<<

Reviews

Human stem models to study host–virus interactions in the central

Oliver Harschnitz 1,2 ✉ and Lorenz Studer 1,2 Abstract | Advancements in human pluripotent technology offer a unique opportunity for the field to study host–virus interactions directly in disease-relevant cells of the human (CNS). Viral encephalitis is most commonly caused by herpesviruses, arboviruses and enteroviruses targeting distinct CNS cell types and often leading to severe neurological damage with poor clinical outcomes. Furthermore, different neurotropic viruses will affect the CNS at distinct developmental stages, from early prenatal development to the aged brain. With the unique flexibility and scalability of human pluripotent stem cell technology, it is now possible to examine the molecular mechanisms underlying acute infection and latency, determine which CNS subpopulations are specifically infected, study temporal aspects of viral susceptibility, perform high-throughput chemical or genetic screens for viral restriction factors and explore complex cell-non-autonomous disease mechanisms. Therefore, human pluripotent stem cell technology has the potential to address key unanswered questions about antiviral immunity in the CNS, including emerging questions on the potential CNS tropism of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).

embryonic stem cells1 Embryonic stem cells The isolation of human and tran- immune system. Animal models can be genetically Pluripotent stem cells scription factor-based of cells altered, and disease can be studied within that are capable of giving into human induced pluripotent stem cells (iPSCs)2 are an entire organism to dissect complex interactions rise to all lineages, but not important breakthroughs in the stem cell field. The between organ systems and diverse cell types. However, to extraembryonic tissues. They are derived from of human pluripotent stem cells there are considerable species-specific differences of the of the (hPSCs; a term referring to both human embryonic stem the immune system that complicate the translation developing . cells and human iPSCs) along the three germ layers ena- of findings in animal models to human disease. General bles the derivation, study and perturbation of potentially differences in metabolism and cell turnover may play a any human . Key applications of hPSCs include role, but species-specific differences of the immune sys- the study of human development3–7, modelling human tem are also profound. Comparing mice and humans, disease8–11 and the development of cell-based we find key differences in many immune components, in regenerative medicine12–18. Past human studies on including in the cellular composition of leukocytes, in neurological and neuroimmunological disease relied signalling components of B cells and T cells, in Toll-like on autopsy or biopsy materials, the isolation of cere- receptors (TLRs), in cytokines and cytokine receptors brospinal fluid or peripheral blood samples. The lack of and in immunoglobulin class switching19,20. In the brain, 1Developmental routine access to primary human central nervous system microglia show species-specific divergence at physio- Program, Sloan Kettering (CNS) cell types has precluded most mechanistic studies, logical levels (for example, in proliferation and in the Institute for Research, prevented the establishment of robust disease expression of sialic acid-binding immunoglobulin-like New York, NY, USA. models and made it impossible to perform high-content lectins (SIGLECs)) and in their neuroinflammatory 2 The Center for Stem Cell chemical or genetic screens directly in human cells. and disease states (for example, responses to trans- Biology, Sloan Kettering Institute for Cancer Research, Therefore, hPSC technology offers a particularly attrac- forming -β (TGFβ) signalling in models 21 New York, NY, USA. tive approach for studying CNS disease and is an ideal of Alzheimer disease) . The use of hPSC-derived cell ✉e-mail: [email protected] tool to probe neuroimmunological disorders. types provides a tractable human in vitro disease plat- https://doi.org/10.1038/ Animal models have yielded important insights form for mechanistic studies, for identifying novel thera- s41577-020-00474-y into the physiology and dysfunction of the mammalian peutic targets and for translating targets into compounds

NATuRe RevIeWS | Immunology volume 21 | July 2021 | 441 Reviews

Induced pluripotent for therapeutic intervention. In addition to capturing ZIKV preferentially targets NPCs and microglia, lead- stem cells the patient’s genetic information via iPSC technology, ing to impaired prenatal brain development resulting (iPSCs). Pluripotent stem powerful tools are used routinely in in microcephaly41,42. In addition to direct CNS tropism, cells derived from terminally hPSCs to genetically correct or induce the expression of viral infections can lead to systemic inflammatory differentiated somatic cells through cellular reprogramming disease-linked . responses that may harm CNS-resident cell types either via the transient expression of To date, hPSC technology has been mostly applied directly or through migration of immune cells into the a defined set of transcription to neurodevelopmental and neurodegenerative disor- CNS43. One such example is the development of post- factors. ders, such as microcephaly22, spinal muscular atrophy23, encephalitic parkinsonism that followed the familial dysautonomia24, amyotrophic lateral sclerosis8, pandemic in 1918. The cause of the resulting - Directed differentiation 9 25 The guided differentiation of Alzheimer disease and Parkinson disease . However, sonism remains unclear, with several potential mech- pluripotent stem cells towards hPSC technology is also increasingly used to study anisms proposed, such as chronic CNS inflammation a desired state autoimmune-induced or virus-induced diseases of the following acute viral infection, invasion of peripheral using and small nervous system26. Such studies have examined how immune cells into the CNS following loss of integrity molecules. autoantibodies target neuronal epitopes leading to of the blood–brain barrier (BBB) or damage due to 44,45 Cerebellar granule cell neuronal dysfunction that closely recapitulates clinical postinfectious autoimmunity . phenotypes and treatment responsiveness27–29. Studies Why certain neuronal or glial cell types show resist- The most numerous neurons on host–virus interactions in hPSC-derived neural lin- ance or susceptibility to a given virus remains largely in the human brain. They are eages include work on herpes simplex virus 1 (HSV-1) unknown. To probe cell type-specific innate immune found in the thick granular layer of the cerebellar cortex. and Zika virus (ZIKV), which affect distinct subpopu- responses, it is imperative to obtain purified neural lations, such as postmitotic neurons versus neural pro- lineages on demand. The derivation of highly enriched Purkinje neurons genitor cells (NPCs), providing important insights into CNS subtypes from hPSCs allows modelling of selec- A class of GABAergic neurons virus-induced CNS pathophysiology30–33. This Review tive vulnerabilities in vitro and the identification of cell located within the Purkinje layer of the cerebellum. They presents an overview of hPSC-based studies of CNS type-specific innate immune mechanisms. Furthermore, arise from progenitors in the host–virus interactions and explores how this technol­ by combining several distinct cell types in co-culture sys- ventricular neuroepithelium ogy can address key outstanding questions in the neuro­ tems, either in two or three dimensions, one can study of the cerebellar primordium immunology field. In the final section of the Review, the role of cell-non-autonomous signalling in anti­viral during embryogenesis. we discuss how such studies may also help to address immunity46. Those may include anti-inflammatory emerging questions concerning CNS involvement and mechanisms during the acute phase47,48, but also pro­ neurological complications in severe acute respiratory inflammatory cascades leading to chronic inflamma- syndrome coronavirus 2 (SARS-CoV-2) infection. tion and long-term neurological deficits43,49–51. Systemic inflammatory signals that negatively impact CNS Using hPSCs to model CNS-resident cell types function, such as virus-induced brain endothelial cell CNS-resident cells show differential susceptibility to inflammation52 or T cell-mediated synaptic pruning43, viral infection. The brain comprises numerous neuronal could also be modelled with hPSC technology by and glial cell types that are essential for CNS function. establishing relevant co-culture systems of neural and The extensive cellular heterogeneity in the CNS entails non-neural cell types. vast heterogeneity in cell type-specific immunologi- cal responses. and microglia play key roles Neural induction and differentiation of hPSCs. in antiviral immunity, and neurons themselves are Sophisticated differentiation strategies have been devel- not mere bystanders. Neurons exhibit innate antiviral oped to yield region-specific neuronal subtypes12,35,53–60, immune responses and produce various interferons and astrocytes61–64 and oligodendrocytes65. The first step of express interferon-stimulated in response to viral in vitro neural differentiation mimics the developmental infection34,35. Selective vulnerability is a frequent, albeit process of neural induction during the gastrula stage66 poorly understood, characteristic of neurological dis- (Fig. 1). Inhibition of morphogenetic (BMP) orders, such as the motor degeneration seen in signalling — and/or activation of growth factor amyotrophic lateral sclerosis or the loss of dopaminergic (FGF) signalling — combined with inhibition sup- neurons in Parkinson disease. Selective vulnerability is presses non-neural, extraembryonic and mesodermal/ similarly seen in viral encephalitis as illustrated for West endodermal fates and enables neural differentiation. Nile virus infection, where autopsy reports describe a This strategy is implemented in a widely used method lack of viral infection in cerebellar granule cell neurons, (‘dual-SMAD inhibition’)54 by concomitant treatment while Purkinje neurons and cortical neurons are highly with the TGFβ inhibitor SB431542 and the BMP inhib- susceptible36. Such differential West Nile virus permis- itor noggin (or LDN193189), a method that forms the sivity in granule cell versus cortical neurons involves basis of many neural differentiation paradigms today. higher basal levels of key interferon-stimulated genes Regional specification similarly mimics in vivo devel- (Ifi27, Irg1 (also known as Acod1) and Rsad2) and a opment where gradients drive patterning in more potent interferon-stimulated response fol- the anterior–posterior (FGFs, WNTs and retinoic acid) lowing interferon-β (IFNβ) stimulus37. In patients and dorsal–ventral (WNTs, BMP and sonic hedgehog with herpes simplex encephalitis, the frontotempo- (SHH)) axis. Exposure to specific combinations of ral regions of the are most commonly morphogens at distinct time points of differentiation affected38,39. By contrast, patients with DBR1 deficiency results in the derivation of diverse neuronal lineages display a brainstem encephalitis, caused by multiple from hPSCs, ranging from forebrain cortical, midbrain neurotropic viruses, including HSV-1 (ref.40). Finally, dopaminergic and hindbrain neurons to spinal cord

