Next Article in Journal
Impact of Graphene Layers on Genetic Expression and Regulation within Sulfate-Reducing Biofilms
Previous Article in Journal
Congenital Suipoxvirus Infection in Newborn Piglets in an Austrian Piglet-Producing Farm
Previous Article in Special Issue
A Retrospective Cross-Sectional Analysis of Viral SARI in Pregnant Women in Southern Brazil
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Central Nervous System Disorders with Auto-Antibodies in People Living with HIV

1
Department of Infectious-Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Via Don Angelo Sempreboni 5, Negrar, 37024 Verona, Italy
2
Unit of Neurology, Ospedale Maria Vittoria, ASL Città di Torino, Via Luigi Cibrario, 72, 10144 Torino, Italy
3
Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Ospedale Amedeo di Savoia, C.so Svizzera 164, 10124 Torino, Italy
*
Author to whom correspondence should be addressed.
Microorganisms 2024, 12(9), 1758; https://doi.org/10.3390/microorganisms12091758
Submission received: 24 June 2024 / Revised: 20 August 2024 / Accepted: 21 August 2024 / Published: 24 August 2024
(This article belongs to the Special Issue Viral Infection and Immunity)

Abstract

:
Abstract: People living with HIV (PLWH) may present atypical neurological complications. Recently, autoimmune manifestations of the central nervous system (CNS) have been described. We retrospectively described the features of PLWH presenting with acute neurological symptoms with positive anti-CNS antibodies. We analyzed relevant CSF characteristics. Twelve patients were identified, with demyelinating, inflammatory, or no MRI lesions. We observed CSF inflammatory features. Aspecific CSF anti-CNS antibodies were found in all subjects and a specific antibody (second-level blotting panel) was found in one. The cases presented a slow resolution of symptoms with sequelae. More studies are needed to better describe the spectrum and prognosis of autoimmune CNS diseases in PLWH.

1. Introduction

People living with HIV (PLWH) may present complications after the introduction of combined antiretroviral therapy (cART), including cardiovascular, metabolic, and neurological issues. HIV encephalitis is currently rare but other neurological disorders are not uncommon, including HIV-associated neurocognitive disorder (HAND), which is now estimated to be as prevalent as 20–30% in PLWH with controlled HIV viremia. While these disorders have a complex pathogenesis, chronic inflammation may also play an important role; abnormal levels of cerebrospinal fluid (CSF) neopterin have been shown even after years of successful antiviral treatment [1,2]. Additionally, autoimmune disorders of the central nervous system (CNS) have been reported in PLWH, and the spectrum now includes anti-NMDAR encephalitis and anti-NSA encephalitis with psychotic onset [3,4,5,6]. Immune response inflammatory syndrome (IRIS) may occur after the start of antiretroviral treatment; recently, a new, uncommon acute CNS inflammatory condition has been described and reported to be related to the brain parenchyma invasion of CD8+ T lymphocytes [7,8]. On the contrary, PLWH with multiple sclerosis have a mild course and a less aggressive disease, suggesting that immune modulation may be beneficial in the context of immune-mediated disorders [9].
Due to loss of immune competence, inflammatory and autoimmune diseases can occur in PLWH, in whom diagnosis is more difficult due to differences in classical antibody titer and onset [10]. Immune system dysregulation is one of the key recognized features of the pathogenesis of non-AIDS-related comorbidities, and recently, autoimmune manifestations of the central nervous system (CNS) have been described. HIV patients have a higher incidence of systemic autoimmune diseases, even on highly active antiretroviral therapy (HAART) [11], commonly with broad spectrum production of non-organ specific antibodies and hypergammaglobulinemia [12]. Treg cells and Th17 cells may also play a role in HIV infection and progression to AIDS and the development of autoimmune diseases. [13]. Additionally, other mechanisms have been proposed: HIV exerts a direct effect on B cells through its binding to B cell surface molecule CD40. Molecular mimicry between HIV proteins and self-antigens could cause antibody cross-reactions and therefore lead to broad autoimmune manifestations [14]. Given the limited number of reported cases and the potential underdiagnosis of these disturbances, we aimed to describe the clinical and diagnostic features of PLWH presenting autoimmune disorders of the CNS.

