Severe Neuro-COVID is associated with peripheral immune signatures, autoimmunity and neurodegeneration: a prospective cross-sectional study

Manina M Etter, Tomás A Martins, Laila Kulsvehagen, Elisabeth Pössnecker, Wandrille Duchemin, Sabrina Hogan, Gretel Sanabria-Diaz, Jannis Müller, Alessio Chiappini, Jonathan Rychen, Noëmi Eberhard, Raphael Guzman, Luigi Mariani, Lester Melie-Garcia, Emanuela Keller, Ilijas Jelcic, Hans Pargger, Martin Siegemund, Jens Kuhle, Johanna Oechtering, Caroline Eich, Alexandar Tzankov, Matthias S Matter, Sarp Uzun, Özgür Yaldizli, Johanna M Lieb, Marios-Nikos Psychogios, Karoline Leuzinger, Hans H Hirsch, Cristina Granziera, Anne-Katrin Pröbstel, Gregor Hutter, Manina M Etter, Tomás A Martins, Laila Kulsvehagen, Elisabeth Pössnecker, Wandrille Duchemin, Sabrina Hogan, Gretel Sanabria-Diaz, Jannis Müller, Alessio Chiappini, Jonathan Rychen, Noëmi Eberhard, Raphael Guzman, Luigi Mariani, Lester Melie-Garcia, Emanuela Keller, Ilijas Jelcic, Hans Pargger, Martin Siegemund, Jens Kuhle, Johanna Oechtering, Caroline Eich, Alexandar Tzankov, Matthias S Matter, Sarp Uzun, Özgür Yaldizli, Johanna M Lieb, Marios-Nikos Psychogios, Karoline Leuzinger, Hans H Hirsch, Cristina Granziera, Anne-Katrin Pröbstel, Gregor Hutter

Abstract

Growing evidence links COVID-19 with acute and long-term neurological dysfunction. However, the pathophysiological mechanisms resulting in central nervous system involvement remain unclear, posing both diagnostic and therapeutic challenges. Here we show outcomes of a cross-sectional clinical study (NCT04472013) including clinical and imaging data and corresponding multidimensional characterization of immune mediators in the cerebrospinal fluid (CSF) and plasma of patients belonging to different Neuro-COVID severity classes. The most prominent signs of severe Neuro-COVID are blood-brain barrier (BBB) impairment, elevated microglia activation markers and a polyclonal B cell response targeting self-antigens and non-self-antigens. COVID-19 patients show decreased regional brain volumes associating with specific CSF parameters, however, COVID-19 patients characterized by plasma cytokine storm are presenting with a non-inflammatory CSF profile. Post-acute COVID-19 syndrome strongly associates with a distinctive set of CSF and plasma mediators. Collectively, we identify several potentially actionable targets to prevent or intervene with the neurological consequences of SARS-CoV-2 infection.

Conflict of interest statement

I.J. has received speaker honoraria or unrestricted grants from Biogen Idec and Novartis and has served as an advisor for Alexion, Biogen, Bristol Myers Squibb, Celgene, Janssen-Cilag, Neuway, Merck, Novartis, Roche and Sanofi Genzyme; none of these are related to this study. G.H. has equity in and is a co-founder of Incephalo Inc. A-K.P. has received speaker honoraria or research/travel support from Roche and Biogen all used for research. The remaining authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. CONSORT diagram and schemes illustrating…
Fig. 1. CONSORT diagram and schemes illustrating the project design.
a Consort flow diagram. Patients who tested positive for SARS-CoV-2 were assessed for eligibility (n = 310), of which 269 declined to participate and 1 failed to meet inclusion criteria. Enrolled patients (n = 40) were allocated to different severity classes of Neuro-COVID according to Fotuhi et. al. with 18 in class I, 7 in class II and 15 in class III. Schemes illustrating the study design:b Paired cerebrospinal fluid (CSF) and plasma samples were collected from 40 COVID-19 patients. Paired CSF and plasma samples from healthy (n = 25) and non-MS inflammatory neurological disease controls (n = 25) were retrospectively obtained. c In 37 of the COVID-19 patients, a contrast-enhanced MRI or CT scan was conducted and evaluated by a board-certified neuroradiologist. d Brain volumetric analysis was performed in 35 COVID-19 patients. This cohort included 22 patients of the main study cohort from whom Magnetization prepared—rapid gradient echo (MPRAGE) pulse sequences and paired CSF and plasma samples were obtained (light blue) and an additional 13 patients who underwent brain MRI during COVID-19 infection (dark blue). A cohort of 36 healthy age and sex-matched individuals served as the control group. b–d Created with Biorender.com.
