S. A. Fluids and barriers of the CNS: a historical viewpoint. Fluids Barriers CNS 8, 2 (2011).
Antila, S. et al. Development and plasticity of meningeal lymphatic vessels. J. Exp. Med. 214, 3645–3667 (2017).
del Rio Serrato, A. et al. Perinatal brain group 3 innate lymphoid cells are involved in the formation of murine dural lymphatics. Preprint at bioRxiv https://doi.org/10.1101/2024.06.12.597323 (2024).
Thion, M. S., Ginhoux, F. & Garel, S. Microglia and early brain development: an intimate journey. Science 362, 185–189 (2018).
Barron, J. J. et al. Group 2 innate lymphoid cells promote inhibitory synapse development and social behavior. Science 386, eadi1025 (2024).
Lenz, K. M. et al. Mast cells in the developing brain determine adult sexual behavior. J. Neurosci. 38, 8044–8059 (2018).
Tanabe, S. & Yamashita, T. B-1a lymphocytes promote oligodendrogenesis during brain development. Nat. Neurosci. 21, 506–516 (2018).
Schepanski, S. et al. Pregnancy-induced maternal microchimerism shapes neurodevelopment and behavior in mice. Nat. Commun. 13, 4571 (2022).
Cansever, D. et al. Lactation-associated macrophages exist in murine mammary tissue and human milk. Nat. Immunol. 24, 1098–1109 (2023).
Viola, M. F. et al. Dedicated macrophages organize and maintain the enteric nervous system. Nature 618, 818–826 (2023).
Stelzer, I. A. et al. Vertically transferred maternal immune cells promote neonatal immunity against early life infections. Nat. Commun. 12, 4706 (2021).
Katsimpardi, L. et al. Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science 344, 630–634 (2014).
Yousefzadeh, M. J. et al. An aged immune system drives senescence and ageing of solid organs. Nature 594, 100–105 (2021).
Yousef, H. et al. Aged blood impairs hippocampal neural precursor activity and activates microglia via brain endothelial cell VCAM1. Nat. Med. 25, 988–1000 (2019).
Das, M. M. et al. Young bone marrow transplantation preserves learning and memory in old mice. Commun. Biol. 2, 73 (2019).
Ruckh, J. M. et al. Rejuvenation of regeneration in the aging central nervous system. Cell Stem Cell 10, 96–103 (2012).
Necula, D., Riviere-Cazaux, C., Shen, Y. & Zhou, M. Insight into the roles of CCR5 in learning and memory in normal and disordered states. Brain Behav. Immun. 92, 1–9 (2021).
Garber, C. et al. T cells promote microglia-mediated synaptic elimination and cognitive dysfunction during recovery from neuropathogenic flaviviruses. Nat. Neurosci. 22, 1276–1288 (2019).
Kaya, T. et al. CD8+ T cells induce interferon-responsive oligodendrocytes and microglia in white matter aging. Nat. Neurosci. 25, 1446–1457 (2022).
Yang, A. C. et al. Physiological blood–brain transport is impaired with age by a shift in transcytosis. Nature 583, 425–430 (2020).
Montagne, A. et al. Blood–brain barrier breakdown in the aging human hippocampus. Neuron 85, 296–302 (2015).
Nation, D. A. et al. Blood–brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat. Med. 25, 270–276 (2019).
Erickson, M. A., Morofuji, Y., Owen, J. B. & Banks, W. A. Rapid transport of CCL11 across the blood–brain barrier: regional variation and importance of blood cells. J. Pharmacol. Exp. Ther. 349, 497–507 (2014).
Fung, I. T. H. et al. Activation of group 2 innate lymphoid cells alleviates aging-associated cognitive decline. J. Exp. Med. 217, e20190915 (2020).
Baruch, K. et al. Aging-induced type I interferon response at the choroid plexus negatively affects brain function. Science 346, 89–93 (2014).
Deczkowska, A. et al. Mef2C restrains microglial inflammatory response and is lost in brain ageing in an IFN-I-dependent manner. Nat. Commun. 8, 717 (2017).
Iram, T. et al. Young CSF restores oligodendrogenesis and memory in aged mice via Fgf17. Nature 605, 509–515 (2022).
Rustenhoven, J. et al. Age-related alterations in meningeal immunity drive impaired CNS lymphatic drainage. J. Exp. Med. 220, e20221929 (2023).