442 | July 2021 | volume 21 www.nature.com/nri Reviews

a b Human gastrula, day 16 Trophoblast Forebrain

Amniotic cavity ZIKV HSV-1 Neural Trigeminal stem neurons (S) Astrocytes (S) cells (S) Yolk sac Microglia (S) Astrocytes (R)

Oligo- Neural dendrocyte (R) progenitor Cortical Primitive Neural induction neurons (R) Cortical haematopoeisis (TGFBi and BMPi) cells (S) neurons (R) Microglia (R)

Forebrain

Midbrain Yolk sac Choroid plexus epithelial cells (S) Haemangioblasts Forebrain Neural stem cells (?) Dopaminergic Cortical neurons (R) neurons (S) EMPs

Midbrain SARS-CoV-2 Oligo- Microglia (R) Astrocytes (R) dendrocytes (?) PMACs

Hindbrain Hindbrain Migration DBR1 deficiency leading to hindbrain encephalitis

CNS Multiple neurotropic viruses

Primitive Spinal HSV-1 Norovirus Influenza virus microglia cord Unknown cellular tropism in the hindbrain

Microglia Trigeminal Cortical Neural Oligo- neurons neurons stem cells Astrocytes dendrocytes Microglia

Fig. 1 | In vivo development guides in vitro differentiation principles to obtain disease-relevant cells. a | The major developmental pathways that govern in vitro differentiation strategies for microglia and neuroectodermal cell derivatives. b | Neurotropic viruses may target the central nervous system (CNS) in a region-specific or cell-specific manner. Indicated in the figure are examples of viruses that specifically target the forebrain (namely Zika virus (ZIKV) and herpes simplex virus 1 (HSV-1)), midbrain (severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)) and hindbrain (multiple neurotropic viruses in the context of DBR1 deficiency). Cells known to be susceptible to viral infection are coloured green and annotated with ‘(S)’. Cells annotated with ‘(R)’ are resistant to infection and ‘(?)’ indicates that the susceptibility of the cell population is not clear. BMPi, bone morphogenetic protein inhibition; EMP, erythromyeloid progenitor; PMAC, premacrophage; TGFBi, transforming growth factor-β inhibition.

motor neurons. An alternative strategy involves the a ‘generic’ neuron that does not exist in vivo overexpression of lineage-specific transcription factors rather than a specific neuronal subtype67,68. to coax hPSCs into neural lineages. Such direct lineage conversion paradigms have been used to generate both Modelling primitive in vitro and the neurons and glia63,65,67,68 as well as NPCs for high-content derivation of human microglia. In contrast to neurons, chemical screens or whole-genome genetic screens69. astrocytes and , which are all neuro- The advantages of direct lineage conversion include the ectoderm derivatives, microglia are yolk sac-derived increased speed at which cells acquire terminal fates CNS-resident macrophages that arise during primi- and, in some instances, reduced variability among cells. tive haematopoiesis70. There are two key components However, their specific identity is often unclear, and that determine the functional and molecular differ- for several protocols the resulting neurons may adopt ences among tissue-resident macrophage populations,

NATuRe RevIeWS | Immunology volume 21 | July 2021 | 443 Reviews

a Starting material Healthy Disease hPSCs Introduction of disease Isogenic disease hPSCs variant by gene editing

Corrected iPSCs Genetic correction Patient iPSCs by gene editing

b Monolayer cultures Co-culture systems 3D hPSCs hPSCs hPSCs

Cortical Oligo- Neurons/ Neurons/ Neurons/ Cerebral Regionally neurons Astrocytes dendrocytes Microglia astrocytes astrocytes/microglia microglia patterned organoid

Viral infection Viral infection Viral infection (WT or GFP (WT or GFP (WT or GFP reporter virus) reporter virus) reporter virus)

Viral Viral Viral Viral phenotype Viral phenotype phenotype phenotype phenotype (subtype (regional susceptibility) susceptibility)

• Study viral tropism • Study cell-autonomous signalling pathways • Recapitulate aspects of brain development • Study virus-induced phenotypes (cytotoxicity, • Co-culture systems are highly defined with and structural organization cytokine production) controllable cell ratios • Incorporates heterogeneity in cell types • Study viral susceptibility in patient • Still amenable to high-throughput screens • Study cell-non-autonomous signalling iPSC-derived cells pathways • Perform mechanistic studies of antiviral • Increased maturation of cells signalling pathways c

Genome-wide screens to investigate host factors • Next-generation sequencing Phenotypic • (enrichment screening (FACS of sgRNAs and functional Genetic perturbation Viral assay for GFP+ cells or classification of enriched (CRISPR knockout) (reporter virus) survival of infection) genes) GFP+ GFP Enrichment hPSC-derived CNS cells FACS Rank

High-content chemical screens to identify antiviral compounds Antiviral effect Viral assay High-content Phenotypic (reporter virus) drug screen assays

Caspase 3/ caspase 7 activity assay Activity readout hPSC-derived hPSC-derived CNS cells CNS cells • Validation in multiple Cell viability hPSC-derived CNS cell types assay • Titration of candidate hits for toxicity and optimal efficacy

444 | July 2021 | volume 21 www.nature.com/nri Reviews

◀ Fig. 2 | Applications of human pluripotent stem cells to study host–virus culture models that incorporate those cell types in a interactions. a | Human pluripotent stem cell (hPSC) technology in combination with defined and highly reproducible manner. genome editing tools allows the use of healthy hPSC lines versus those with genetically induced disease alleles, and the use of patient-derived induced pluripotent stem cell Tailoring hPSC-based models for neuroimmunology. (iPSC) lines versus iPSC lines that have been genetically corrected. b | By differentiation of hPSCs into central nervous system (CNS) cells, host–virus interactions can be When hPSC-based disease models are used to study probed in monolayer cultures, co-culture systems or in more complex 3D organoids. host–virus interactions in the CNS, there are several Each of these differentiation paradigms can be applied to patient iPSCs or genetically aspects to consider (Fig. 2). If the virus of interest affects engineered lines. c | Whole-genome genetic screens in hPSC-derived CNS cells a specific neurodevelopmental stage, an hPSC-based can identify host factors that either promote viral replication or identify antiviral model must recapitulate that specific timing of devel- restriction factors in disease relevant cells that can lead to new therapeutic targets opment and include, for example, NPCs, in either 2D or (top). An alternative approach to identify novel therapeutic candidates is the 3D organoid cultures. For modelling region-specific dis- application of high-content chemical screens, which can yield antiviral compounds ease, cells should be directed towards the respective CNS or compounds that ameliorate cytotoxicity (bottom). Cells (or nuclei) in green region. If a specific subpopulation of neurons or glial denote productive infection by the virus. FACS, fluorescence-activated ; cells is affected, for example, cortical neurons, dopamin- sgRNA, single guide RNA; WT, wild type. ergic neurons, astrocytes or microglia, appropriate cell type-specific protocols should be used. Finally, for the including microglia: namely, their ontogeny and their study of cell-non-autonomous mechanisms, co-culture subsequent conditioning by tissue-specific niche models or 3D cerebral organoids are required to examine factors71. Only recently have differentiation protocols cell–cell interactions. Remaining technical challenges been developed to recapitulate the developmental origin include the derivation of fully mature (adult or aged) of microglia. By mimicking primitive streak specification cell types and the need to further reduce variability, (WNT and nodal)72 followed by mesodermal differentia- particularly in 3D organoid models. tion (BMP4 and FGF2), haemogenic endothelial specifi- cation and haematopoietic transition (VEGF, IL-3, IL-6 Limitations of hPSC-based disease modelling. and stem cell factor (SCF)), hPSCs convert sequentially Considerable progress has been made in using hPSC into yolk sac macrophages and microglia-like cells73–77. technology and directed differentiation methods to In monocultures, these hPSC-derived microglia express derive disease-relevant cell types in a reproducible several hallmark microglial markers, such as P2RY12, manner. Despite this, technical challenges remain that GPR34, TMEM119 and IBA1 (albeit at relatively low prevent hPSC-based models from reaching their full levels). However, they lack the required tissue-specific potential. A key limitation is that the current models are conditioning of the CNS, which can be overcome in part more suited towards developmental disorders rather than via co-culture with hPSC-derived neurons or astrocytes, those occurring in the adult or aged CNS. Therefore, incorporation into 3D cerebral organoids or transplan- hPSC-based models are more suitable for studying tation into the mouse CNS78–82. While tissue-specific viruses that target the developing brain, such as ZIKV, conditioning seems to play an important role in micro- rather than infectious diseases that occur at later stages glial differentiation, the specific factors involved in this of life. Attempts to age hPSC-derived cells in vitro have process remain to be determined. been pursued90; however, those strategies may not repli- cate in a physiological manner. Organoid models Increasing complexity by generating hPSC-derived allow extended in vitro periods ranging from months to cerebral organoids. There has been rapid progress in years of culture, which can increase the extent of in vitro the development of hPSC-derived 3D cultures, such maturation and ageing, and the first studies comparing as cerebral spheroids and organoids. Early methods hPSC-derived organoid models with their in vivo coun- were based on promoting self-organization83–85, result- terparts have proven hopeful91. However, such long-term ing in complex neuroepithelial structures, including culture is laborious and costly for routine use, and may cortical-like tissues85. Cerebral organoids can model be difficult to replicate more broadly. While the devel- features of early development, including NPC prolifer- opment of 3D organoid models has provided further ation and differentiation and the establishment of cor- opportunities to study development and disease in the tical layers22. Furthermore, organoids provide a model CNS, these more complex models often show more var- where NPCs, neuronal cells and glial cells interact and iability than their 2D counterparts. It will be essential develop in parallel. In addition to ‘unguided’ cerebral to further optimize these differentiation paradigms to organoids22, region-specific forebrain, midbrain or make them more robust and carefully characterize their hindbrain organoids can be obtained by adding specific cellular composition and development over time. patterning factors86,87. Alternative strategies include the Finally, while the available hPSC-based models are introduction of a polarized patterning centre, resulting ideally suited to study the tropism of neurotropic viruses in multiple forebrain regions88 within the same organoid, and potential virus-induced disease, they fail to address or the fusion of independently patterned organoids to how viruses enter the CNS as they lack a BBB or diffusion study the interaction of distinct brain regions. Finally, kinetics. However, advances have been made by devel- cells from different lineages, such as microglia or endo­ oping hPSC-derived BBB models using microfluidic thelial cells, can be introduced into cerebral organoids technology92, hPSC-derived organoids with a functional to obtain a closer representation of the CNS milieu82. vascular system93 and most recently choroid plexus orga- While some of those lineages can develop spontaneously noids, which established that infection with SARS-CoV-2 in unguided protocols89, it will be important to develop disrupts the blood–cerebrospinal fluid barrier94.