2. Materials and Methods

We retrospectively reviewed cases of PLWH with CNS disorders who had been admitted to the Unit of Infectious Diseases, Amedeo di Savoia Hospital, Torino (Italy), between 2001 and 2019 and presented anti-CNS auto-antibodies. The patients signed a written informed consent form for CSF withdrawal, storage, and analysis. The study was approved by the local Ethics Committee (Comitato Interaziendale di Orbassano, “PRODIN study”, protocol number 103). We included patients with confirmed positivity for serum/CSF anti-CNS antibodies identified using immunofluorescence and immunoblot methods (Euroimmun AG PerkinElmer company (Waltham, MA, USA)). Any patient with active central nervous system infections, such as acute confirmed infectious encephalitis of any etiology, mycobacteriosis, herpes viruses (beyond just isolated detection of EBV DNA), neurosyphilis, or cryptococcosis, was excluded from the study. Any patient with a known autoimmune disease was excluded from the study. EBV DNA was tested when possible for its role in neuro-inflammatory diseases. Beyond first-level CSF markers (cells, proteins, and glucose ratio), CSF was tested for blood–brain barrier (BBB) permeability (CSF to serum albumin ratio, “CSAR”), inflammation (CSF to serum IgG ratio, neopterin), amyloid deposition (1–42 β-amyloid), neuronal damage (Total tau (T-tau)), phosphorylated tau (P-tau), 14-3-3 protein), and astrocyte damage (S-100 β). Quantitative determination of albumin in serum and CSF was measured using immunoturbidimetric methods (AU 5800. Beckman Coulter, Brea, CA, USA); 14-3-3 protein was detected using the Western blot (WB) technique; and CSF tau, P-tau, and 1–42 β-amyloid were measured using immunoenzymatic methods (Fujirebio diagnostics, Malvern, PA, USA). Neopterin and S-100β were measured through validated ELISA methods [DRG Diagnostics (Marburg, Germany) and DIAMETRA S.r.l. (Spello, Italy), respectively]. Imaging (either MRI or CT) and electrophysiological studies (EEG) were performed. Data were analyzed using standard statistical methods: variables were described as numbers (percentages) with medians [interquartile ranges (IQR) or ranges (minimum–maximum)]. Due to limited power, further subgroup analysis was not feasible. Data analysis was performed using SPSS software for Mac (version 26.0. IBM Corp (Armonk, NY, USA)).

3. Results

We identified twelve patients: they were more frequently male (75%) with a median age of 51 years [range: 42–55], 75% had European ancestry, and 50% took HAART. The median and nadir CD4 cell count were 302/mm3 [range: 120–434] and 93/mm3 [range: 18–259]; median plasma and CSF HIV RNA were respectively 922 [range: 0–31,556] and 2765 copies/mL [range: 32–21,829]. A median of 4 years [range: 1–7] since the diagnosis of HIV infection was recorded. When considering the MRI results, the participants presented with demyelinating (n = 3, 25%) or inflammatory lesions (2, 17%) while one (8%) presented with brain atrophy; a mixed pattern was observed in three subjects (25%) while the remaining three individuals showed a normal MRI (25%) at the onset of symptoms. Additional clinical and radiological features of the participants are presented in Table 1 and the Supplementary Data, Table S1.
Symptoms included headache, ataxia, dizziness, shaking, and agitation as well as walking, fluency, and memory disorders. CSF was clear in all patients with a median of 10 cells/mm3 [0–40], 65 mg/dL of proteins [41–126], and 52 mg/dL of glucose [42–65]. Median CSF neuro-markers were as follows: T-tau 160 pg/mL [87.5–362.2], P-Tau 29.5 pg/mL [19.2–37.2], neopterin 2 ng/mL [1–12.2], S100 Beta 120.5 pg/mL [52.2–179.2], and Beta Amyloid1–42 889 pg/mL [401.5–1189.2]. CSF 14-3-3 protein was positive in three patients (25%). Increased BBB permeability was observed in six participants (50%). No patient tested positive for any common pathogen responsible for meningoencephalitis, as per the inclusion criteria. In two patients in which EEG was studied, a diffuse cortical wave reduction was observed. Broad CSF anti-CNS antibodies (immunofluorescence technique) were found in all subjects and only one patient tested positive for specific antibodies (Recoverin, Zic4) when using the second-level blotting panel. The pattern of immunofluorescence was nuclear in five patients, synaptic in three, synaptic and nuclear in one, and non-specific in three (Table 2, Figure 1, Figure 2 and Figure 3 for negative and positive controls).
All but one patient had both serum and CSF antibody positivity. In the majority of cases, we observed a slow resolution of symptoms with sequelae (67%); specifically, five patients experienced residual sensory disorders, and three patients experienced neuropsychological (mainly memory) disorders. Two cases of concomitant symptomatic CSF escape were registered with complete resolution of symptoms after HAART optimization (Table 1). In patients 4, 5, and 8, CSF, EBV, and DNA were 54, 82, and 315 cp/mL, respectively (Table 1), and negative in the other four patients in which they were assessed (patients 6, 7, 11, and 12).

