You are on page 1of 3

https://doi.org/10.

1093/brain/awad290 BRAIN 2023: 146; 3963–3965 | 3963

Post-herpes simplex virus encephalitis


autoimmunity: more the rule than
the exception

Downloaded from https://academic.oup.com/brain/article/146/10/3963/7252803 by guest on 11 October 2023


This scientific commentary refers to ‘Neurologic complications in antibodies at the time of the HSE, yet some (30%) went on to develop
herpes simplex encephalitis: clinical, immunological and genetic neuronal antibodies without clinical relapses. Indeed, the presence
studies’ by Armangué et al. (https://doi.org/10.1093/brain/ of any neuronal antibodies at 3 weeks following HSE was predictive
awad238). of a subsequent symptomatic relapse later in the course of the dis­
ease (odds ratio, OR 11.5).8 However, the reason why some patients
Serendipity is a factor in many discoveries in science and medicine, developed symptomatic or asymptomatic secondary autoimmun­
but perseverance is the key to success. The scientific story of post- ity following HSE, whereas others did not, and the factors that pre­
herpes simplex virus I autoimmune encephalitis (HSVE-AE) is laden disposed patients to symptomatic relapses, remained unclear.
with keen observations, chance findings and coincidences, but it is Early hypotheses for the mechanisms underlying post-HSVE-AE
also a story of persistence. It began with the unexpected discovery considered the possibility of molecular mimicry between HSV
of N-methyl-D-aspartate receptor (NMDAR) antibodies in the sera of surface molecules and NMDARs. Yet post-viral encephalitis does
a number of patients with herpes simplex virus I encephalitis (HSE) not appear to be limited to HSE but also occurs following a wide
recruited as a control group during the development of a standar­ range of other viral encephalitis variants, including Japanese
dized NMDAR antibody assay. A retrospective analysis of 44 HSE pa­ B-encephalitis,9 arguing against this idea. Instead, it is likely that
tients treated at a German university hospital then identified NMDAR the co-occurrence of a tissue-destructive viral encephalitis releas­
antibodies in 30% of these individuals.1 This discovery in turn led to ing neuronal antigens, together with the strong pro-inflammatory
the prospective identification of NMDAR antibodies in the CSF of an stimulus mediated by viral particles via conserved pattern-
adult patient with a post-HSE relapse. Interestingly, NMDAR anti­ recognition receptors (PRRs), such as toll-like receptors (TLRs),
bodies had not been present in CSF obtained during this patient’s causes auto-immunization (Fig. 1). The PRR chiefly responsible for
HSE episode 4 weeks earlier. This case came to the attention of detecting DNA viruses like HSV is TLR3. Upon detection of intracel­
Josep Dalmau via a visiting German neuroscientist.2 lular double-stranded RNA produced during viral replication, TLR3
In parallel (https://doi.org/10.1016/j.jpeds.2012.10.011), Thais induces the production of type I interferons (IFN-I), the body’s first
Armangué—a neuropaediatrician working in Josep Dalmau’s lab line of antiviral defence. By regulating the expression of hundreds
—put together a small case series of children who had developed of different genes, IFNs are able to block virus replication at numer­
post-HSV choreoathetosis, a syndrome familiar to paediatricians ous levels. Correspondingly, mutations in the TLR3 gene or compo­
but mostly considered to be the result of viral relapse, rather than nents of the associated signalling cascade are known to predispose
autoimmunity. Some aspects of this syndrome resembled the spec­ to relapsing HSE. Furthermore, there is a strong link between hy­
trum of movement disorders associated with anti-NMDAR enceph­ peractivation of the IFN-I system and autoimmunity (systemic lu­
alitis, which had been described by Josep Dalmau and colleagues3 a pus erythematosus) as well as hyperinflammation syndromes
few years earlier. Armangué demonstrated that these patients also (Aicardi-Goutières syndrome). Therefore, it was tempting to specu­
showed seroconversion, with the appearance of NMDAR antibodies late that the TLR3/IFN pathway might have a role in the develop­
in CSF in association with clinical relapses. These observations in ment of secondary, post-viral autoimmunity.
children and adults were published together4 and were confirmed In their latest work, published in this issue of Brain, Armangué,
almost in parallel by British5 and Australian6 groups. Dalmau and colleagues10 add novel and exciting findings to the
Having picked up the scent, Armangué then orchestrated a field of post-HSVE-AE, based on their extensive prospective
Spanish prospective observational multicentric study to recruit pa­ Spanish cohort of 93 patients with HSE and an international cohort
tients with HSV in a standardized fashion.7 This commendable ef­ of 97 patients with post-HSVE-AE. They confirmed that 42% of pa­
fort resulted in a large prospective series of 51 patients with HSV, tients in the Spanish cohort developed neuronal antibodies follow­
27% of whom went on to experience secondary autoimmune en­ ing HSE, 54% of whom (23% of all HSE patients) went on to develop
cephalitis relapses—often therapy-responsive—within 2 months symptomatic autoimmune relapses, usually within 3–6 weeks. This
after HSE. While most harboured NMDAR antibodies in their CSF figure strengthens earlier observations and supports previous
(64%), others had antibodies targeting unknown neural antigens in­ calls for the adoption of a systematic management strategy in
stead or in addition (36%). None had NMDAR or other neuronal patients following HSV, with clinical visits and further lumbar