NATuRe RevIeWS | Immunology volume 21 | July 2021 | 445 Reviews

Table 1 | Human stem cell models for CNS viral infections Pathogen Virus family Viral Target cell hPSC-based model Phenotype Refs genome Zika virus Flaviviridae ssRNA NPCs, microglia, Monocultures (NPCs), Impaired organoid growth, premature 32,33,86, macrophages co-cultures (neurons differentiation and apoptosis of NPCs, viral 95,97–102, and microglia), replication in macrophages and microglia 107,109, cortical organoids 132–134 Dengue virus Flaviviridae ssRNA NPC, macrophages Monocultures Proinflammatory cytokine production in 132,133 (NPCs, neurons) NPCs, decreased migration of macrophages West Nile virus Flaviviridae ssRNA NPCs Monocultures Proinflammatory cytokine production and 132 (NPCs, neurons) cell death of infected NPCs Herpes simplex Herpesviridae dsDNA NPCs, cortical Monocultures Cortical neurons and oligodendrocytes 30,31,35, virus 1 neurons, sensory (NPCs, astrocytes, derived from TLR3-and UNC93B-deficient 113,134–137 neurons, astrocytes, neurons, hPSCs show increased susceptibility to oligodendrocytes oligodendrocytes) infection, as is also the case for hPSC-derived cortical neurons deficient in SNORA31, while wild-type trigeminal neurons lack cell-intrinsic viral resistance Varicella zoster Herpesviridae dsDNA NPCs, sensory Monocultures Establishment of viral latency and 111,112,134, virus neurons (NPCs, sensory reactivation of productive infection 138,139 neurons) in sensory neurons Human Herpesviridae dsDNA NPCs Monocultures Cytotoxicity in NPCs, impaired organoid 134,138–141 cytomegalovirus (NPCs) growth and structural development Rabies lyssavirus Rhabdoviridae ssRNA Neurons Monocultures Proinflammatory cytokine production and 142 (neurons) axonal transmission of virus HIV Retroviridae ssRNA Microglia Tricultures Microglia are susceptible to HIV infection 46 (neurons, astrocytes and show inflammatory transcriptional and microglia) profiles and impaired phagocytosis CNS, central nervous system; dsDNA, double-stranded DNA; hPSC, human pluripotent stem cell; NPC, neural ; ssRNA, single-stranded RNA; TLR3, Toll-like 3.

Using hPSCs to study CNS infections that ZIKV preferentially infected NPCs, causing cell Viral infections of the CNS. Viral encephalitis is most cycle dysregulation and cell death32,33,95. As a control, commonly caused by herpesviruses, arboviruses and were exposed to dengue virus serotype 2, enteroviruses38 and often triggers severe neurological a flavivirus with genetic similarities to ZIKV, which did damage. The factors that determine susceptibility to not trigger cell death or abrogate NPC growth33. Qian and viral encephalitis remain poorly understood. While colleagues examined the neurotropism of ZIKV in more age-related decline in innate and adaptive immunity detail using a platform to establish forebrain-, midbrain- can cause increased susceptibility in older individuals and hypothalamic-specific cerebral organoids86. By (age >60 years), inborn errors of immunity that impair deriving well-defined forebrain organoids which mimic cell-intrinsic antiviral mechanisms can predispose chil- the multilayer organization seen in human CNS devel- dren to viral encephalitis. Furthermore, some viruses opment, they tested ZIKV infection at multiple develop- induce disease at specific developmental stages, such as mental stages in vitro. While NPCs could be infected at in the case of ZIKV infection, a disease process that has all stages of in vitro differentiation, only early-stage fore- been studied in particular detail using hPSC-based mod- brain organoids, containing a high proportion of + els (see Table 1 for an overview of hPSC-based disease NPCs as opposed to TBR2+ progenitor cells or CTIP2+ modelling of host–virus interactions). neurons, were vulnerable to ZIKV infection, resulting in cell death and decreased proliferation. These findings hPSC-based models for ZIKV infection. ZIKV is an arbo- indicate a narrow temporal window during which the virus belonging to the genus Flavivirus discovered in is at risk. An additional study using both mouse Uganda in 1947. Transfer of ZIKV to humans via Aedes pluripotent stem cell-based and hPSC-based models, aegypti mosquitoes was first reported in 2007. Infection described similar results and reported ZIKV-induced with ZIKV in adults typically leads to mild symptoms. intrauterine growth retardation and in However, concurrent neurological manifestations, mice (however, this was mouse strain dependent). including microcephaly in newborns and Guillain– In hPSC-derived cerebral organoids, the study authors Barré syndrome in adults, have caused major concerns. observed ZIKV-mediated cell death due to apoptosis hPSC technology has played a key role in determining and autophagy, and proposed that increased suscep- the tropism of ZIKV for different CNS cell types and in tibility in NPCs could be attributed to their higher addressing the molecular mechanisms underlying the expression of AXL, a candidate receptor for ZIKV devastating microcephaly. Two early landmark studies infection32,96. Nevertheless, genetic ablation of AXL in using 2D monolayer cultures of hPSC-derived NPCs and hPSC-derived NPCs and cerebral organoids did not pro- neurons95 or 3D neurospheres32,33, respectively, showed tect from ZIKV infection or ZIKV-induced cell death97.

446 | July 2021 | volume 21 www.nature.com/nri Reviews

TYRO3, an alternative candidate viral receptor, was also in viral entry, replication, packaging and release, and are highly expressed in both NPCs and cerebral organoids, performed directly in disease-relevant hPSC-derived cell and could play a role in ZIKV entry97. Alternatively, types. Li et al. used this strategy in hPSC-derived NPCs AXL and TYRO3 could serve as co-receptors in NPCs. to identify molecular targets for potential therapeutic intervention107. They made several interesting observa- ZIKV disrupts NPC proliferation through distinct mecha- tions; firstly, genes encoding putative ZIKV receptors nisms. While multiple studies reported effects of ZIKV on (AXL and TYRO3) identified in previous studies in NPCs32,33,86,95, the molecular mechanisms underlying this immortalized cell lines were not among the positive hits, phenotype remained elusive. With use of a combination highlighting the importance of using disease-relevant of mouse models and hPSC-derived forebrain organoids, cells. Among the positive hits were genes involved in NS2A, one of the seven non-structural ZIKV , heparan sulfation, endocytosis, endoplasmic reticulum was shown to interact with and destabilize adherens processing, Golgi apparatus function and interferon junctions, resulting in aberrant radial glial organization98. activity. These potentially relate to different stages of Importantly, the interaction between ZIKV NS2A and the viral cycle, namely viral entry (heparan sulfation, adherens junction components impairs NPC prolifera- endocytosis), translation and replication of the viral tion and induces premature differentiation. An earlier genome (endoplasmic reticulum), egress from the host study using fetal neural stem cells demonstrated inhibi- (Golgi apparatus) and host antiviral response (inter- tion of the AKT–mechanistic target of rapamycin signal- feron activity). Hit validation was performed with use ling pathway by two other ZIKV non-structural proteins, of single-gene knockouts and pharmacological inter- NS4A and NS4B, leading to reduced and ventions. A second whole-genome screen was based on increased autophagy99. Additional evidence for ZIKV NPCs derived via direct lineage conversion of hPSCs, cytotoxicity in NPCs was presented via the heterodimeric and confirmed several hits and pathways involved (hep- protease of two ZIKV proteins, NS2B and NS3, interact- aran sulfation, endocytosis, and Golgi apparatus func- ing with the host protein septin 2 and leading to defects tion)69. Again, no evidence was found that either AXL in cytokinesis and cell death100. or TYRO3 plays a role in ZIKV infection in NPCs. Two other studies identified promising therapeutic candi- The role of non-neural cells in ZIKV infection. The role dates by performing high-content chemical screens of of during ZIKV infection has been addressed in FDA-approved compounds108,109. Such drug-repurposing hPSC-derived astrocytes and microglia-like cells101,102. screens are designed to rapidly identify compounds of Muffat and colleagues showed divergent innate immune direct therapeutic relevance. The first study screened responses in NPCs versus astrocytes or microglia. In line more than 6,000 compounds in hPSC-derived astro- with previous studies, NPCs were highly susceptible to cytes and NPCs to either inhibit ZIKV infection or ZIKV infection, resulting in significant cell death102. prevent subsequent cell death. Combining compounds By contrast, astrocytes and microglia did not show signs of from both groups improved response and was validated cell death but rather served as reservoirs for productive in hPSC-derived cerebral organoids. In the second viral replication101,102. The upregulation of candidate viral study, we performed a screen of more than 1,000 FDA- entry receptors on microglia (AXL, TYRO3 and TLR7) approved drugs in hPSC-derived NPCs109. We validated and proinflammatory markers (IL-6, IL-1β and CCR5) two positive hits not only in hPSC-derived forebrain was ZIKV strain dependent101, potentially explaining dis- organoids in vitro but also in vivo in the adult mouse parate effects of African and Asian ZIKV strains on CNS brain, and established that the key hit (hippeastrine phenotypes. Transfer of infected microglia in co-culture hydro­bromide) is not only prophylactic in preventing induced NPC infection and death101. These results led infection but can exert strong therapeutic anti-ZIKV to the hypothesis that microglia may be a ZIKV vector activity several days after initial infection. enabling crossing of the placental–fetal barrier, as they originate in the yolk sac near the maternal vasculature Modelling viral latency and reactivation before migrating into the developing CNS. This is in An area of where hPSC models could have agreement with data showing ZIKV infecting and rep- a major impact is the study of viral latency and reac- licating in Hofbauer cells103, a type of human placental tivation. After an initial productive or lytic phase of macrophage, and with a study in mice describing a very infection, some viruses undergo latency, whereby the full narrow time window when intrauterine ZIKV injection viral genome is retained in the host cell but with only triggers fetal brain infection104 matching the exact time few viral latency genes expressed and without produc- point when microglial progenitors enter the CNS104,105. tion of infectious progeny. Latency can persist despite Alternatively, as fetal macrophages as a whole are suscep- host immune responses and is reversible; thus, the virus tible to ZIKV infection, peripheral blood macrophages can be reactivated, after which a new phase of productive could also serve as a Trojan horse for ZIKV infection infection occurs. While latency is mostly established in into the developing brain by crossing the BBB42. cells of the peripheral nervous system (PNS), it is impor- tant to CNS disease as reactivation of a latent infection Chemical and genetic screens for ZIKV. hPSC models may cause spread to the CNS35,110. are ideally positioned for high-content chemical or Herpesviruses can initiate both a lytic infection and a genetic screens to identify therapeutic targets of ZIKV latent infection, with alphaherpesviruses, such as HSV-1 infection or other viral infections106. Whole-genome and varicella zoster virus, establishing latency specifi- CRISPR–Cas screens can identify host factors involved cally in the nervous system. The molecular mechanisms