Da Mesquita, S. et al. Aging-associated deficit in CCR7 is linked to worsened glymphatic function, cognition, neuroinflammation, and β-amyloid pathology. Sci. Adv. 7, eabe4601 (2021).
Lee, C. H. et al. Hypothalamic macrophage inducible nitric oxide synthase mediates obesity-associated hypothalamic inflammation. Cell Rep. 25, 934–946.e935 (2018).
Ronan, L. et al. Obesity associated with increased brain age from midlife. Neurobiol. Aging 47, 63–70 (2016).
Kedia, S. et al. T cell-mediated microglial activation triggers myelin pathology in a mouse model of amyloidosis. Nat. Neurosci. 27, 1468–1474 (2024).
Dulken, B. W. et al. Single-cell analysis reveals T cell infiltration in old neurogenic niches. Nature 571, 205–210 (2019).
Escoubas, C. C. et al. Type-I-interferon-responsive microglia shape cortical development and behavior. Cell 187, 1936–1954.e1924 (2024).
Lin, C. J. et al. Mast cell deficiency improves cognition and enhances disease-associated microglia in 5XFAD mice. Cell Rep. 42, 113141 (2023).
Taranov, A. et al. The choroid plexus maintains adult brain ventricles and subventricular zone neuroblast pool, which facilitates poststroke neurogenesis. Proc. Natl Acad. Sci. USA 121, e2400213121 (2024).
Zhu, Y. et al. Opioid-induced fragile-like regulatory T cells contribute to withdrawal. Cell 186, 591–606.e523 (2023).
Steffensen, A. B. et al. Cotransporter-mediated water transport underlying cerebrospinal fluid formation. Nat. Commun. 9, 2167 (2018).
Iliff, J. J. et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci. Transl. Med. 4, 147ra111 (2012).
Mestre, H. et al. Flow of cerebrospinal fluid is driven by arterial pulsations and is reduced in hypertension. Nat. Commun. 9, 4878 (2018).
Iliff, J. J. et al. Cerebral arterial pulsation drives paravascular CSF-interstitial fluid exchange in the murine brain. J. Neurosci. 33, 18190–18199 (2013).
Jiang-Xie, L.-F. et al. Neuronal dynamics direct cerebrospinal fluid perfusion and brain clearance. Nature 627, 157–164 (2024).
Upton, M. L. & Weller, R. O. The morphology of cerebrospinal fluid drainage pathways in human arachnoid granulations. J. Neurosurg. 63, 867–875 (1985).
Ahn, J. H. et al. Meningeal lymphatic vessels at the skull base drain cerebrospinal fluid. Nature 572, 62–66 (2019).
Yoon, J.-H. et al. Nasopharyngeal lymphatic plexus is a hub for cerebrospinal fluid drainage. Nature 625, 768–777 (2024).
Absinta, M. et al. Human and nonhuman primate meninges harbor lymphatic vessels that can be visualized noninvasively by MRI. Elife 6, e29738 (2017).
Choi, B.-R., Johnson, K. R., Maric, D. & McGavern, D. B. Monocyte-derived IL-6 programs microglia to rebuild damaged brain vasculature. Nat. Immunol. 24, 1110–1123 (2023).
Ligocki, A. P. et al. Cerebrospinal fluid flow extends to peripheral nerves further unifying the nervous system. Sci. Adv. 10, eadn3259 (2024).
Acknowledgements
We thank the members of Deczkowska lab for discussions and apologize to colleagues whose work was discussed without citation owing to space constraints. The Deczkowska lab for Brain-Immune Communication is supported by G5 funding from Institut Pasteur, the European Research Council Starting grant (BrainGate), Agence Nationale de la Recherche (ANR-PRC), Ville de Paris EMERGENCE(S) grant, Alzheimer’s Association Research Grant (AARG-22-917964) and Fédération pour la Recherche sur le Cerveau and Don Explore AD (Programme Explore de l’Institut Pasteur). Our lab is part of the DIM C-BRAINS, funded by the Conseil Régional d’Île-de-France. A.D.-B. is supported by Paris Region Fellowship Programme.
Author information
Authors and Affiliations
Contributions
M.K., A.D.-B. and S.A.-M. contributed equally. All the authors contributed to all aspects of the article.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Reviews Immunology thanks Adrian Liston, Maria Rescigno and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Kovacs, M., Dominguez-Belloso, A., Ali-Moussa, S. et al. Immune control of brain physiology. Nat Rev Immunol 25, 515–527 (2025). https://doi.org/10.1038/s41577-025-01129-6
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41577-025-01129-6