Received August 24, 2023. Accepted August 24, 2023. Advance access publication August 28, 2023
© The Author(s) 2023. Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For permissions, please e-mail:
journals.permissions@oup.com
3964 | BRAIN 2023: 146; 3963–3965 Scientific Commentary

Downloaded from https://academic.oup.com/brain/article/146/10/3963/7252803 by guest on 11 October 2023


Figure 1 Current hypotheses for the mechanism underlying post-viral encephalitis autoimmune encephalitis relapses. (1) Neurotropic viruses (e.g.
herpes simplex virus I) cause neuronal destruction leading to widespread release of neuronal antigens (e.g. NMDAR subunits) and viral components.
(2) Viral components, such as double-stranded RNA, activate pattern recognition receptors (PRRs, e.g. toll-like receptor 3; TLR3), which induce an inter­
feron type I (IFN-I) response as well as co-stimulatory signalling. (3) Together with the presentation of neuronal antigens by antigen-presenting cells
(dendritic cell in purple, left part of circle) to T lymphocytes (center cell in circle, green), this results in breach of tolerance. The autoreactive T cells can
in turn stimulate autoantigen-specific B cells (right cell within circle, green with surface antibodies) to develop into short-lived plasma blasts or cells (4)
whose autoantibodies then target dendritic NMDARs (5) and cause receptor internalization, net loss of surface NMDARs, synaptic and eventually net­
work dysfunction. In parallel, IFN-I causes expression of major histocompatibility complex type I (MHC-I) on the surface of neurons (not shown), there­
by rendering them susceptible to cytotoxic T cells. Many questions remain unanswered: Where does the breach of tolerance take place, in perivascular
spaces in the brain or in cervical lymph nodes? Why is the NMDAR such a frequent autoantigen and which other endogenous factors limit/propagate
autoimmunity in the context of post-HSE autoimmunity? Created with BioRender.com.