Fig. 2. Routine inflammatory CSF parameters and…
Fig. 2. Routine inflammatory CSF parameters and B-cell response in Neuro-COVID patients.
a Box plot representation of routine cerebrospinal fluid (CSF) parameters including glucose (mmol/L), glucose CSF/plasma ratio (log2 scale), lactate (mmol/L), albumin CSF/plasma ratio (log2 scale), total protein (mg/L, log10 scale) and leukocytes (cell count × 106/L − log10 scale) (center line at the median, upper bound at 75th percentile, lower bound at 25th percentile) with whiskers at minimum and maximum values. Patients that were excluded from the analysis are indicated in violet. Statistics: The data for each parameter, except the leukocyte count, was marginalized on sex and age. Statistics: The data for each parameter was marginalized on sex and age. Two-sided Mann–Whitney-U test was applied, p value correction was performed with Benjamin-Hochberg (BH)-procedure (adjusted p: *<0.05; **<0.01; ***<0.001; if not otherwise indicated: not significant). b Non-metric multidimensional scaling (NMDS) plots of merged anti-BSA-, anti-dsDNA- and anti-gut bacteria antibodies in the CSF (left plot) and plasma (right plot) of each class. Data points are colored by category. Each point represents one patient. Patients with a more similar antibody composition are closer together and, conversely, those that were more dissimilar depict a greater distance. The ellipses represent the 95% confidence interval within subgroups. c Box plot representation of CSF levels (OD450; optical density at 450 nm) of anti-BSA, anti-dsDNA-and anti-gut bacteria (RePOOPulate)-IgG/IgA per patient and control group. Patients with anti-SARS-CoV-2 Spike protein antibodies in the CSF indicated in red, those with intrathecal IgG or IgA production in orange, respectively. Statistics: The data for each parameter except the IgG data was marginalized on sex and age. Two-sided Mann–Whitney-U test was applied, p value correction was performed with Benjamin-Hochberg (BH)-procedure (adjusted p: *<0.05; **<0.01; ***<0.001; if not otherwise indicated: not significant). Source data of (a, c) are provided as a Source Data file.
Fig. 3. B-cell receptor sequencing reveals a…
Fig. 3. B-cell receptor sequencing reveals a more specific antibody response and a higher B-cell clone number in class I patients, whereas class III patients depicted a more diverse response due to polyclonality.
a Box plot representation of B-cell clone number in class I (n = 4) and class III patients (n = 4) respectively. B-cell clone number was significantly higher in the blood of class I patients compared to class III (Mann–Whitney-U test: p: =0.02857, spearman’s rank correlation test: R = 0.738, p = 0.04583). Class I patients (n = 4) had higher plasma IgG levels which correlated with the number of B-cell clones in the blood. b Box plot representation of Shannon Diversity and Evenness in class I (n = 4) and class III (n = 4) patients. Diversity analysis showed that there was no significant difference between class I and II. c Representation of all B-cell clones from each class clustered together and ranked according to their total frequency (relative abundance) in the immune repertoire of a patient. Then, top 10 highest frequency clones from each class selected, grouped together and ranked again according to the total frequency. As indicated in the dot plot, the clones making up more than 5% of BCR immune repertoire belonged to class I patients (n = 4). Only two B-cell clones of class III patients were in the top 10. d B-cell immune repertoire of both classes (class I and III patients: n = 8) showed a similar Gaussian distribution in CDR3 nucleotide length and same median CDR3 nucleotide length. Each bar represents the number of B cells having a specific CDR3 nucleotide length. Boxplots indicate median and lower (25th)–upper (75th) quartile and whiskers show the minimum-maximum values. Each dot represents individual samples. Statistics: Two-sided Wilcoxon rank-sum test and spearman’s rank correlation were applied (adjusted p: *<0.05; if not otherwise indicated: not significant, R Spearman correlation coefficient). BCR B-cell receptor.