NATuRe RevIeWS | Immunology volume 21 | July 2021 | 447 Reviews

Healthy control TLR3–/– patient iPSCs TLR3–/– isogenic hPSCs

hPSCs hPSCs

Trigeminal Cortical Neural Oligo- Trigeminal Cortical Neural Oligo- neurons neurons progenitor cells Astrocytes dendrocytes neurons neurons progenitor cells Astrocytes dendrocytes

Viral infection Viral infection (WT or GFP (WT or GFP reporter virus) reporter virus)

Viral phenotype Viral phenotype Viral phenotype

• Cell type-specific cell-intrinsic immunity • Cortical neurons and oligodendrocytes deficient in TLR3 (or other • Cortical neuron and innate antiviral immunity is factors in the TLR–interferon signalling pathway) lose HSV-1 cell- TLR3–interferon dependent intrinsic immunity • WT trigeminal neurons are highly susceptible to HSV-1 infection, • Deficiencies in SNORA31 and DBR1, as reported in patients with closely mimicking the trigeminal ganglion as a reservoir of latent HSE, also lead to HSV-1 susceptibility in hPSC-derived cortical infection neurons, despite intact TLR3–interferon signalling pathways

Fig. 3 | CNS immunity to HSV-1 is due to a cell-intrinsic, cell type-dependent TLR3–interferon response. Studies using human pluripotent stem cells (hPSCs) have dissected the differences in cell-intrinsic antiviral immunity to herpes simplex virus 1 (HSV-1) between various central nervous system (CNS) cell types. In healthy controls, hPSC-derived cortical neurons and oligodendrocytes show cell-intrinsic antiviral immunity to HSV-1, which is dependent on intact Toll-like receptor 3 (TLR3)–interferon signalling. This cell-intrinsic immunity is lost in patients who have deficiencies in the TLR3–interferon signalling pathway (due to genetic variants in TLR3, UNC93B or STAT1 for example) or in TLR3−/− isogenic control cells. hPSC-derived trigeminal neurons lack TLR3–interferon-dependent anti-HSV-1 immunity and show productive viral infection also in wild-type (WT) cells, closely mimicking the in vivo situation where trigeminal neurons are the reservoir for latent HSV-1 infection even in healthy individuals. Cells coloured green are productively infected by the virus. HSE, herpes simplex encephalitis.

of latency are poorly understood, with a lack of robust on the selective vulnerability of distinct neuronal sub- experimental models. While most in vitro models reca- types in the PNS, on whether all latent viral genomes pitulate only the lytic stage of infection, animal models are competent to reactivate and on whether a reactivated are generally inefficient, expensive and time-consuming virus can re-establish latency110. Future studies using to study this phenomenon as latency is often established human stem cell models may address some of those in very few cells and over protracted periods, making it key questions and facilitate the development of novel difficult to acquire sufficient material to study viral gene therapeutic strategies to prevent viral reactivation. expression. Furthermore, animal models lack the flex- ibility and scalability of hPSC-based models to enable Modelling primary immunodeficiencies. To date, there whole-genome or chemical screens to determine host are at least 430 inborn errors of immunity (also known factors and therapeutic targets in studying viral latency. as primary immunodeficiencies)114. Two known clini- With advances in hPSC-based disease modelling, cal entities where a genetic cause can enable CNS viral studies have aimed to recapitulate viral latency and reac- infection are herpes simplex encephalitis affecting the tivation of HSV-1 and varicella zoster virus in vitro in forebrain, caused by deficiencies in TLR3, UNC93B human sensory neurons111–113. Some key criteria of viral (also known as UNC93B1), TRAF3, TRIF (also known latency could be established in vitro in hPSC-derived as TICAM1), TBK1 and IRF3 (ref.39), and viral enceph- neurons: viral gene expression restricted to only a few alitis of the brainstem due to DBR1 deficiency, which key latency genes, viral genomes being episomal and can be caused by HSV-1, influenza virus or norovirus40. non-replicative, and lack of infectious viral particle Human iPSC technology has offered an ideal plat- production. A key aspect in those studies was the main- form to study primary CNS immunodeficiencies. An tenance of viral latency and the reactivation of latent early example of iPSC-based disease modelling for infection. Reactivation of latent alphaherpesviruses can an inborn error of immunity affecting the human nerv- be debilitating and painful in the PNS, and can lead to ous system was the study of anti-HSV-1 immunity using life-threatening encephalitis on spread to the CNS. Many multiple hPSC-derived CNS cells30 (Fig. 3). In this study, questions remain on how viral latency is established, we reported that cell-intrinsic immunity for HSV-1