punctures. The authors delineated three somewhat distinct clinical of infectious encephalitis: a significant proportion (at least 20%) of
syndromes associated with these relapses: (i) mostly NMDAR patients with HSE will go on to develop symptomatic—sometimes
antibody-associated choreoathetosis in children; (ii) idiopathic difficult to discriminate from residual symptoms—post-HSE auto­
anti-NMDAR encephalitis-like syndromes (psychiatric symptoms, immune relapse, a treatable complication. We would all be well
seizures, decreased level of consciousness and dysautonomia) in advised to accept that post-HSVE AE is more the rule than the
teenagers/adults; and (iii) isolated neuropsychiatric/behavioural exception and to change our post-acute management of these
phenotypes (often associated with non-NMDAR, neuronal autoanti­ patients accordingly.
bodies). Intriguingly, they identified a moderate (yet significant) in­
crease in specific type I interferon signals in the blood 3 weeks Frank Leypoldt1,2 and Klaus-Peter Wandinger1
after HSE as the strongest predictor of developing symptomatic
1 Institute of Clinical Chemistry, University-Hospital Schleswig Holstein
post-HSVE-AE (OR 34.8). Patients with detectable neuronal autoanti­
Kiel/Lübeck, 24105 Kiel, Germany
bodies but without clinical relapse did not show this phenotype. Of
2 Department of Neurology, University-Hospital Schleswig Holstein,
note, a very high type I IFN signature at 3 weeks following HSE was
24105 Kiel, Germany
associated with uncontrolled viral encephalitis. Previously hypothe­
sized polymorphisms or mutations in TLR3/IFN-related genes—pre­
Correspondence to: Frank Leypoldt
disposing to HSE—were not detected in the present cohort. The
E-mail: Frank.Leypoldt@uksh.de
authors did, however, observe some additional risk of autoimmune
relapses conferred by the absence of a very common HLA class I
gene (HLA-A*02), an observation that further implicates CD8+
T-lymphocytes in the pathogenesis of post-HSVE-AE.
Funding
In summary, the discovery of post-HSVE-AE and its systematic F.L. is funded by the Bundesministerium für Bildung und Forschung
clinical characterization is of the utmost importance in the field BMBF (01GM1908A und 01GM2208), E-Rare Joint Transnational
Scientific Commentary BRAIN 2023: 146; 3963–3965 | 3965

research support (ERA-Net, LE3064/2-1), Stiftung Pathobiochemie of 4. Armangue T, Leypoldt F, Málaga I, et al. Herpes simplex virus en­
the German Society for Laboratory Medicine and HORIZON MSCA cephalitis is a trigger of brain autoimmunity. Ann Neurol. 2014;
2022 Doctoral Network 101119457 IgG4-TREAT and discloses speak­ 75:317-323.
er honoraria from Grifols, Teva, Biogen, Bayer, Roche, Novartis, 5. Hacohen Y, Deiva K, Pettingill P, et al. N-methyl-D-aspartate re­
Fresenius, travel funding from Merck, Grifols and Bayer and serving ceptor antibodies in post-herpes simplex virus encephalitis
on advisory boards for Roche, Biogen and Alexion. neurological relapse. Movement Disord. 2014;29:90-96.
6. Mohammad SS, Sinclair K, Pillai S, et al. Herpes simplex enceph­
alitis relapse with chorea is associated with autoantibodies to
N-methyl-D-aspartate receptor or dopamine-2 receptor.
Competing interests Movement Disord. 2014;29:117-122.
The authors report no competing interests. 7. Armangue T, Moris G, Cantarín-Extremera V, et al. Autoimmune
post-herpes simplex encephalitis of adults and teenagers.
Neurology. 2015;85:1736-1743.
8. Armangue T, Spatola M, Vlagea A, et al. Frequency, symptoms,
References risk factors, and outcomes of autoimmune encephalitis after

Downloaded from https://academic.oup.com/brain/article/146/10/3963/7252803 by guest on 11 October 2023


1. Prüss H, Finke C, Höltje M, et al. N-methyl-D-aspartate receptor herpes simplex encephalitis: A prospective observational study
antibodies in herpes simplex encephalitis. Ann Neurol. 2012;72: and retrospective analysis. Lancet Neurol. 2018;17:1-13.
902-911. 9. Ma BJ, Han W, Li J. Japanese encephalitis-induced anti-N-
2. Leypoldt F, Titulaer MJ, Aguilar E, et al. Herpes simplex virus-1 methyl-d-aspartate receptor encephalitis: A hospital-based
encephalitis can trigger anti-NMDA receptor encephalitis: prospective study. Brain Dev. 2019;42:179-184.
Case report. Neurology. 2013;81:1637-1639. 10. Armangué T, Olive-Cirera G, Martinez-Hernandez E, et al.
3. Dalmau J, Tüzün E, Wu HY, et al. Paraneoplastic anti-N-methyl- Neurologic complications in herpes simplex encephalitis:
D-aspartate receptor encephalitis associated with ovarian tera­ clinical, immunological and genetic studies. Brain. 2023;146:
toma. Ann Neurol. 2007;61:25-36. 4306-4319.

You might also like