4. Discussion

In this case series, we measured several cerebrospinal fluid biomarkers of neuronal damage and inflammation of neurologically symptomatic PLWH. Our findings are consistent with classical CSF inflammatory features suggestive of encephalitis. More widely, our series may represent a good example of the variety of clinical manifestations in HIV-associated neurological disorders, moving through different pathological biotypes, as proposed in a recent study [15]. CSF biomarkers were altered in several patients with non-homogenously distributed patterns, yet they were consistent with disruption of the BBB, neuronal damage, and inflammation. There were no other factors favoring autoimmune disorders that may explain these features and no cases of para-neoplastic diseases were recorded; the role of EBV in such alterations remains uncertain. All patients had CNS-antibody positivity confirmed in CSF and all but one in plasma. We were not able to determine a specific antibody pattern to any of the standard antigens except for one case (patient n. 12 in Table 1) in which Recovering and Zic4 antibodies were found; given the association with neoplasias and the higher risk observed in PLWH, this subject is on active follow-up [14,16]. The majority of our patients had altered imaging with a high proportion of demyelinating and mixed lesions upon brain MRI; while HIV infection per se has been associated with such patterns, it would be interesting to understand whether auto-antibodies are the cause or consequence of neuronal damage. Viral infections have been described as potential triggers for several autoimmune disorders including encephalitis (the commonest being anti-nMDAR encephalitis after HSV CNS infections) [17]. It is also worrisome that several patients had neurological sequelae and this should prompt the early diagnosis and treatment of patients showing unexpected course or poor response to antiretroviral therapy. To our knowledge, few examples of such a correlation have previously been described, making this an important field for future research, especially considering the high prevalence of symptoms of such presentations and the current difficulty in reaching a reliable diagnosis. It is noteworthy that all of these patients presented with acute onset and neurological symptoms that could not be recognized using prompt classical classification and diagnostic tools (first-line assessment). A possible role of human herpesviridae is under investigation as a factor that leads to the progression of autoimmune encephalitis; HIV could actively have a similar role due to unique characteristics such as latency and its reversion, DNA total viral load, chronic microglial activation, and CNS inflammation in which other non-AIDS-defining conditions add load to the total pathological burden and promote the auto-inductive mechanism of progression to neurological issues that clinicians encounter daily [18,19]. This has been largely proven for HAND and aspecific conditions such as white matter hyperintensities, but the field of autoimmunity has not been explored thoroughly [20,21]. The occurrence of aspecific auto-antibodies in patients with symptomatic CSF escape may support the role of HIV replication as a trigger for immune-mediated self-damage [5,20].
A possible role for endogenous human viruses and for persistent herpes viruses has been highlighted repetitively for many inflammatory conditions and Multiple Sclerosis (MS) is just one of them [22,23]. EBV, which was positive in three patients from our series, may manifest complex immune patterns that are not unique to this setting. Lentiviruses can induce a wide range of immune responses and lead to the development of antibodies, which have been tested in animal models. Autoimmune CNS disorders are largely underdiagnosed, and despite many reports describing autoimmunity spectra in PLWH, none of them include CNS complications. To our knowledge, few cases of autoimmune anti-CNS anti-NMDAr acute encephalitis have been described in PLWH, while only one case of anti-NSA+ encephalitis with psychotic onset has been reported [3,4,5,6]. NMDAR hyperactivation has been described in HIV infection, and gp120, a major component of HIV virus, may trigger NMDAr’s dephosphorylation [24]. Reports suggest that AE may be triggered by (or superposed to) herpes simplex infection which is common in such conditions, and the role of EBV is still under debate with some evidence of raised inflammation in its presence [25]. Other possible pathogenetic mechanisms are chronic immune CNS activation, the role of other pathogens in enhancing auto-antibody production, and the presence of a leaking BBB, as already discussed.
Several implications may deserve further discussion given the general high proportion of undiagnosed encephalitis. Classical findings help in the diagnostic process, but up to 30–40% of encephalitis cases remain of unknown origin despite considering autoimmune encephalitis (AE) as a major differential diagnosis, and AE may be more common than viral encephalitis when considering specifying settings [26,27]. Additional explanations could relate immune activation to viral products not normally found in the CSF of patients with acute disease or, looking further, potentially a large number of microorganisms could be reclassified as pathogens if a clear relation between their presence and development of acute symptoms were ever proven. New frontiers such as metagenomics may be helpful in combination with a standard strategy to improve highlighting credible etiological pathogens, but experience is still poor, and frequently, results of uncertain significance endure [28]. Nevertheless, it remains an attractive tool whose use is nowadays difficult to access due to cost and availability. To date, we lack working and reliable markers to test for additional diagnostics.
Moreover, it is important to stress how crucial this differential diagnosis could be in the absence of the recognition of classical pathogens and implications for early treatment at large, following the example of other inflammatory diseases and the potential impact on neurological sequelae when a more aggressive diagnostic path and management are delayed [29,30]. Immune dysregulation and chronic microglial activation, as already highlighted, can explain only part of these pathological patterns. The possibility of intervening promptly with immunomodulatory agents in order to ameliorate prognosis and sequelae may be attractive, specifically in cases where CD8+ or auto-antibody-mediated encephalitis is suspected [7,8,30].
This study has several limitations: its retrospective nature and the heterogeneity and diversity of symptom onset and patient characteristics, in particular with regard to treated and untreated patients. In some cases, uncertainty in the ultimate diagnosis remains. Moreover, we could not test myelin oligodendrocyte glycoprotein (MOG) antibodies. Finally, due to limited power, further subgroup analysis was not feasible. More studies are needed to better describe the spectrum and prognosis of autoimmune CNS diseases in PLWH