448 | July 2021 | volume 21 www.nature.com/nri Reviews

infection in the CNS relies on an intact TLR3–interferon a powerful model to study molecular mechanisms of signalling pathway and is cell type dependent (see Box 1 viral latency, as human primary trigeminal neurons are for details on cell-intrinsic antiviral signalling). Viral extremely difficult to isolate. Furthermore, trigeminal susceptibility and interferon production were studied neurons from human cadavers have the caveat that they in hPSC-derived, purified neural stem cells, cortical may have previously undergone HSV-1 infection and neurons, astrocytes and oligodendrocyte progenitor thus harbour latent viral infection. cells. Intriguingly, while cortical neurons and oligoden­ In a recent study on the genetic susceptibility under- drocyte progenitor cells derived from healthy indi- lying herpes simplex encephalitis, hPSC-derived corti- viduals could restrict viral replication of HSV-1, the cal neurons were used to establish a role for SNORA31, corresponding cell types derived from TLR3- and a small nucleolar RNA of the H/ACA class, as a CNS UNC93B-deficient cells could not. This viral pheno- cell-intrinsic viral restriction factor31. Whole-exome type was explained by a lack of IFN-β and IFN-λ pro- sequencing identified five individuals susceptible to duction in response to poly(I:C) (a TLR3 agonist) or herpes simplex encephalitis carrying heterozygous rare HSV-1. By contrast, no such differences were seen in variants in SNORA31. Using cortical neurons from both neural stem cells or astrocytes from either healthy or patient iPSCs and hPSCs with CRISP–Cas9-induced TLR3- and UNC93B-deficient cells. Furthermore, inter- biallelic and monoallelic mutations, we established feron production varied among neural stem cells, corti- SNORA31 as essential for anti-HSV-1 immunity. Both cal neurons, astrocytes and oligodendrocyte progenitor patient and gene-edited lines for SNORA31 showed cells following poly(I:C) stimulation. These experiments intact TLR3–interferon signalling and normal responses elegantly demonstrated that anti-HSV-1 immunity to TLR3 and IFN-α/β stimulation. However, transcrip- of the CNS is due to a cell-intrinsic, TLR3–interferon tomic analysis in hPSC-derived cortical neurons showed response in cortical neurons and oligodendrocyte an impaired response to HSV-1 infection. Hence, while progenitor cells. SNORA31 is a viral restriction factor for HSV-1, it A second study describes a directed differentia- does so independently of the known TLR3–interferon tion paradigm to obtain human trigeminal neurons signalling pathways. from hPSCs35. Trigeminal neurons, which are part of the PNS and developmentally originate from cranial Models for SARS-CoV-2 infection placodes, play a key role in CNS infection as a site of The coronavirus disease 2019 (COVID-19) pandemic entry and viral latency for HSV-1. Intriguingly, we and the broad tropism of SARS-CoV-2 for multiple reported distinct roles for trigeminal versus cortical organ systems and cell types highlights the importance neurons in HSV-1 host defence. While we confirmed of using disease-relevant models that can capture the cel- a TLR3–interferon-dependent cell-intrinsic immunity lular diversity of the , such as hPSC-based to HSV-1 in cortical neurons, trigeminal neurons were models. The importance of using human in vitro mod- highly susceptible to HSV-1 independently of whether els is underscored by species barriers that can hinder they were derived from TLR3-deficient patients or from translation of findings in primary cell lines to the clinic, healthy individuals. This result mimics the viral life even leading to false positive results that can waste pre- cycle of HSV-1, where the trigeminal ganglion becomes cious time and resources that are of importance during infected and a source for viral latency even in healthy a pandemic when a new infectious disease emerges115–117. individuals. Such hPSC-based platforms may provide Although COVID-19 initially presented as a mostly acute respiratory disorder, it has become clear that that there are many extrapulmonary and even chronic Box 1 | Cell-intrinsic innate antiviral signalling manifestations118. These extrapulmonary manifestations A range of neurotropic viruses, including Zika virus, herpes simplex virus 1 (HSV-1), include myocarditis, acute kidney injury, hepatocellular dengue virus, West Nile virus and varicella zoster virus, are capable of entering and injury, gastrointestinal disease and increasing evidence infecting the central nervous system. To protect the host from viral infection, the for both PNS and CNS pathology118,119. There has been innate immune system consists of four main classes of pattern recognition receptors a rapid response from the stem cell field in engineer- (PRRs) that detect pathogen-associated molecular patterns143. The four classes of ing both 2D models and 3D organoids to study the tro- PRRs are Toll-like receptors (TLRs), retinoic acid-inducible gene 1 protein (RIG1) like pism of SARS-CoV-2 in human gut enterocytes120 and receptors, nucleotide-binding oligomerization domain (NOD)-like receptors and intestinal organoids121,122, cardiomyocytes123–125, kidney cytosolic DNA sensors (such as cyclic GMP–AMP synthase). Signalling through these organoids126, organoids124, pancreatic endocrine PRRs initiates antiviral immune pathways by activating the transcription factors cells124 and, of course, lung organoids127. nuclear factor-κB and interferon regulatory factors, leading to direct intrinsic antiviral immunity. Different subclasses of PRRs activate distinct innate immune signalling In an early, modular hPSC-based platform to study pathways; for example, detection of pathogen-associated molecular patterns by SARS-CoV-2 tropism, we identified hPSC-derived TLRs, such as HSV-1 by TLR3, leads to activation of myeloid differentiation primary dopaminergic neurons to be susceptible to SARS-CoV-2 response protein MYD88-dependent and Toll/IL-1 receptor domain-containing infection, while hPSC-derived cortical neurons and adaptor inducing interferon-β (TRIF)-dependent signalling pathways and subsequent microglia are resistant124. A second study that used both production of antiviral cytokines and type I interferons. The specificity of PRRs for a hPSC-derived monolayer cultures (neurons, astrocytes virus can be cell type specific. An example is that of human cortical neurons, which are and microglia) and region-specific organoids (cortical, dependent on TLR3–interferon signalling for their cell-intrinsic anti-HSV-1 immunity. hippocampal, hypothalamic, midbrain and choroid By contrast, TLR3 is redundant for anti-HSV-1 immunity in haematopoietic cell types, plexus organoids) found that in all these human model explaining why patients with TLR3 deficiencies have infections limited to their central systems, only choroid plexus organoids showed signifi­cant nervous system without dissemination to leukocytes or macrophages144. SARS-CoV-2 infection and replication (10–20%), leading

NATuRe RevIeWS | Immunology volume 21 | July 2021 | 449 Reviews

to inflammatory cellular responses and cell death128. study both sporadic viral infections and primary CNS These findings were corroborated by Pellegrini et al., immunodeficiencies in disease-relevant human cells. who used hPSC-derived cortical organoids and choroid While 3D organoid models have shown considerable plexus organoids to study the tropism of SARS-CoV-2, promise for the study of host–virus interactions, further and similarly found that while cortical neurons and progress is required to achieve models that more closely astrocytes are relatively resistant to infection, there was represent their in vivo equivalent and show reduced productive infection in choroid plexus organoids94. These vari­ability. Strategies towards this goal include the use of two studies indicate that the choroid plexus could poten- specialized spinning bioreactors86 or the introduction tially serve as an entry site for SARS-CoV-2 into the CNS, of signalling centres into organoids to ensure spatial as productive infection in choroid plexus epithelial cells, topography88. The fusion of multiple organoids into a due to virus circulating in nearby capillaries, could lead single assembloid could represent a model to recreate the to viral spread into the cerebrospinal fluid. physiological path of infection for viruses such as HSV-1. However, conflicting data report a direct neuro­ Other ways to increase complexity include the addition tropism of SARS-CoV-2 with use of hPSC-derived orga- of multiple cell types in 2D co-culture systems or by noids, a mouse model overexpressing human ACE2, and use of microfluidic devices. Most CNS models do not COVID-19 patient brain autopsy material. The study include several non-neural cell types, such as endothe- authors present evidence for the neuroinvasive capac- lial cells, pericytes or microglia. While such lineages can ity of SARS-CoV-2 in neural stem cells and cortical develop spontaneously in unguided protocols89, it will be neurons129, findings in direct contrast to those of previ- important to develop culture models that incorporate ous reports. While the brain autopsy reports showed evi- those cell types in a defined manner. dence of neuroinvasion by staining for anti-SARS-CoV-2 The combination of hPSC technology with genetic spike protein129, there was a striking lack of immune cell or chemical screens is a particularly attractive strategy infiltration around infected sites. Furthermore, the asso- to identify genetic host factors that drive viral suscepti- ciation of SARS-CoV-2 infection in this study with sites bility and to nominate candidate therapeutic targets for of ischaemic infarcts that the patients had experienced drug design or clinical treatment. Future screens may may point to an alternative route for viral invasion into harness the potential of automated imaging technol­ the CNS. ogies to apply such screens in more complex co-culture It was perhaps thought that the extrapulmonary systems or 3D organoid models. With the incorpora- manifestations would occur mostly in patients with tion of multiple CNS cell types in a single model, it severe COVID-19. This, however, does not seem to be will be intriguing to assess cell type-specific responses, the case, as is illustrated by the occurrence of myocar- and cell–cell interactions following a viral stimu- ditis in relatively young and often otherwise asymp- lus. One method to detect heterogeneity in pathogen tomatic patients130. Whether this is also true for the responses is single-cell RNA sequencing (scRNA-seq). neurological manifestations of COVID-19 remains to Studies using scRNA-seq should facilitate the dissec- be seen, as the true incidence is unknown. This is fur- tion of cell type-specific innate immune responses (for ther complicated by the fact that neurological damage example, neuronal versus microglial responses) and may not become apparent until much later, leading to identify differences in cellular states within a popu­ postinfectious disease such as postencephalitic parkin- lation (for example, infected cells versus bystander cells sonism, which was described following the 1918 influ- and inflammatory states). Furthermore, scRNA-seq enza epidemic44,45. There are still many open questions may offer unique insights into activation states after on how SARS-CoV-2 can affect the human nervous infection and help to unravel the mechanisms under- system, such as which subpopulations of neurons and lying long-term neurological deficits following viral non-neuronal cells are susceptible to SARS-CoV-2 infec­ encephalitis. By combining CRISPR–Cas-based gene tion, which clinical phenotypes are a direct result of editing with scRNA-seq (through a technique known infection in the CNS versus secondary to SARS-CoV-2- as Perturb-seq), it is now possible to assay single-cell mediated vascular damage or cytokine release, and transcriptional profiles linked to a specific genotype whether CNS involvement could lead to long-term and phenotype within a single experiment131. Such neurological damage or increased susceptibility to techniques could probe genetic regulators of an anti- neuro­degeneration. Studies with carefully character- viral response in human disease-relevant cells, while ized hPSC-derived CNS cell types are needed to resolve simultaneously obtaining transcriptional data on the questions about the precise tropism of SARS-CoV-2 in gene regu­latory pathways involved during immune the brain and to establish model systems that allow the activation. This may lead to the rapid identifica- study of SARS-CoV-2-mediated long-term effects. tion of important host factors that regulate antiviral immunity and the corresponding regulatory pathways Conclusions involved. To date, hPSC-based models have successfully been Finally, in a similar way that the stem cell field reacted used to study viral infections of the CNS in NPCs, to the ZIKV health crisis, we currently have an oppor- neurons, astrocytes, oligodendrocytes and microglia. tunity to harness the power of hPSC-based models to Additionally, differentiation paradigms exist to derive address the COVID-19 pandemic. There are many open cells of the PNS, such as trigeminal neurons, that play questions that hPSC-based models can help to address, a role in viral latency and CNS entry. More broadly, such as the exact neurotropism of SARS-CoV-2, the hPSC technology has enabled neuroimmunologists to molecular mechanisms underlying a differential viral