Fig. 4. Neuro-COVID patients display a vigorous…
Fig. 4. Neuro-COVID patients display a vigorous peripheral immune response and specific CSF alterations including analytes with high predictive value for class III development and strong CSF-plasma correlation.
a Rose plots representing Z-scores of marginalized normalized protein expression (NPX) of 192 soluble proteins in CSF and plasma. For better visualization, analytes have been grouped into ‘inflammatory’ (left panels) and ‘neurological’ (right panels) proteins. b Non-metric multidimensional scaling (NMDS) plots of 192 examined soluble proteins in CSF and plasma. Each patient is presented by one dot, and colored according to healthy controls, CNS inflammatory controls or Neuro-COVID class I-III. The ellipses represent the 95% confidence interval within subgroups. Source data are provided as a Source Data file.
Fig. 5. Individual CSF and plasma analytes…
Fig. 5. Individual CSF and plasma analytes discriminating different groups.
Box plot representations (center line at the median, upper bound at 75th percentile, lower bound at 25th percentile) of marginalized normal protein expression (NPX) of individual analytes significantly discriminating selected groups with whiskers at minimum and maximum values. Each dot represents one participant a Plasma, increasing NPX from class I (n = 18) to III (n = 15), and higher than in controls (n = 50): IL-6 (class III vs. I: p = 0.007, class III vs. infl. ctrl: p = 0.001), IL-8 (class III vs. I: p = 0.003, class III vs. infl. ctrl: p = 0.0002), HGF (class III vs. I: p = 0.04, class III vs. infl. ctrl: p = 0.0007), VEGFA (class III vs. I: p = 0.01, class III vs. infl. ctrl: p = 0.0005), EN-RAGE (class III vs. I: p = 0.003, class III vs. infl. ctrl: p = 0.0002), TNFRSF12A (class III vs. I: p = 0.006, class III vs. infl. ctrl: p = 0.002), PD-L1 (class III vs. I: p = 0.04, class III vs. infl. ctrl: p = 0.002), CCL23 (class III vs. infl. ctrl: p = 0.01), EZR (class III vs. infl. ctrl: p = 0.002), TNFRSF11B (class III vs. infl. ctrl: p = 0.049). b Plasma, decreasing NPX from class I (n = 18) to III (n = 15), higher in controls (n = 50) than in COVID-19 patients (n = 40): BMP-4, CLEC10A, CNTN5, GDF-8, NTRK2, GDNFRalpha, ROBO2. c Plasma, class-independent, higher NPX in COVID-19 (n = 35) than in controls (n = 50): 4E-BP1 (class III vs infl. ctrl: p = 0.007). d Plasma, higher NPX in COVID-19 (n = 40) than in controls (n = 50), decreasing from class I (n = 18) to III (n = 15): HAGH (class III vs. I: p = 0.008). e Cerebrospinal fluid (CSF), increasing NPX from class I (n = 16) to III (n = 14): IL-8 (class III vs. I: p = 0.012), MSR1 (class III vs. I: p = 0.016), 4E-BP1 (class III vs. I: p = 0.02), CD200R1 (class III vs. I: p = 0.04), TNFRSF12A (class III vs. I: p = 0.008), EZR (class III vs. I: p = 0.01). f CSF, increasing NPX from class I (n = 16) to III (n = 14), and higher in class III than in inflammatory controls (n = 25): TNFRSF11B (class III vs. I: p = 0.04, class III vs. infl. ctrl: p = 0.02). Source data are provided as a Source Data file. Statistics (af): statistical significance was calculated using two-sided Mann–Whitney-U test and p values were adjusted using Benjamin-Hochberg (BH)-procedure (p: *<0.05; **<0.01; ***<0.001, if not otherwise indicated: not significant).