5. Conclusions

In conclusion, our series highlights the importance of considering autoimmune mechanisms in the management of PLWH presenting with acute and sub-acute CNS symptoms. Including unconventional autoimmune encephalitis among the causes of acute and subacute CNS manifestations should be considered in the diagnostic workup.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/microorganisms12091758/s1. Table S1: The table describes additional clinical and radiological characteristics of patients included in the analysis. HBV: hepatitis B virus; HBeAg: hepatitis B e antigen; HCV: hepatitis C virus; HSV: herpes simplex virus; PML: progressive multifocal leukoencephalopathy; IRIS: immune reconstitution inflammatory syndrome; PCNSL: primary central nervous system lymphoma; D: dorsal.

Author Contributions

Conceptualization, G.S., A.C., D.I. and G.D.P.; methodology, G.S., A.C., C.A., M.T. and G.D.P.; software, G.S. and A.C.; validation, M.T., C.A., G.D.P. and D.I.; formal analysis, G.S., A.C., C.A. and M.T.; investigation, G.S. and A.C.; resources, A.C., G.D.P. and D.I.; data curation, G.S., A.C. and M.T.; writing—original draft preparation, G.S.; writing—review and editing, G.S., A.C. and M.T.; visualization, C.A., G.S., and A.C.; supervision, D.I., A.C., G.D.P. and M.T.; project administration, A.C.; funding acquisition, A.C., G.D.P. and D.I. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Italian Ministry of Health “Fondi Ricerca Corrente” to IRCCS Sacro Cuore Don Calabria Hospital.