450 | July 2021 | volume 21 www.nature.com/nri Reviews

susceptibility (for instance, expression of specific viral We have described several remaining hurdles to be co-receptors by dopaminergic neurons versus cortical overcome for hPSC-based models to reach their full neurons), whether SARS-CoV-2 infection may lead to potential in neuroimmunology. However, hPSC-based long-term neurological damage and the identification models have already proven to be invaluable tools for of therapeutic targets to treat neuroinflammation in immunologists in a spectacularly short time, both in patients with COVID-19. The first studies performing the case of sporadic viral infections of the CNS and for high-content chemical screens to identify therapeutic inborn errors of immunity leading to viral encephalitis. compounds that may alleviate SARS-CoV-2-induced Human stem cell technology will provide an essential disease using human cells from hPSC-derived or pri- addition to the scientific toolbox of neuroimmunologists mary organoids have been published122,127, and it will be and help to translate findings to patients in need. important to see whether hits identified in these studies can be translated to a clinical setting. Published online 4 January 2021

1. Thomson, J. A. lines derived from 20. Sinmaz, N., Nguyen, T., Tea, F., Dale, R. C. & Brilot, F. 37. Cho, H. et al. Differential innate immune response human . 282, 1145–1147 (1998). Mapping autoantigen epitopes: molecular insights programs in neuronal subtypes determine susceptibility This study describes the first example of deriving into autoantibody-associated disorders of the nervous to infection in the brain by positive-stranded RNA human embryonic stem cell lines from blastocysts. system. J. Neuroinflammation 13, 219 (2016). viruses. Nat. Med. 19, 458–464 (2013). 2. Takahashi, K. et al. Induction of pluripotent stem 21. Smith, A. M. & Dragunow, M. The human side of 38. Tyler, K. L. Acute viral encephalitis. N. Engl. J. Med. cells from adult human by defined factors. microglia. Trends Neurosci. 37, 125–135 (2014). 379, 557–566 (2018). Cell 131, 861–872 (2007). 22. Lancaster, M. A. et al. Cerebral organoids model 39. Jouanguy, E. et al. Human inborn errors of immunity This is first example of cellular reprogramming human brain development and microcephaly. Nature to herpes viruses. Curr. Opin. Immunol. 62, 106–122 leading to the generation of human iPSCs from 501, 373–379 (2014). (2020). terminally differentiated, somatic cells. This is one of the early reports of hPSC-derived 40. Zhang, S.-Y. et al. Inborn errors of RNA lariat 3. Nicholas, C. R. et al. Functional maturation of cerebral organoids and their application for the metabolism in humans with brainstem viral infection. hPSC-derived forebrain interneurons requires disease modelling of microcephaly. Cell 172, 952–965.e18 (2018). an extended timeline and mimics human neural 23. Ebert, A. D. et al. Induced pluripotent stem cells 41. Qian, X., Nguyen, H. N., Jacob, F., Song, H. & development. Cell Stem Cell 12, 573–586 (2013). from a spinal muscular atrophy patient. Nature 457, Ming, G.-L. Using brain organoids to understand 4. Gafni, O. et al. Derivation of novel human ground 277–280 (2009). Zika virus-induced microcephaly. Development 144, state naive pluripotent stem cells. Nature 504, 24. Zeltner, N. et al. Capturing the biology of 952–957 (2017). 282–286 (2013). disease severity in a PSC-based model of familial 42. Lum, F.-M. et al. Zika virus infects human fetal brain 5. Deglincerti, A. et al. Self-organization of the in vitro dysautonomia. Nat. Med. 22, 1421–1427 (2016). microglia and induces inflammation. Clin. Infect. Dis. attached human embryo. Nature 533, 251–254 25. Soldner, F. et al. Generation of isogenic pluripotent 64, 914–920 (2017). (2016). stem cells differing exclusively at two early onset 43. Di Liberto, G. et al. Neurons under T cell attack 6. Martyn, I., Kanno, T. Y., Ruzo, A., Siggia, E. D. & Parkinson point mutations. Cell 146, 318–331 coordinate phagocyte-mediated synaptic stripping. Brivanlou, A. H. Self-organization of a human (2011). Cell 175, 458–471.e19 (2018). organizer by combined Wnt and Nodal signalling. 26. Harschnitz, O. Human stem cell–derived models: 44. Dale, R. C. et al. Encephalitis lethargica syndrome: Nature 558, 132–135 (2018). lessons for autoimmune diseases of the nervous 20 new cases and evidence of basal ganglia 7. Karzbrun, E., Kshirsagar, A., Cohen, S. R., Hanna, J. H. system. Neuroscientist 25, 199–207 (2018). autoimmunity. Brain 127, 21–33 (2004). & Reiner, O. Human brain organoids on a chip reveal 27. Steinbeck, J. A. et al. Functional connectivity 45. Hoffman, L. A. & Vilensky, J. A. Encephalitis the physics of folding. Nat. Phys. 14, 515–522 under optogenetic control allows modeling of human lethargica: 100 years after the epidemic. Brain (2018). neuromuscular disease. Cell Stem Cell 18, 134–143 140, 2246–2251 (2017). 8. Dimos, J. T. et al. Induced pluripotent stem cells (2015). 46. Ryan, S. K. et al. Neuroinflammation and EIF2 generated from patients with ALS can be 28. Harschnitz, O. & van den Berg, L. H. Autoantibody signaling persist despite antiretroviral treatment differentiated into motor neurons. Science 321, pathogenicity in a multifocal motor neuropathy in an hiPSC tri-culture model of HIV infection. 1218–1221 (2008). induced pluripotent stem cell–derived model. Stem Cell Rep. 14, 703–716 (2020). This study provides an early example of iPSC-based Ann. Neurol. 80, 71–88 (2016). 47. Reinert, L. S. et al. Sensing of HSV-1 by the cGAS-STING disease modelling of neurological disease using 29. Clark, A. J., Kaller, M. S., Galino, J., Willison, H. J., pathway in microglia orchestrates antiviral defence patient-derived cells. Rinaldi, S. & Bennett, D. L. H. Co-cultures with stem in the CNS. Nat. Commun. 7, 13348 (2016). 9. , M. A. et al. Probing sporadic and familial cell-derived human sensory neurons reveal regulators 48. Chhatbar, C. et al. Type I interferon receptor signaling Alzheimer’s disease using induced pluripotent stem of peripheral myelination. Brain 140, 898–913 of neurons and astrocytes regulates microglia activation cells. Nature 482, 216–220 (2012). (2017). during viral encephalitis. Cell Rep. 25, 118–129.e4 10. Soldner, F. et al. Parkinson-associated risk variant in 30. Lafaille, F. G. et al. Impaired intrinsic immunity to (2018). distal enhancer of α-synuclein modulates target gene HSV-1 in human iPSC-derived TLR3-deficient CNS 49. Vasek, M. J. et al. A complement-microglial axis expression. Nature 533, 95–99 (2016). cells. Nature 491, 769–773 (2012). drives synapse loss during virus-induced memory 11. Paquet, D. et al. Efficient introduction of specific 31. Lafaille, F. G. et al. Human SNORA31 variations impairment. Nature 534, 538–543 (2016). homozygous and heterozygous mutations using impair cortical neuron-intrinsic immunity to HSV-1 50. Bialas, A. R. et al. Microglia-dependent synapse CRISPR/Cas9. Nature 533, 125–129 (2016). and underlie herpes simplex encephalitis. Nat. Med. loss in type I interferon-mediated lupus. Nature 546, 12. Kriks, S. et al. Dopamine neurons derived from 25, 1873–1884 (2019). 539–543 (2017). human ES cells efficiently engraft in animal models Lafaille et al. (2012) and Lafaille et al. (2019) 51. Garber, C. et al. T cells promote microglia-mediated of Parkinson’s disease. Nature 480, 547–551 describe examples of hPSC-based disease synaptic elimination and cognitive dysfunction (2011). modelling for primary immunodeficiencies that during recovery from neuropathogenic flaviviruses. 13. Shiba, Y. et al. Human ES-cell-derived cardiomyocytes lead to a loss of cell-intrinsic immunity in cortical Nat. Neurosci. 22, 1276–1288 (2019). electrically couple and suppress arrhythmias in injured neurons to HSV-1 infection. 52. Blank, T. et al. Brain endothelial- and epithelial-specific hearts. Nature 489, 322–325 (2012). 32. Cugola, F. R. et al. The Brazilian Zika virus strain interferon receptor chain 1 drives virus-induced 14. Ma, L. et al. Human embryonic stem cell-derived causes birth defects in experimental models. Nature sickness behavior and cognitive impairment. Immunity GABA neurons correct locomotion deficits in quinolinic 534, 267–271 (2016). 44, 901–912 (2016). acid-lesioned mice. Cell Stem Cell 10, 455–464 33. Garcez, P. P. et al. Zika virus impairs growth in human 53. Lee, S. H., Lumelsky, N., Studer, L., Auerbach, J. M. (2012). neurospheres and brain organoids. Science 352, & McKay, R. D. Efficient generation of midbrain and 15. Wang, S. et al. Human iPSC-derived oligodendrocyte 816–818 (2016). hindbrain neurons from mouse embryonic stem cells. progenitor cells can myelinate and rescue a mouse Cugola et al. (2016) and Garcez et al. (2016) Nat. Biotechnol. 18, 675–679 (2000). model of congenital hypomyelination. Cell Stem Cell are early examples of cerebral organoids being 54. Chambers, S. M. et al. Highly efficient neural 12, 252–264 (2013). used to study the tropism and pathogenesis of conversion of human ES and iPS cells by dual 16. Gornalusse, G. G. et al. HLA-E-expressing pluripotent ZIKV, identifying NPCs as a susceptible cell type. inhibition of SMAD signaling. Nat. Biotechnol. 27, stem cells escape allogeneic responses and lysis by 34. Delhaye, S. et al. Neurons produce type I interferon 275–280 (2009). NK cells. Nat. Biotechnol. 35, 765–772 (2017). during viral encephalitis. Proc. Natl Acad. Sci. USA 55. Chambers, S. M. et al. Combined small-molecule 17. Kikuchi, T. et al. Human iPS cell-derived dopaminergic 103, 7835–7840 (2006). inhibition accelerates developmental timing and neurons function in a primate Parkinson’s disease 35. Zimmer, B. et al. Human iPSC-derived trigeminal converts human pluripotent stem cells into model. Nature 548, 592–596 (2017). neurons lack constitutive TLR3-dependent immunity nociceptors. Nat. Biotechnol. 30, 715–720 (2012). 18. Deuse, T. et al. Hypoimmunogenic derivatives of that protects cortical neurons from HSV-1 infection. 56. Liu, Y. et al. Medial ganglionic eminence-like cells induced pluripotent stem cells evade immune rejection Proc. Natl Acad. Sci. USA 115, E8775–E8782 derived from human embryonic stem cells correct in fully immunocompetent allogeneic recipients. (2018). learning and memory deficits. Nat. Biotechnol. 31, Nat. Biotechnol. 37, 252–258 (2019). 36. Omalu, B. I., Shakir, A. A., Wang, G., Lipkin, W. I. 440–447 (2013). 19. Mestas, J. & Hughes, C. C. W. Of mice and not men: & Wiley, C. A. Fatal fulminant pan-meningo- 57. Liu, Y. et al. Directed differentiation of forebrain differences between mouse and human immunology. polioencephalitis due to West Nile virus. Brain Pathol. GABA interneurons from human pluripotent stem J. Immunol. 172, 2731–2738 (2004). 13, 465–472 (2003). cells. Nat. Protoc. 8, 1670–1679 (2013).