Fig. 6. Specific CSF and plasma analytes…
Fig. 6. Specific CSF and plasma analytes correlate with COVID-19 severity and have predictive value for class III development.
a Venn diagram representing cerebrospinal fluid (CSF) and plasma mediators associated with COVID-19 severity assessed with the WHO progression scale. The association of protein sets and COVID-19 severity was assessed using a complement of four models (ordinal backward, ordinal-forward, best linear, most-regularized ordinal). Results of each model were finally cross-referenced to provide robust data sets of mediators associated with severe COVID-19. Plasma MSR1 and CSF TNFRSF12A and IL-8 represent the most robust set of proteins associated with COVID-19 severity (high association in each model used). Plasma IL-8 and IL-6, TNFRSF11B and EZR CSF levels depicted strong association using two models, whereas plasma 4E-BP1 and CSF PD-L1, BMP-4, CLEC10A and ROBO2 were strongly associated with COVID-19 severity in one model only. b ROC-AUC analysis of class I and II vs. class III. Five predictive plasma markers, including IL-8, EN-RAGE, TNFRSF12A, MCP-3, and 4 CSF markers, including 4E-BP1, EZR, TNFRSF12A, MSR1, emerged for the prediction of class III development. The Y-axis represents the sensitivity, the X-axis represents the 1-specificity (represented for IL-8, plasma). The names of relevant proteins in the study are compiled in Supplementary Data 4. c Plot representing the ROC-AUC values on the Y-axis vs. the random forest importance score on the X-axis. Relative importance of each single protein is represented by a high random forest importance score. Red points: plasma proteins, light blue points: CSF proteinsd Venn diagram representing CSF and plasma mediators associated with COVID-19 severity assessed with the WHO progression scale (COVID-19 severity) and mediators with high predictive value for class III development (Neuro-COVID severity). Plasma IL-8, CSF TNFRSF12A and CSF EZR depicted a high predictive value for severe COVID-19 and also class III development. lm linear model, RMSE root mean squared error.
Fig. 7. Routine brain imaging, regional GMVs…
Fig. 7. Routine brain imaging, regional GMVs and association with inflammatory CSF parameters.
a–c Conventional brain MRI and CT scans depicting exemplary imaging findings. Scale bar 15 mm (MRI) and 15.4 mm (CT). a Class I: Axial FLAIR images of the same class I patient show multifocal hyperintensities in the right precentral gyrus (top left), semioval center, left frontal cortex (top right), deep white matter, periventricular region (bottom left), right temporal lobe, left parietal white matter (bottom right). b Class II: Axial FLAIR images of a class II patient depict multifocal hyperintensities in the left frontal superior gyrus (top left), white matter of left frontal lobe (top right), left parahippocampal white matter (bottom left), right mesial temporal region (bottom right). c Class III: Axial FLAIR image shows bilateral thalamic hyperintensities (top left). Axial T1-weighted image depicts left middle cerebral artery (M2-segment) enhancement in the insular cistern (top right). Coronal CT scans demonstrate right cerebellar infarction (bottom left) and right temporo-occipital intracerebral hemorrhage (bottom right) (secondary to thrombosis of the right sigmoid sinus). d–f Conventional pre-COVID-19 MRIs (left) and MRIs during COVID-19 (right). d, e Axial DWI of the same class I patient depicts hyperintense signal alterations during COVID-19 (right sided MRI scans, orange arrows) in the left cerebellar hemisphere (d) and frontal juxtacortical region (e). f Axial FLAIR imaging demonstrates bilateral hyperintense signal alterations in the cerebellar peduncles (right sided MRI scan, orange arrows) of a class III patient. g–i Map (g) and 3D view (h, i) of the 16 brain regions with significant correlation values of gray matter volume (GMV) and clinical variables in the Neuro-COVID group after multiple comparison correction (FDR). These regions are represented in different colors on a T1-weighted template. j shows a matrix representing the association significance (significant p-corrected <0.05 in red squares). Associations between regional volumes and clinical measures were assessed using partial correlation, allowing to calculate the linear partial correlation between variables of interest adjusting for different covariates (age, sex, age*sex interaction, MRI magnetic field strength, total intracranial volume (TIV)). Statistical analysis was performed using the JASP software (https://jasp-stats.org/). MRIcroGL software was used to generate this figure (https://www.nitrc.org/projects/mricrogl). Source data of (gj) are provided as a Source Data file. L left, R right, leuk leukocytes, prot protein, albR Albumin CSF-plasma ratio.