Data Availability Statement

The data that support the findings of this study are available from the local Ethics Committee (Comitato Interaziendale di Orbassano, “PRODIN study”, protocol number 103). The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Williams, M.E.; Stein, D.J.; Joska, J.A.; Naudé, P.J.W. Cerebrospinal fluid immune markers and HIV-associated neurocognitive impairments: A systematic review. J. Neuroimmunol. 2021, 358, 577649. [Google Scholar] [CrossRef]
  2. Ulfhammer, G.; Edén, A.; Mellgren, Å.; Fuchs, D.; Zetterberg, H.; Hagberg, L.; Nilsson, S.; Yilmaz, A.; Gisslén, M. Persistent central nervous system immune activation following more than 10 years of effective HIV antiretroviral treatment. AIDS 2018, 32, 2171–2178. [Google Scholar] [CrossRef] [PubMed]
  3. Cunill, V.; Arboleya, S.; Jiménez, M.d.L.R.; Campins, A.; Herbera, P.; Mestre, L.; Clemente, A.; Barceló, M.I.; Leyes, M.; Canellas, F.; et al. Neuronal surface antibodies in HIV-infected patients with isolated psychosis. J. Neuroimmunol. 2016, 301, 49–52. [Google Scholar] [CrossRef]
  4. Haneche, F.; Demeret, S.; Psimaras, D.; Katlama, C.; Pourcher, V. An anti-NMDA receptor encephalitis mimicking an HIV encephalitis. Clin. Immunol. 2018, 193, 10–11. [Google Scholar] [CrossRef] [PubMed]
  5. Moloney, P.B.; Hutchinson, S.; Heskin, J.; Mulcahy, F.; Langan, Y.; Conlon, N.P.; Linas, B.P.; Takahashi, C.; Cervantes-Arslanian, A.M. Possible N-methyl-D-aspartate receptor antibody-mediated encephalitis in the setting of HIV cerebrospinal fluid escape. J. Neurol. 2020, 267, 1348–1352. [Google Scholar] [CrossRef]
  6. Atchayaram, N.; Nagabushana, D.; Nishamol, T.; Bhattacharya, K.; Saini, J.; Chowdary, R.; Mahadevan, A.; Polavarapu, K. Anti-N-methyl-D-aspartate-receptor Encephalitis as a Harbinger of Pediatric HIV Infection. J. Pediatr. Neurosci. 2021, 16, 327–331. [Google Scholar] [CrossRef] [PubMed]
  7. Santana, L.M.; Valadares, E.A.; Ferreira-Júnior, C.U.; Santos, M.F.; Albergaria, B.-H.; Rosa-Júnior, M. CD8 + T-lymphocyte Encephalitis: A Systematic Review. AIDS Rev. 2020, 22, 112–122. [Google Scholar] [CrossRef] [PubMed]
  8. Wood, A.C.; Parker, R.; Allinson, K.; Scoffings, D. CD8 encephalitis presenting as autoimmune encephalitis in HIV-1 infection. BMJ Case Rep. 2022, 15, e246290. [Google Scholar] [CrossRef]
  9. Gold, J.; Goldacre, R.; Maruszak, H.; Giovannoni, G.; Yeates, D.; Goldacre, M. HIV and lower risk of multiple sclerosis: Beginning to unravel a mystery using a record-linked database study. J. Neurol. Neurosurg. Psychiatry 2015, 86, 9–12. [Google Scholar] [CrossRef]
  10. Iordache, L.; Bengoufa, D.; Taulera, O.; Rami, A.; Lascoux-Combe, C.; Day, N.; Parrinello, M.; Sellier, P.-O.; Molina, J.-M.; Mahr, A. Nonorgan-specific autoantibodies in HIV-infected patients in the HAART era. Medicine 2017, 96, e6230. [Google Scholar] [CrossRef]
  11. Roszkiewicz, J.; Smolewska, E. Kaleidoscope of autoimmune diseases in HIV infection. Rheumatol. Int. 2016, 36, 1481–1491. [Google Scholar] [CrossRef] [PubMed]
  12. Stratton, R.; Slapak, G.; Mahungu, T.; Kinloch-de Loes, S. Autoimmunity and HIV. Curr. Opin. Infect. Dis. 2009, 22, 49–56. [Google Scholar] [CrossRef] [PubMed]
  13. Sempere, J.M.; Soriano, V.; Benito, J.M. T regulatory cells and HIV infection. AIDS Rev. 2007, 9, 54–60. [Google Scholar]
  14. Ryu, H.S.; Lee, S.Y.; Park, D.H.; Lee, J.M. A Case of Paraneoplastic Neurological Syndrome Expressing Dual Antineuronal Antibodies: Anti-Hu and Recoverin. Ann. Indian Acad. Neurol. 2020, 23, 133–135. [Google Scholar]