NATuRe RevIeWS | Immunology volume 21 | July 2021 | 451 Reviews

58. Maroof, A. M. et al. Directed differentiation and neural precursors from human embryonic stem cells. virus infection in fetal-like organoids and adult brain. functional maturation of cortical interneurons from Nat. Biotechnol. 19, 1129–1133 (2001). Cell Stem Cell 21, 274–283.e5 (2017). human embryonic stem cells. Cell Stem Cell 12, 84. Watanabe, K. et al. A ROCK inhibitor permits Xu et al. (2016) and Zhou et al. (2017) provide 559–572 (2013). survival of dissociated human embryonic stem cells. examples of how hPSC-based models are scalable 59. Du, Z.-W. et al. Generation and expansion of Nat. Biotechnol. 25, 681–686 (2007). so that they can be used to perform high-content highly pure motor neuron progenitors from human 85. Eiraku, M. et al. Self-organized formation of polarized drug screens, here discovering therapeutic pluripotent stem cells. Nat. Commun. 6, 6626 (2015). cortical tissues from ESCs and its active manipulation candidates that act against ZIKV infection. 60. Lu, J. et al. Generation of serotonin neurons from by extrinsic signals. Cell Stem Cell 3, 519–532 110. Wilson, A. C. & Mohr, I. A cultured affair: HSV latency human pluripotent stem cells. Nat. Biotechnol. 34, (2008). and reactivation in neurons. Trends Microbiol. 20, 89–94 (2016). 86. Qian, X. et al. Brain-region-specific organoids using 604–611 (2012). 61. Krencik, R., Weick, J. P., Liu, Y., Zhang, Z.-J. & mini- for modeling ZIKV exposure. Cell 111. Markus, A., Lebenthal-Loinger, I., Yang, I. H., Zhang, S.-C. Specification of transplantable 165, 1238–1254 (2016). Kinchington, P. R. & Goldstein, R. S. An in vitro model astroglial subtypes from human pluripotent stem 87. Jo, J. et al. Midbrain-like organoids from human of latency and reactivation of varicella zoster virus in cells. Nat. Biotechnol. 29, 528–534 (2011). pluripotent stem cells contain functional dopaminergic human stem cell-derived neurons. PLoS Pathog. 11, 62. Li, X. et al. Fast generation of functional subtype and neuromelanin-producing neurons. Cell Stem Cell e1004885 (2015). astrocytes from human pluripotent stem cells. 19, 248–257 (2016). 112. Sadaoka, T. et al. In vitro system using human neurons Stem Cell Rep. 11, 998–1008 (2018). 88. Cederquist, G. Y. et al. Specification of positional demonstrates that varicella-zoster vaccine virus is 63. Tchieu, J. et al. NFIA is a gliogenic switch enabling identity in forebrain organoids. Nat. Biotechnol. 37, impaired for reactivation, but not latency. Proc. Natl rapid derivation of functional human astrocytes from 436–444 (2019). Acad. Sci. USA 113, E2403–E2412 (2016). pluripotent stem cells. Nat. Biotechnol. 37, 267–275 89. Ormel, P. R. et al. Microglia innately develop within 113. Pourchet, A., Modrek, A. S., Placantonakis, D. G., (2019). cerebral organoids. Nat. Commun. 9, 4167 (2018). Mohr, I. & Wilson, A. C. Modeling HSV-1 latency 64. Bradley, R. A. et al. Regionally specified human 90. Miller, J. D. et al. Human iPSC-based modeling in human embryonic stem cell-derived neurons. pluripotent stem cell-derived astrocytes exhibit of late-onset disease via -induced aging. Pathogens 6, 24 (2017). different molecular signatures and functional Cell Stem Cell 13, 691–705 (2013). Markus et al. (2015), Sadaoka et al. (2016) and properties. Development 146, dev170910 (2019). 91. Cowan, C. S. et al. Cell types of the human retina Pourchet et al. (2017) apply hPSC-derived neuronal 65. Yang, N. et al. Generation of oligodendroglial cells and its organoids at single-cell resolution. Cell 182, cultures to study viral latency. by direct lineage conversion. Nat. Biotechnol. 31, 1623–1640.e34 (2020). 114. Tangye, S. G. et al. Human inborn errors of immunity: 434–439 (2013). 92. Vatine, G. D. et al. Human iPSC-derived blood-brain 2019 update on the classification from the 66. Tao, Y. & Zhang, S.-C. Neural subtype specification barrier chips enable disease modeling and personalized international union of immunological societies expert from human pluripotent stem cells. Cell Stem Cell 19, medicine applications. Cell Stem Cell 24, 995–1005.e6 committee. J. Clin. Immunol. 40, 24–64 (2020). 573–586 (2016). (2019). 115. Hoffmann, M. et al. Chloroquine does not inhibit 67. Vierbuchen, T. et al. Direct conversion of fibroblasts 93. Cakir, B. et al. Engineering of human brain organoids infection of human lung cells with SARS-CoV-2. to functional neurons by defined factors. Nature 463, with a functional vascular-like system. Nat. Methods Nature 585, 588–590 (2020). 1035–1041 (2010). 16, 1169–1175 (2019). 116. Wang, M. et al. Remdesivir and chloroquine effectively 68. Pang, Z. P. et al. Induction of human neuronal cells by 94. Pellegrini, L. et al. SARS-CoV-2 infects the brain choroid inhibit the recently emerged novel coronavirus defined transcription factors. Nature 476, 220–223 plexus and disrupts the blood-CSF-barrier in human (2019-nCoV) in vitro. Cell Res. 30, 269–271 (2020). (2012). brain organoids. Cell Stem Cell 27, 951–961.e5 117. Liu, J. et al. Hydroxychloroquine, a less toxic derivative 69. Wells, M. F. et al. Genome-wide screens in accelerated (2020). of chloroquine, is effective in inhibiting SARS-CoV-2 human stem cell- derived neural progenitor cells 95. Tang, H. et al. Zika virus infects human cortical neural infection in vitro. Cell Discov. 6, 16 (2020). identify Zika virus host factors and drivers of progenitors and attenuates their growth. Cell Stem Cell 118. Gupta, A. et al. Extrapulmonary manifestations proliferation. Preprint at bioRxiv https://doi.org/ 18, 587–590 (2016). of COVID-19. Nat. Med. 26, 1017–1032 (2020). 10.1101/476440 (2018). 96. Nowakowski, T. J. et al. Expression analysis highlights 119. Ellul, M. A. et al. Neurological associations of 70. Thion, M. S., Ginhoux, F. & Garel, S. Microglia and AXL as a candidate Zika virus entry receptor in neural COVID-19. Lancet Neurol. 19, 767–783 (2020). early brain development: an intimate journey. Science stem cells. Cell Stem Cell 18, 591–596 (2016). 120. Lamers, M. M. et al. SARS-CoV-2 productively infects 362, 185–189 (2018). 97. Wells, M. F. et al. Genetic ablation of AXL does not human gut enterocytes. Science 369, 50–54 (2020). 71. Lee, C. Z. W., Kozaki, T. & Ginhoux, F. Studying protect human neural progenitor cells and cerebral 121. Zhou, J. et al. Infection of bat and human intestinal tissue macrophages in vitro: are iPSC-derived cells organoids from Zika virus infection. Cell Stem Cell 19, organoids by SARS-CoV-2. Nat. Med. 26, 1077–1083 the answer? Nat. Rev. Immunol. 18, 716–725 703–708 (2016). (2020). (2018). 98. Yoon, K.-J. et al. Zika-virus-encoded NS2A disrupts 122. Duan, X. et al. Identification of drugs blocking 72. Sturgeon, C. M., Ditadi, A., Awong, G., Kennedy, M. mammalian cortical neurogenesis by degrading SARS-CoV-2 infection using human pluripotent stem & Keller, G. Wnt signaling controls the specification adherens junction proteins. Cell Stem Cell 21, cell-derived colonic organoids. Preprint at bioRxiv of definitive and primitive hematopoiesis from human 349–358.e6 (2017). https://doi.org/10.1101/2020.05.02.073320 (2020). pluripotent stem cells. Nat. Biotechnol. 32, 554–561 99. Liang, Q. et al. Zika Virus NS4A and NS4B proteins 123. Pérez-Bermejo, J. A. et al. SARS-CoV-2 infection of (2014). deregulate Akt-mTOR signaling in human fetal human iPSC-derived cardiac cells predicts novel 73. Muffat, J. et al. Efficient derivation of microglia-like neural stem cells to inhibit neurogenesis and induce cytopathic features in hearts of COVID-19 patients. cells from human pluripotent stem cells. Nat. Med. autophagy. Cell Stem Cell 19, 663–671 (2016). Preprint at bioRxiv https://doi.org/10.1101/2020.08. 22, 1358–1367 (2016). 100. Li, H. et al. Zika virus protease cleavage of host 25.265561 (2020). 74. Abud, E. M. et al. iPSC-derived human microglia-like protein Septin-2 mediates mitotic defects in neural 124. Yang, L. et al. A human pluripotent stem cell-based cells to study neurological diseases. Neuron 94, progenitors. Neuron 101, 1089–1098.e4 (2019). platform to study SARS-CoV-2 tropism and model 278–293.e9 (2017). 101. Mesci, P. et al. Modeling neuro-immune interactions virus infection in human cells and organoids. 75. Pandya, H. et al. Differentiation of human and murine during Zika virus infection. Hum. Mol. Genet. 27, Cell Stem Cell 27, 125–136.e7 (2020). induced pluripotent stem cells to microglia-like cells. 41–52 (2018). This study is the first to report the selective Nat. Neurosci. 20, 753–759 (2017). 102. Muffat, J. et al. Human induced pluripotent stem tropism of SARS-CoV-2 for specific subpopulations 76. Douvaras, P. et al. Directed differentiation of human cell-derived glial cells and neural progenitors display of CNS cells. pluripotent stem cells to microglia. Stem Cell Rep. 8, divergent responses to Zika and dengue infections. 125. Sharma, A. et al. Human iPSC-derived cardiomyocytes 1516–1524 (2017). Proc. Natl Acad. Sci. USA 115, 7117–7122 (2018). are susceptible to SARS-CoV-2 infection. Cell Rep. 77. Takata, K. et al. Induced-pluripotent-stem-cell-derived 103. Quicke, K. M. et al. Zika virus infects human placental Med. 1, 100052 (2020). primitive macrophages provide a platform for modeling macrophages. Cell Host Microbe 20, 83–90 (2016). 126. Monteil, V. et al. Inhibition of SARS-CoV-2 infections tissue-resident macrophage differentiation and 104. Vermillion, M. S. et al. Intrauterine Zika virus in engineered human tissues using clinical-grade function. Immunity 47, 183–198.e6 (2017). infection of pregnant immunocompetent mice models soluble human ACE2. Cell 181, 905–913.e7 (2020). 78. Haenseler, W. et al. A highly efficient human transplacental transmission and adverse perinatal 127. Han, Y. et al. Identification of candidate COVID-19 pluripotent stem cell microglia model displays a outcomes. Nat. Commun. 8, 14575 (2017). therapeutics using hPSC-derived lung organoids. neuronal-co-culture-specific expression profile and 105. Ginhoux, F. & Guilliams, M. Tissue-resident macrophage Preprint at bioRxiv https://doi.org/10.1101/2020.05. inflammatory response. Stem Cell Rep. 8, 1727–1742 ontogeny and homeostasis. Immunity 44, 439–449 05.079095 (2020). (2017). (2016). 128. Jacob, F. et al. Human pluripotent stem cell-derived 79. Hasselmann, J. et al. Development of a chimeric 106. Puschnik, A. S., Majzoub, K., Ooi, Y. S. & Carette, J. E. neural cells and brain organoids reveal SARS-CoV-2 model to study and manipulate human microglia A CRISPR toolbox to study virus-host interactions. neurotropism predominates in choroid plexus in vivo. Neuron 103, 1016–1033.e10 (2019). Nat. Rev. Microbiol. 15, 351–364 (2017). . Cell Stem Cell S1934-5909, 30463-X 80. Svoboda, D. S. et al. Human iPSC-derived 107. Li, Y. et al. Genome-wide CRISPR screen for Zika virus (2020). microglia assume a primary microglia-like state resistance in human neural cells. Proc. Natl Acad. Sci. This is early example of using regionalized after transplantation into the neonatal mouse brain. USA 116, 9527–9532 (2019). brain organoids to model SARS-CoV-2 tropism, Proc. Natl Acad. Sci. USA 116, 25293–25303 This study highlights the potential to use hPSC- establishing susceptibility of choroid plexus (2019). based platforms to perform whole-genome organoids for SARS-CoV-2 infection. 81. Mancuso, R. et al. Stem-cell-derived human microglia screens in human, disease-relevant cells, in this 129. Song, E. et al. Neuroinvasion of SARS-CoV-2 in human transplanted in mouse brain to study human disease. case identifying critical host factors involved in and mouse brain. Preprint at bioRxiv https://doi.org/ Nat. Neurosci. 22, 2111–2116 (2019). ZIKV infection. 10.1101/2020.06.25.169946 (2020). 82. Schmunk, G. et al. Human microglia upregulate 108. Xu, M. et al. Identification of small-molecule 130. Topol, E. J. COVID-19 can affect the heart. Science cytokine signatures and accelerate maturation of inhibitors of Zika virus infection and induced neural 370, 408–409 (2020). neural networks. Preprint at bioRxiv https://doi.org/ cell death via a drug repurposing screen. Nat. Med. 131. Dixit, A. et al. Perturb-seq: dissecting molecular 10.1101/2020.03.24.006874 (2020). 22, 1101–1107 (2016). circuits with scalable single-cell RNA profiling of 83. Zhang, S.-C., Wernig, M., Duncan, I. D., Brüstle, O. & 109. Zhou, T. et al. High-content screening in hPSC-neural pooled genetic screens. Cell 167, 1853–1866.e17 Thomson, J. A. In vitro differentiation of transplantable progenitors identifies drug candidates that inhibit Zika (2016).