Fig. 8. Specific CSF and plasma mediators…
Fig. 8. Specific CSF and plasma mediators have high predictive value to forecast long-COVID.
ROC-AUC analysis of CSF and plasma parameters and long-COVID. The Y-axis represents the sensitivity, the X-axis represents the 1-Specificity (represented for plasma MCP-3). a Assuming an AUC cut-off of >0.75 for single mediators, high MCP-3 and CLM-6, as well as low RGMA plasma levels were predictive for long-COVID. Confusion matrix analysis of predictive plasma proteins revealed high predictive value of a plasma mediator signature consisting of RGMA, EZR, FcRL2 and ST1A1. b Assuming an AUC cut-off of >0.75, three single CSF proteins emerged for the prediction of long-COVID development. Low CSF levels of TRANCE, as well as high TNFRSF9 and IFN-γ levels were the best predictors. Confusion matrix of CSF proteins revealed high predictive power of a mediator pattern composed of TNFRSF9, IFN-γand TRAIL.
Fig. 9. Overview of proposed pathomechanisms leading…
Fig. 9. Overview of proposed pathomechanisms leading to Neuro-COVID.
The proposed main determinants of severe Neuro-COVID are: (1) peripherally induced cytokine derangements, followed by (2) impaired BBB with ingressing polyreactive autoantibodies, resulting in (3) microglia reactivity and neuronal damage. Created with Biorender.com.

References

    1. Schwabenland M, et al. Deep spatial profiling of human COVID-19 brains reveals neuroinflammation with distinct microanatomical microglia-T-cell interactions. Immunity. 2021;54:1594–1610.e11.
    1. Heming M, et al. Neurological Manifestations of COVID-19 Feature T Cell Exhaustion and Dedifferentiated Monocytes in Cerebrospinal Fluid. Immunity. 2021;54:164–175.e6.
    1. Frontera JA, et al. A Prospective Study of Neurologic Disorders in Hospitalized Patients With COVID-19 in New York City. Neurology. 2021;96:e575–e586.
    1. Deigendesch N, et al. Correlates of critical illness-related encephalopathy predominate postmortem COVID-19 neuropathology. Acta Neuropathologica. 2020;140:583–586.
    1. Yang AC, et al. Dysregulation of brain and choroid plexus cell types in severe COVID-19. Nature. 2021;595:565–571.
    1. Fuchs V, et al. Presence of SARS-CoV-2 Transcripts in the Choroid Plexus of MS and Non-MS Patients With COVID-19. Neurology - Neuroimmunology Neuroinflammation. 2021;8:e957.
    1. Pröbstel AK, et al. SARS-CoV-2-specific neuropathology: fact or fiction? Trends in Neurosciences. 2021;44:933–935.
    1. Douaud G, et al. SARS-CoV-2 is associated with changes in brain structure in UK Biobank. Nature. 2022;604:697–707.
    1. Neumann B, et al. Cerebrospinal fluid findings in COVID-19 patients with neurological symptoms. J. Neurol. Sci. 2020;418:117090.
    1. Bellon M, et al. Cerebrospinal Fluid Features in Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Reverse Transcription Polymerase Chain Reaction (RT-PCR) Positive Patients. Clin. Infect. Dis. Publ. Infect. Dis. Soc. Am. 2021;73:e3102–e3105.
    1. Chang SE, et al. New-onset IgG autoantibodies in hospitalized patients with COVID-19. Nat. Commun. 2021;12:5417.
    1. Song E, et al. Divergent and self-reactive immune responses in the CNS of COVID-19 patients with neurological symptoms. Cell Rep. Med. 2021;2:100288.
    1. Fotuhi M, Mian A, Meysami S, Raji CA. Neurobiology of COVID-19. J. Alzheimers Dis. JAD. 2020;76:3–19.
    1. WHO Working Group on the Clinical Characterisation and Management of COVID-19 infection. A minimal common outcome measure set for COVID-19 clinical research. Lancet Infect. Dis. 2020;20:e192–e197.
    1. Chaudhary, N. & Wesemann, D. R. Analyzing Immunoglobulin Repertoires. Front. Immunol.9, 462 (2018).
    1. Yang L, et al. The signal pathways and treatment of cytokine storm in COVID-19. Signal Transduct. Target. Ther. 2021;6:255.