  15. Johnson, T.P.; Nath, A. Biotypes of HIV-associated neurocognitive disorders based on viral and immune pathogenesis. Curr. Opin. Infect. Dis. 2022, 35, 223–230. [Google Scholar] [CrossRef]
  16. Bataller, L.; Wade, D.F.; Graus, F.; Stacey, H.D.; Rosenfeld, M.R.; Dalmau, J. Antibodies to Zic4 in paraneoplastic neurologic disorders and small-cell lung cancer. Neurology 2004, 62, 778–782. [Google Scholar] [CrossRef] [PubMed]
  17. Armangue, T.; Spatola, M.; Vlagea, A.; Mattozzi, S.; Cárceles-Cordon, M.; Llufriu, S.; Muchart, J.; Erro, M.E.; Abraira, L.; Moris, G.; et al. Frequency, symptoms, risk factors, and outcomes of autoimmune encephalitis after herpes simplex encephalitis: A prospective observational study and retrospective analysis. Lancet Neurol. 2018, 17, 760–772. [Google Scholar] [CrossRef]
  18. Chen, N.C.; Partridge, A.T.; Sell, C.; Torres, C.; Martín-García, J. Fate of microglia during HIV-1 infection: From activation to senescence? Glia 2017, 65, 431–446. [Google Scholar] [CrossRef]
  19. Spudich, S.S. Immune activation in the central nervous system throughout the course of HIV infection. Curr. Opin. HIV AIDS 2016, 11, 226–233. [Google Scholar] [CrossRef]
  20. Mastrangelo, A.; Turrini, F.; de Zan, V.; Caccia, R.; Gerevini, S.; Cinque, P. Symptomatic cerebrospinal fluid escape. AIDS 2019, 33 (Suppl. S2), S159–S169. [Google Scholar] [CrossRef]
  21. Watson, C.; Busovaca, E.; Foley, J.M.; Allen, I.E.; Schwarz, C.G.; Jahanshad, N.; Nir, T.M.; Esmaeili-Firidouni, P.; Milanini, B.; Rosen, H.; et al. White matter hyperintensities correlate to cognition and fiber tract integrity in older adults with HIV. J. Neurovirol. 2017, 23, 422–429. [Google Scholar] [CrossRef] [PubMed]
  22. Rasmussen, H.B.; Perron, H.; Clausen, J. Do endogenous retroviruses have etiological implications in inflammatory and degenerative nervous system diseases? Acta Neurol. Scand. 1993, 88, 190–198. [Google Scholar] [CrossRef] [PubMed]
  23. Bjornevik, K.; Cortese, M.; Healy, B.C.; Kuhle, J.; Mina, M.J.; Leng, Y.; Elledge, S.J.; Niebuhr, D.W.; Scher, A.I.; Munger, K.L.; et al. Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science 2022, 375, 296–301. [Google Scholar] [CrossRef] [PubMed]
  24. Ru, W.; Tang, S.J. HIV-1 gp120Bal down-Regulates Phosphorylated NMDA Receptor Subunit 1 in Cortical Neurons via Activation of Glutamate and Chemokine Receptors. J. Neuroimmune Pharmacol. 2016, 11, 182–191. [Google Scholar] [CrossRef] [PubMed]
  25. Lupia, T.; Milia, M.G.; Atzori, C.; Gianella, S.; Audagnotto, S.; Imperiale, D.; Mighetto, L.; Pirriatore, V.; Gregori, G.; Lipani, F.; et al. Presence of Epstein–Barr virus DNA in cerebrospinal fluid is associated with greater HIV RNA and inflammation. AIDS 2020, 34, 373–380. [Google Scholar] [CrossRef]
  26. Singh, T.D.; Fugate, J.E.; Rabinstein, A.A. The spectrum of acute encephalitis: Causes, management, and predictors of outcome. Neurology 2015, 84, 359–366. [Google Scholar] [CrossRef]
  27. Gable, M.S.; Sheriff, H.; Dalmau, J.; Tilley, D.H.; Glaser, C.A. The frequency of autoimmune N-methyl-D-aspartate receptor encephalitis surpasses that of individual viral etiologies in young individuals enrolled in the California Encephalitis Project. Clin. Infect. Dis. 2012, 54, 899–904. [Google Scholar] [CrossRef]
  28. Brown, J.R.; Bharucha, T.; Breuer, J. Encephalitis diagnosis using metagenomics: Application of next generation sequencing for undiagnosed cases. J. Infect. 2018, 76, 225–240. [Google Scholar] [CrossRef]