452 | July 2021 | volume 21 www.nature.com/nri Reviews

132. Desole, G. et al. Modelling neurotropic flavivirus 138. Sison, S. L. et al. Human cytomegalovirus disruption Acknowledgements infection in human induced pluripotent stem of calcium signaling in neural progenitor cells and O.H. and L.S. are supported by grants from the US National cell-derived systems. Int. J. Mol. Sci. 20, (2019). organoids. J. Virol. 93, e00954–e01019 (2019). Institutes of Health. O.H. was supported by a training award 133. Lang, J. et al. An hPSC-derived tissue-resident 139. Poole, E. et al. An iPSC-derived myeloid lineage model from the NYSTEM grant #C32559GG and the Center for macrophage model reveals differential responses of herpes virus latency and reactivation. Front. Microbiol. Stem Cell Biology at MSK. The authors thank all members of of macrophages to ZIKV and DENV infection. 10, 2233 (2019). the Studer laboratory for their critical input and stimulating Stem Cell Rep. 11, 348–362 (2018). 140. D’Aiuto, L. et al. Human induced pluripotent scientific discussions. 134. Cosset, É. et al. Human three-dimensional engineered stem cell-derived models to investigate human neural tissue reveals cellular and molecular events cytomegalovirus infection in neural cells. PLoS ONE Author contributions following cytomegalovirus infection. Biomaterials 53, 7, e49700 (2012). The authors contributed equally to all aspects of the article. 296–308 (2015). 141. Brown, R. M. et al. Human cytomegalovirus Competing interests 135. Markus, A. et al. Varicella-zoster virus (VZV) infection compromises development of cerebral organoids. The authors declare no competing interests. of neurons derived from human embryonic stem cells: J. Virol. 93, 00957–01019 (2019). direct demonstration of axonal infection, transport 142. Sundaramoorthy, V. et al. Modelling lyssavirus Peer review information of VZV, and productive neuronal infection. J. Virol. 85, infections in human stem cell-derived neural cultures. Nature Reviews Immunology thanks M. Deleidi and the other, 6220–6233 (2011). Viruses 12, 359 (2020). anonymous, reviewer(s) for their contribution to the peer 136. Kurapati, S. et al. Role of the JNK pathway in varicella- 143. Carty, M., Reinert, L., Paludan, S. R. & Bowie, A. G. review of this work. zoster virus lytic infection and reactivation. J. Virol. Innate antiviral signalling in the central nervous 91, 00640 (2017). system. Trends Immunol. 35, 79–87 (2014). Publisher’s note 137. Lee, K. S. et al. Human sensory neurons derived 144. Guo, Y. et al. Herpes simplex virus encephalitis Springer Nature remains neutral with regard to jurisdictional from induced pluripotent stem cells support in a patient with complete TLR3 deficiency: TLR3 claims in published maps and institutional affiliations. Varicella-Zoster virus infection. PLoS ONE 7, is otherwise redundant in protective immunity. e53010 (2012). J. Exp. Med. 208, 2083–2098 (2011). © Springer Nature Limited 2021

NATuRe RevIeWS | Immunology volume 21 | July 2021 | 453