    1. Perreau M, et al. The cytokines HGF and CXCL13 predict the severity and the mortality in COVID-19 patients. Nat. Commun. 2021;12:4888.
    1. Liu Q, et al. Cathepsin C promotes microglia M1 polarization and aggravates neuroinflammation via activation of Ca2+-dependent PKC/p38MAPK/NF-κB pathway. J. Neuroinflammation. 2019;16:10.
    1. Zamanian JL, et al. Genomic analysis of reactive astrogliosis. J. Neurosci. J. Soc. Neurosci. 2012;32:6391–6410.
    1. Lempp FA, et al. Lectins enhance SARS-CoV-2 infection and influence neutralizing antibodies. Nature. 2021;598:342–347.
    1. Kovarik JJ, et al. Identification of Immune Activation Markers in the Early Onset of COVID-19 Infection. Front. Cell. Infect. Microbiol. 2021;11:651484.
    1. Li L, et al. Interleukin-8 as a Biomarker for Disease Prognosis of Coronavirus Disease-2019 Patients. Front. Immunol. 2021;11:602395.
    1. Moshkovits I, et al. CMRF35-like molecule 1 (CLM-1) regulates eosinophil homeostasis by suppressing cellular chemotaxis. Mucosal Immunol. 2014;7:292–303.
    1. Hoel H, et al. Elevated markers of gut leakage and inflammasome activation in COVID-19 patients with cardiac involvement. J. Intern. Med. 2021;289:523–531.
    1. Zhang Y, et al. Potential contribution of increased soluble IL-2R to lymphopenia in COVID-19 patients. Cell. Mol. Immunol. 2020;17:878–880.
    1. Tian, Y. et al. Activation of RARα Receptor Attenuates Neuroinflammation After SAH via Promoting M1-to-M2 Phenotypic Polarization of Microglia and Regulating Mafb/Msr1/PI3K-Akt/NF-κB Pathway. Front. Immunol. 13, 839796 (2022).
    1. Kossmann T, et al. Interleukin-8 Released into the Cerebrospinal Fluid after Brain Injury is Associated with Blood–Brain Barrier Dysfunction and Nerve Growth Factor Production. J. Cereb. Blood Flow. Metab. 1997;17:280–289.
    1. Geiger KD, Stoldt P, Schlote W, Derouiche A. Ezrin Immunoreactivity Reveals Specific Astrocyte Activation in Cerebral HIV. J. Neuropathol. Exp. Neurol. 2006;65:87–96.
    1. Lööv C, Nadadhur AG, Hillered L, Clausen F, Erlandsson A. Extracellular ezrin: a novel biomarker for traumatic brain injury. J. Neurotrauma. 2015;32:244–251.
    1. Sabbatino F, et al. PD-L1 Dysregulation in COVID-19 Patients. Front. Immunol. 2021;12:2198.
    1. Dastidar SG, et al. 4E-BP1 Protects Neurons from Misfolded Protein Stress and Parkinson’s Disease Toxicity by Inducing the Mitochondrial Unfolded Protein Response. J. Neurosci. 2020;40:8734–8745.
    1. Divolis G, et al. Activation of both transforming growth factor-β and bone morphogenetic protein signalling pathways upon traumatic brain injury restrains pro-inflammatory and boosts tissue reparatory responses of reactive astrocytes and microglia. Brain Commun. 2019;1:fcz028.
    1. Ilarregui JM, et al. Macrophage galactose-type lectin (MGL) is induced on M2 microglia and participates in the resolution phase of autoimmune neuroinflammation. J. Neuroinflammation. 2019;16:130.
    1. Yaşar Ş, Çolak C, Yoloğlu S. Artificial Intelligence-Based Prediction of Covid-19 Severity on the Results of Protein Profiling. Comput. Methods Prog. Biomed. 2021;202:105996.
    1. Zhang J-Y, et al. Single-cell landscape of immunological responses in patients with COVID-19. Nat. Immunol. 2020;21:1107–1118.
    1. Maddison DC, Giorgini F. The kynurenine pathway and neurodegenerative disease. Semin. Cell Dev. Biol. 2015;40:134–141.
    1. Callahan V, et al. The Pro-Inflammatory Chemokines CXCL9, CXCL10 and CXCL11 Are Upregulated Following SARS-CoV-2 Infection in an AKT-Dependent Manner. Viruses. 2021;13:1062.
    1. Soriano JB, et al. A clinical case definition of post-COVID-19 condition by a Delphi consensus. Lancet Infect. Dis. 2022;22:e102–e107.
    1. Simhadri VR, Mariano JL, Gil-Krzewska A, Zhou Q, Borrego F. CD300c is an Activating Receptor Expressed on Human Monocytes. J. Innate Immun. 2013;5:389–400.
    1. Yang Y, et al. Plasma IP-10 and MCP-3 levels are highly associated with disease severity and predict the progression of COVID-19. J. Allergy Clin. Immunol. 2020;146:119–127.e4.
    1. Fujita Y, Yamashita T. The roles of RGMa-neogenin signaling in inflammation and angiogenesis. Inflamm. Regen. 2017;37:6.
    1. Guillot F, et al. Transcript analysis of laser capture microdissected white matter astrocytes and higher phenol sulfotransferase 1A1 expression during autoimmune neuroinflammation. J. Neuroinflammation. 2015;12:130.
    1. Jackson TA, Haga CL, Ehrhardt GRA, Davis RS, Cooper MD. FcR-Like 2 Inhibition of B Cell Receptor-Mediated Activation of B Cells. J. Immunol. 2010;185:7405–7412.
    1. Blank, A. E. et al. Tumour necrosis factor receptor superfamily member 9 (TNFRSF9) is up-regulated in reactive astrocytes in human gliomas. Neuropathol Appl Neurobiol.41, e56–67 (2015).
    1. Glasnović A, O’Mara N, Kovačić N, Grčević D, Gajović S. RANK/RANKL/OPG Signaling in the Brain: A Systematic Review of the Literature. Front. Neurol. 2020;11:590480.
    1. Shimamura M, et al. Development of a novel RANKL-based peptide, microglial healing peptide1-AcN (MHP1-AcN), for treatment of ischemic stroke. Sci. Rep. 2018;8:17770.
    1. Bossi F, Bernardi S, Zauli G, Secchiero P, Fabris B. TRAIL Modulates the Immune System and Protects against the Development of Diabetes. J. Immunol. Res. 2015;2015:e680749.
    1. Cartland SP, et al. TRAIL-Expressing Monocyte/Macrophages Are Critical for Reducing Inflammation and Atherosclerosis. iScience. 2019;12:41–52.
    1. Chyuan I-T, Tsai H-F, Liao H-J, Wu C-S, Hsu P-N. An apoptosis-independent role of TRAIL in suppressing joint inflammation and inhibiting T-cell activation in inflammatory arthritis. Cell. Mol. Immunol. 2018;15:846–857.
    1. Pröbstel A-K, et al. Association of antibodies against myelin and neuronal antigens with neuroinflammation in systemic lupus erythematosus. Rheumatol. Oxf. Engl. 2019;58:908–913.
    1. Patiño-Trives AM, et al. Anti-dsDNA Antibodies Increase the Cardiovascular Risk in Systemic Lupus Erythematosus Promoting a Distinctive Immune and Vascular Activation. Arterioscler. Thromb. Vasc. Biol. 2021;41:2417–2430.
    1. Azevedo RB, et al. Covid-19 and the cardiovascular system: a comprehensive review. J. Hum. Hypertens. 2021;35:4–11.
    1. Ryan FJ, et al. Colonic microbiota is associated with inflammation and host epigenomic alterations in inflammatory bowel disease. Nat. Commun. 2020;11:1512.
    1. Kim HS. Do an Altered Gut Microbiota and an Associated Leaky Gut Affect COVID-19 Severity? mBio. 2021;12:e03022–20.
    1. Sun Z, et al. Gut microbiome alterations and gut barrier dysfunction are associated with host immune homeostasis in COVID-19 patients. BMC Med. 2022;20:24.
    1. Pröbstel A-K, et al. Gut microbiota-specific IgA+ B cells traffic to the CNS in active multiple sclerosis. Sci. Immunol. 2020;5:eabc7191.
    1. Rojas OL, et al. Recirculating Intestinal IgA-Producing Cells Regulate Neuroinflammation via IL-10. Cell. 2019;176:610–624.e18.
    1. Kuchroo, V. K. et. al. Dysregulation of immune homeostasis in autoimmune diseases. Nat Med.18, 42–7 (2012).
    1. Leuzinger K, et al. Comparing Immunoassays for SARS-CoV-2 Antibody Detection in Patients with and without Laboratory-Confirmed SARS-CoV-2 Infection. J. Clin. Microbiol. 2021;59:e01381–21.
    1. Wen J, et al. TNF-like weak inducer of apoptosis promotes blood brain barrier disruption and increases neuronal cell death in MRL/lpr mice. J. Autoimmun. 2015;60:40–50.
    1. Stephan D, et al. TWEAK/Fn14 pathway modulates properties of a human microvascular endothelial cell model of blood brain barrier. J. Neuroinflammation. 2013;10:9.
    1. Wood, H. VEGFA mediates blood-brain barrier disruption in Parkinson disease. Nat. Rev. Neurol. 10.1038/s41582-021-00594-6 (2021).
    1. Zhang ZG, et al. VEGF enhances angiogenesis and promotes blood-brain barrier leakage in the ischemic brain. J. Clin. Investig. 2000;106:829–838.
    1. Chen J, et al. Assessment of Advanced Glycation End Products and Receptors and the Risk of Dementia. JAMA Netw. Open. 2021;4:e2033012.
    1. Kook S-Y, Seok Hong H, Moon M, Mook-Jung I. Disruption of blood-brain barrier in Alzheimer disease pathogenesis. Tissue Barriers. 2013;1:e23993.
    1. DePaula-Silva AB, et al. Differential transcriptional profiles identify microglial- and macrophage-specific gene markers expressed during virus-induced neuroinflammation. J. Neuroinflammation. 2019;16:152.
    1. Wardemann H, et al. Predominant autoantibody production by early human B cell precursors. Science. 2003;301:1374–1377.
    1. Gwo, C.-Y., Zhu, D. C. & Zhang, R. Brain White Matter Hyperintensity Lesion Characterization in T2 Fluid-Attenuated Inversion Recovery Magnetic Resonance Images: Shape, Texture, and Potential Growth. Front. Neurosci. 13, 353 (2019).
    1. Nalugo, M., Schulte, L. J., Masood, M. F. & Zayed, M. A. Microvascular Angiopathic Consequences of COVID-19. Front. Cardiovasc. Med. 8, 636843 (2021).
    1. Czaja W, et al. Myostatin regulates pituitary development and hepatic IGF1. Am. J. Physiol. Endocrinol. Metab. 2019;316:E1036–E1049.
    1. Hayashi Y, et al. Myostatin expression in the adult rat central nervous system. J. Chem. Neuroanat. 2018;94:125–138.
    1. Fernández-Castañeda, A. et al. Mild respiratory COVID can cause multi-lineage neural cell and myelin dysregulation. Cell S0092867422007139 10.1016/j.cell.2022.06.008 (2022).
    1. Phetsouphanh DR, et al. Immunological dysfunction persists for 8 months following initial mild-to-moderate SARS-CoV-2 infection. Nature Immunology. 2022;23:210–216.
    1. Bardina SV, et al. Differential Roles of Chemokines CCL2 and CCL7 in Monocytosis and Leukocyte Migration during West Nile Virus Infection. J. Immunol. 2015;195:4306–4318.
    1. Mathey EK, et al. Neurofascin as a novel target for autoantibody-mediated axonal injury. J. Exp. Med. 2007;204:2363–2372.
    1. Pröbstel AK, et al. Antibodies to MOG are transient in childhood acute disseminated encephalomyelitis. Neurology. 2011;77:580–588.
    1. Pröbstel A-K, et al. Anti-MOG antibodies are present in a subgroup of patients with a neuromyelitis optica phenotype. J. Neuroinflammation. 2015;12:46.
    1. Ashburner J, Friston KJ. Diffeomorphic registration using geodesic shooting and Gauss-Newton optimisation. NeuroImage. 2011;55:954–967.

Source: PubMed

3
Subscribe