  29. Pender, M.P.; Csurhes, P.A.; Smith, C.; Douglas, N.L.; Neller, M.A.; Matthews, K.K.; Beagley, L.; Rehan, S.; Crooks, P.; Hopkins, T.J.; et al. Epstein-Barr virus-specific T cell therapy for progressive multiple sclerosis. JCI Insight. 2018, 3, e124714, Erratum in JCI Insight. 2020, 5, e144624. [Google Scholar] [CrossRef]
  30. Abboud, H.; Probasco, J.C.; Irani, S.; Ances, B.; Benavides, D.R.; Bradshaw, M.; Christo, P.P.; Dale, R.C.; Fernandez-Fournier, M.; Flanagan, E.P.; et al. Autoimmune Encephalitis Alliance Clinicians Network. Autoimmune encephalitis: Proposed best practice recommendations for diagnosis and acute management. J. Neurol. Neurosurg. Psychiatry 2021, 92, 757–768. [Google Scholar] [CrossRef]
Figure 1. Anti-oligodendrocytes immunofluorescence on monkey cerebellar tissue samples exposed to the CSF of our patient (n. 5) collected at the onset of her symptoms. The diffuse positivity of the fluorescence among the white matter (A) and molecular (B) and granular layers (C) of the grey matter revealed the presence of anti-oligodendrocyte antibodies in the CSF of our patient. (Purkinje cells shown in D). This pattern is not specific.
Figure 1. Anti-oligodendrocytes immunofluorescence on monkey cerebellar tissue samples exposed to the CSF of our patient (n. 5) collected at the onset of her symptoms. The diffuse positivity of the fluorescence among the white matter (A) and molecular (B) and granular layers (C) of the grey matter revealed the presence of anti-oligodendrocyte antibodies in the CSF of our patient. (Purkinje cells shown in D). This pattern is not specific.
Microorganisms 12 01758 g001
Figure 2. The figure demonstrates a perisomatic pattern, positive upon immunofluorescence for anti-CNS antibodies. Purkinje cells show fluorescence at the axonal level. Figure from patient case n. 8. This pattern is not specific.
Figure 2. The figure demonstrates a perisomatic pattern, positive upon immunofluorescence for anti-CNS antibodies. Purkinje cells show fluorescence at the axonal level. Figure from patient case n. 8. This pattern is not specific.
Microorganisms 12 01758 g002
Figure 3. The figure on the left side demonstrates a negative control; the figure in the middle demonstrates a pattern for anti-Tr positivity: fluorescent in the cytoplasm of Purkinje cells. The figure on the right side demonstrates a pattern for anti-Hu positivity: fluorescent neuronal nuclei (both in the molecular layer, the granular layer, and Purkinje nuclei).
Figure 3. The figure on the left side demonstrates a negative control; the figure in the middle demonstrates a pattern for anti-Tr positivity: fluorescent in the cytoplasm of Purkinje cells. The figure on the right side demonstrates a pattern for anti-Hu positivity: fluorescent neuronal nuclei (both in the molecular layer, the granular layer, and Purkinje nuclei).
Microorganisms 12 01758 g003
Table 1. Patients and main features. HIV: human immunodeficiency virus; CSF: cerebrospinal fluid; EEG: electroencephalography; HAART: highly active antiretroviral therapy; M: male; F: female; N/A: not assessed; BBBp: blood–brain barrier permeability; VDRL: venereal disease research laboratory test; CSAR: CSF to serum albumin ratio; PCNSL: primary central nervous system lymphoma; PML: progressive multifocal leukoencephalopathy; IRIS: immune reconstitution inflammatory syndrome.
Table 1. Patients and main features. HIV: human immunodeficiency virus; CSF: cerebrospinal fluid; EEG: electroencephalography; HAART: highly active antiretroviral therapy; M: male; F: female; N/A: not assessed; BBBp: blood–brain barrier permeability; VDRL: venereal disease research laboratory test; CSAR: CSF to serum albumin ratio; PCNSL: primary central nervous system lymphoma; PML: progressive multifocal leukoencephalopathy; IRIS: immune reconstitution inflammatory syndrome.
Patient n°AgeGenderImaging PatternOnset SymptomsCD4+HIV RNA Plasma (cp/mL)HIV RNA CSF (cp/mL)EBV DNA CSF (cp/mL)CSF Ab PatternEEGAltered NeuromarkersHAARTFinal Diagnosis
152MDemyelinatingSensory and dysautonomic disorders28566794466N/ASynapticNormal14.3.3, Neopterin, BBBp, CSAR, and IgG indexTreatedMyelitis
255MNormalAsymptomatic, control in previous neurolue, and VDRL serum positivity436NegativeNegativeN/ANuclearNormalNoneNaïveNone
328MInflammatory/demyelinatingConfusion25116,1971064N/ANuclearNormal14.3.3, Tau, and Beta AmyloidNaïveBalo concentric sclerosis
460FDemyelinatingSensory and extrapyramidal disorders111Negative16054Nuclear and SynpaticNormalIntrathecal synthesisTreatedPML-IRIS
550FInflammatory/demyelinatingExtrapiramidal disorders and vertigo516Negative756682AspecificNormalIntrathecal synthesis, CSAR, BBBp, Neopterin, and Beta AmyloidTreatedCNS viral escape
641MNormalCefalea and sensory disorders14739571NegativeNuclearNormalNoneNaïveHAART neurotoxicity
741MDemyelinatingVertigo, sensory and extrapyramidal disorders, and ataxia1936,67616,030NegativeSynapticNormalNeopterin, BBBp, and CSARNaïvePML
844FInflammatoryDysautonomic and extrapuramidal disorders, confusion, and agitation320NegativeNegative315AspecificAlteredBBBp and intrathecal synthesisTreatedNone
960FInflammatoryMovement disorders and ataxia42988,982330,273N/AAspecificAlteredIntrathecal synthesis, BBBp, Neopterin, and Beta AmyloidTreatedCSF viral escape
1044MDemyelinating, inflammatory, and mass effectDysautonomic, sensory, and motor disorders37758,98823,762N/ASynapticNormalTau, Neopterin, 14.3.3, and BBBpNaïvePCNSL
1150MAtrophicExtrapyramidal and movement disorders, ataxia, and confusion379NegativeNegativeNegativeNuclearNormalNoneTreatedNone
1255MNormalSensory disorders7151450330,000NegativeNuclear, Recoverin, and Zic 4 on BlotNormalBBBp and Intrathecal synthesisNaïveNone
Table 2. Antibody patterns and neuromarkers. In black, the patterns with corresponding neuromarkers outside the normal range of values are highlighted.
Table 2. Antibody patterns and neuromarkers. In black, the patterns with corresponding neuromarkers outside the normal range of values are highlighted.
Altered Neuromarkers and Correspondances with Antibody PatternsNuclearSynapticAspecificZic4Recoverine
Neopterin
1–42 Beta Amyloid
BBB damage
Intrathecal synthesis
CSAR
Tau
FTau
14.3.3.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Stroffolini, G.; Atzori, C.; Imperiale, D.; Trunfio, M.; Di Perri, G.; Calcagno, A. Central Nervous System Disorders with Auto-Antibodies in People Living with HIV. Microorganisms 2024, 12, 1758. https://doi.org/10.3390/microorganisms12091758

AMA Style

Stroffolini G, Atzori C, Imperiale D, Trunfio M, Di Perri G, Calcagno A. Central Nervous System Disorders with Auto-Antibodies in People Living with HIV. Microorganisms. 2024; 12(9):1758. https://doi.org/10.3390/microorganisms12091758

Chicago/Turabian Style

Stroffolini, Giacomo, Cristiana Atzori, Daniele Imperiale, Mattia Trunfio, Giovanni Di Perri, and Andrea Calcagno. 2024. "Central Nervous System Disorders with Auto-Antibodies in People Living with HIV" Microorganisms 12, no. 9: 1758. https://doi.org/10.3390/microorganisms12091758

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop