Sachin P. Gadani’s research while affiliated with Johns Hopkins University and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (19)


NLRX1 expression and inflammatory mediators during acute EAE. (A) Representative Western blot and densitometry analysis of NLRX1 protein expression in the spinal cord of CFA-treated (n = 3) and EAE mice (PID 5, 10, 15 and 20; n = 4–6). (B) WT and Nlrx1-/- mice were immunized with MOG35 − 55 and their paralysis score was evaluated every day until tissue collection for RNA extraction. (C) Gene expression analysis of proinflammatory mediators in the spinal cord of WT (n = 5) and Nlrx1-/- (n = 6) mice at PID14. Gene expression was normalized to CFA treated mice (data not shown). Data presented as mean at ± SEM. Western blot data was compared using one-way ANOVA with Holm-Sidak’s multiple comparison test. Gene expression data was analyzed using Fisher’s LSD test. *p < 0.05, ***p < 0.001, ****p < 0.0001
Clinical disease and anterior visual pathway degeneration in WT and Nlrx1−/− active EAE mice. (A) WT and Nlrx1−/− mice were immunized with subcutaneous injections of MOG35 − 55 in complete Freund’s adjuvant (CFA) and intraperitoneal injection of pertussis toxin (PTX). Control WT and Nlrx1−/− mice received CFA and PTX without MOG35 − 55 (CFA-only controls). Mice were clinically monitored for (B) EAE onset and (C) EAE paralysis severity overtime (n = 22 WT, n = 23 Nlrx1−/−). EAE paralysis score data represents mean ± standard error of the mean (SEM). On day 42 post-immunization (PID 42) mice were euthanized, and eyes and optic nerves were collected for immunofluorescence staining (IF). (D) Retinas were dissected and flat-mounted for IHC and Brn3a + retinal ganglion cells (RGCs) were quantified in 12 regions of interests (ROIs) using a MATLAB semi-automated counting algorithm. Each RGC density/count data point corresponds to an individual mouse, showing the averaged counts from 12 regions in the retina (4 central, 4 middle, and 4 peripheral). (E) Representative IF images of Brn3a⁺ RGCs in flat-mounted retinas from CFA-only and EAE WT and Nlrx1−/− mice. (F) Average RGC density for EAE (n = 20 WT, n = 22 Nlrx1−/−) and control mice, (CFA-controls = 10, 5 WT and 5 Nlrx1−/−). (G) Representative IF images of Brn3a + RGCs and phosphorylated (Ser73) c-Jun (pc-Jun) expression in flat-mounted retinas from WT and Nlrx1−/− EAE mice. (H) Density quantification of total Brn3a + RGCs and pc-Jun + Brna3 + RGCs in WT and Nlrx1−/− EAE mice. (I) Correlation between pc-Jun and Brna3 intensity in RGCs from WT and Nlrx1−/− EAE mice. The mean intensity of each marker in a single RGC was measured in retina using Zeiss ZEN software. (J) Representative IF staining of wholemount retina from an Nlrx1−/− EAE mouse, demonstrating pc-JunlowBrn3ahigh RGCs (red arrows) and pc-JunhighBrn3alow RGCs (white arrows). (K) Representative IF images of neurofilament light chain (NFL) and (L) average NFL percent area positive in the optic nerve of EAE and control mice. RGC density and NFL percent area positive data are represented as mean ± SEM with statistical analysis by Student’s t-test or one-way ANOVA with Dunnett’s T3 multiple comparison test. Linear associations were determined by Pearson correlation analysis. *p < 0.05, ***p < 0.001, ****p < 0.0001
EAE clinical severity and anterior pathway neurodegeneration in Nlrx1−/− opticospinal encephalomyelitis (OSE) mice. (A) OSE mice have T-cells that express a transgenic T-cell receptor (TCRMOG) and B-cells that express transgenic antibodies (IgHMOG), both of which recognize MOG35 − 55 peptide. (B) Paralysis time of onset, incidence, and (C) survival, (D) weight, and (E) EAE paralysis score of all WT (n = 14), Nlrx1+/− (n = 11), and Nlrx1−/− (n = 21) OSE mice and IgHMOG-only mice (n = 26). (F) Area under the curve of EAE paralysis score at peri-peak (days 3–7 post-paralysis onset/PPO) and chronic time points (22–31 PPO). (G) EAE paralysis scores over time and (H) Brn3⁺ RGC density of surviving WT, Nlrx1+/−, and Nlrx1−/− OSE mice (excludes scores of 5.0). Square and circle data points identify male and female mice, respectively. Filled in and open data points identify mice that developed clinical EAE and those that did not, respectively. (I) Brn3a⁺ RGC density in mice with WT TCR (n = 16) and TCRMOG mice with either Nlrx1+/+ (n = 5) or Nlrx1−/− (n = 6). Averages are presented as mean ± SEM with statistical analysis performed by one-way ANOVA with post hoc Dunnett’s multiple comparison test. *p < 0.05, **p < 0.01, ***p < 0.001
T-cell infiltration and glial activation in the optic nerve of Rag−/− and Nlrx1−/− Rag−/− mice after adoptive transfer of activated anti-MOG CD4 + T-cells. (A) CD4 + T-cells are isolated from TCRMOG mice and are proliferated, stimulated, and Th17 polarized in vitro. Subsequently these activated anti-MOG T-cells are injected intraperitoneally into Rag−/− and Nlrx1−/−;Rag−/− recipient mice. (B) Paralysis time of onset and incidence; and (C) paralysis severity by EAE clinical score in Rag\n−/− (n = 25) and Nlrx1−/−Rag−/− (n = 17) recipient mice. Optic nerves were isolated from adoptively transferred mice two weeks after injection of anti-MOG T-cells and proximal, medial, and distal segments were analyzed by IHC for CD3, IBA1, and GFAP. Representative immunofluorescence (IF) images of cross-sectional optic nerves for DAPI and (D) CD3 and (E) IBA1 and GFAP in Rag−/− (n = 17–19) and Nlrx1−/− Rag−/− (n = 14) recipient mice that developed EAE. Optic nerve and region of interest for quantification outlined with a dotted white line. Quantification of (F) CD3⁺ T-cell infiltration, (G) Iba1⁺ cell density, (H) Iba1 mean fluorescent intensity (MFI), and (I) GFAP MFI in Rag−/− and Nlrx1−/− Rag−/− recipient mice. Data are presented as mean ± SEM with statistical analysis performed by Student’s t-test. **p < 0.01
Transcriptome in LPS- and IFNγ-stimulated WT and Nlrx1−/− primary astrocytes. (A) Schematic of the experimental design. Primary astrocyte cultures were derived from WT and Nlrx1−/− mice and were then subsequently stimulated with IFNγ and lipopolysaccharide (LPS) or vehicle (phosphate buffered saline) for 24 h at which point RNA was isolated and analyzed by bulk RNA sequencing. (B) Multidimensional scaling (MDS) plot of the top 1000 highly variable genes from normalized RNA-seq libraries from Nlrx1+/+ and Nlrx1−/− astrocyte cultures treated with or without LPS + IFNγ (C) Barcode plot showing genes associated with TNFα Signaling Via NF-κB (top, red) or genes associated with Oxidative Phosphorylation (bottom. blue). Genes associated with the pathway in question are represented by solid vertical line and are ranked from left to right using the moderated t statistic based on the difference in how Nlrx1−/− cells responded to LPS + IFNγ vs. Nlrx1+/+. Genes toward the left had a higher induction in the WT than in Nlrx1−/− while genes on the right had a higher induction in the Nlrx1−/− mice. Dotted lines represent neutral enrichment, and solid lines represent local enrichment in that area of the ranked list generated by a weighted tricube average in a sliding window, so that a value of 1 represents enrichment if distribution of genes were uniform, and a value of 1.5 would mean a 50% increased enrichment within the window. FDR are false discovery rate values calculated by the competitive gene set test CAMERA. (D and E) Heatmap showing scaled log 2-fold change values (LPS + IFNγ  stimulated vs. unstimulated) where row represents a gene, and column represents an independent biological replicate for a selection of genes associated with (D) TNFα Signaling via NF-κB and (E) Oxidative Phosphorylation. Each column’s genotype is indicated below the heatmap. for each gene and each biological replicate

+2

NLRX1 limits inflammatory neurodegeneration in the anterior visual pathway
  • Article
  • Full-text available

January 2025

·

49 Reads

Journal of Neuroinflammation

·

Matthew D. Smith

·

·

[...]

·

Chronic innate immune activation in the central nervous system (CNS) significantly contributes to neurodegeneration in progressive multiple sclerosis (MS). Using multiple experimental autoimmune encephalomyelitis (EAE) models, we discovered that NLRX1 protects neurons in the anterior visual pathway from inflammatory neurodegeneration. We quantified retinal ganglion cell (RGC) density and optic nerve axonal degeneration, gliosis, and T-cell infiltration in Nlrx1−/− and wild-type (WT) EAE mice and found increased RGC loss and axonal injury in Nlrx1−/− mice compared to WT mice in both active immunization EAE and spontaneous opticospinal encephalomyelitis (OSE) models. To minimize the effects of Nlrx1−/− on peripheral lymphocyte priming during EAE, we performed adoptive transfer experiments, in which activated myelin-specific T cells were transferred into lymphocyte-deficient Rag−/− or Nlrx1−/−Rag−/− mice. In this model, we found more severe microgliosis and astrogliosis in the optic nerve of Nlrx1−/−Rag−/− mice compared to Rag−/− mice, suggesting a regulatory role of NLRX1 in innate immune cells. Transcriptome analysis in primary astrocytes activated with LPS and IFNγ demonstrated that NLRX1 suppresses NF-κB activation and regulates mitochondrial oxidative phosphorylation in inflammatory reactive astrocytes. The novel pharmacologic NLRX1 activators NX-13 and LABP-66 decreased LPS-mediated gene expression of inflammatory cytokines and chemokines in mixed glial cultures. Moreover, treating EAE mice with oral LABP-66, compared to vehicle, after the onset of paralysis resulted in less anterior visual pathway neurodegeneration. These data suggest that pharmacologic NLRX1 activators have the potential to limit inflammatory neurodegeneration. This study highlights that NLRX1 could serve as a promising target for neuroprotection in progressive MS and other neurodegenerative diseases. Further studies are needed to better understand the cell-specific mechanisms underlying the neuroprotective role of NLRX1 in response to inflammation in the CNS.

Download

Spatial transcriptomics of meningeal inflammation reveals inflammatory gene signatures in adjacent brain parenchyma

October 2024

·

31 Reads

eLife

While modern high efficacy disease modifying therapies have revolutionized the treatment of relapsing-remitting multiple sclerosis, they are less effective at controlling progressive forms of the disease. Meningeal inflammation is a recognized risk factor for cortical gray matter pathology which can result in disabling symptoms such as cognitive impairment and depression, but the mechanisms linking meningeal inflammation and gray matter pathology remain unclear. Here, we performed magnetic resonance imaging (MRI)-guided spatial transcriptomics in a mouse model of autoimmune meningeal inflammation to characterize the transcriptional signature in areas of meningeal inflammation and the underlying brain parenchyma. We found broadly increased activity of inflammatory signaling pathways at sites of meningeal inflammation, but only a subset of these pathways active in the adjacent brain parenchyma. Subclustering of regions adjacent to meningeal inflammation revealed the subset of immune programs induced in brain parenchyma, notably complement signaling and antigen processing/presentation. Trajectory gene and gene set modeling analysis confirmed variable penetration of immune signatures originating from meningeal inflammation into the adjacent brain tissue. This work contributes a valuable data resource to the field, provides the first detailed spatial transcriptomic characterization in a model of meningeal inflammation, and highlights several candidate pathways in the pathogenesis of gray matter pathology.



Spatial Transcriptomics of Meningeal Inflammation Reveals Inflammatory Gene Signatures in Adjacent Brain Parenchyma

September 2024

·

7 Reads

·

1 Citation

While modern high efficacy disease modifying therapies have revolutionized the treatment of relapsing-remitting multiple sclerosis, they are less effective at controlling progressive forms of the disease. Meningeal inflammation is a recognized risk factor for cortical grey matter pathology which can result in disabling symptoms such as cognitive impairment and depression, but the mechanisms linking meningeal inflammation and grey matter pathology remain unclear. Here, we performed MRI-guided spatial transcriptomics in a mouse model of autoimmune meningeal inflammation to characterize the transcriptional signature in areas of meningeal inflammation and the underlying brain parenchyma. We found broadly increased activity of inflammatory signaling pathways at sites of meningeal inflammation, but only a subset of these pathways active in the adjacent brain parenchyma. Sub-clustering of regions adjacent to meningeal inflammation revealed the subset of immune programs induced in brain parenchyma, notably complement signaling and antigen processing/presentation. Trajectory gene and gene set modeling analysis confirmed variable penetration of immune signatures originating from meningeal inflammation into the adjacent brain tissue. This work contributes a valuable data resource to the field, provides the first detailed spatial transcriptomic characterization in a model of meningeal inflammation, and highlights several candidate pathways in the pathogenesis of grey matter pathology.


Spatial Transcriptomics of Meningeal Inflammation Reveals Variable Penetrance of Inflammatory Gene Signatures into Adjacent Brain Parenchyma

June 2024

·

10 Reads

While modern high efficacy disease modifying therapies have revolutionized the treatment of relapsing-remitting multiple sclerosis, they are less effective at controlling progressive forms of the disease. Meningeal inflammation is a recognized risk factor for cortical grey matter pathology which can result in disabling symptoms such as cognitive impairment and depression, but the mechanisms linking meningeal inflammation and grey matter pathology remain unclear. Here, we performed MRI-guided spatial transcriptomics in a mouse model of autoimmune meningeal inflammation to characterize the transcriptional signature in areas of meningeal inflammation and the underlying brain parenchyma. We found broadly increased activity of inflammatory signaling pathways at sites of meningeal inflammation, but only a subset of these pathways active in the adjacent brain parenchyma. Sub-clustering of regions adjacent to meningeal inflammation revealed the subset of immune programs induced in brain parenchyma, notably complement signaling and antigen processing/presentation. Trajectory gene and gene set modeling analysis confirmed variable penetration of immune signatures originating from meningeal inflammation into the adjacent brain tissue. This work contributes a valuable data resource to the field, provides the first detailed spatial transcriptomic characterization in a model of meningeal inflammation, and highlights several candidate pathways in the pathogenesis of grey matter pathology.


Neurodegeneration and demyelination in multiple sclerosis

June 2024

·

87 Reads

·

17 Citations

Neuron

Progressive multiple sclerosis (PMS) is an immune-initiated neurodegenerative condition that lacks effective therapies. Although peripheral immune infiltration is a hallmark of relapsing-remitting MS (RRMS), PMS is associated with chronic, tissue-restricted inflammation and disease-associated reactive glial states. The effector functions of disease-associated microglia, astrocytes, and oligodendrocyte lineage cells are beginning to be defined, and recent studies have made significant progress in uncovering their pathologic implications. In this review, we discuss the immune-glia interactions that underlie demyelination, failed remyelination, and neurodegeneration with a focus on PMS. We highlight the common and divergent immune mechanisms by which glial cells acquire disease-associated phenotypes. Finally, we discuss recent advances that have revealed promising novel therapeutic targets for the treatment of PMS and other neurodegenerative diseases.


Demographics and clinical characteristics of the observational cohort
Associations of bile acid metabolite pathways and imaging outcomes
Demographics and clinical characteristics of clinical trial cohort
Bile acid metabolites predict multiple sclerosis progression and supplementation is safe in progressive disease

January 2024

·

77 Reads

·

3 Citations

Background: Bile acid metabolism is altered in multiple sclerosis (MS) and tauroursodeoxycholic acid (TUDCA) supplementation ameliorated disease in mouse models of MS. Methods: Global metabolomics was performed in an observational cohort of people with MS followed by pathway analysis to examine relationships between baseline metabolite levels and subsequent brain and retinal atrophy. A double-blind, placebo-controlled trial, was completed in people with progressive MS (PMS), randomized to receive either TUDCA (2g daily) or placebo for 16 weeks. Participants were followed with serial clinical and laboratory assessments. Primary outcomes were safety and tolerability of TUDCA, and exploratory outcomes included changes in clinical, laboratory and gut microbiome parameters. Results: In the observational cohort, higher primary bile acid levels at baseline predicted slower whole brain, brain substructure and specific retinal layer atrophy. In the clinical trial, 47 participants were included in our analyses (21 in placebo arm, 26 in TUDCA arm). Adverse events did not significantly differ between arms (p=0.77). The TUDCA arm demonstrated increased serum levels of multiple bile acids. No significant differences were noted in clinical or fluid biomarker outcomes. Central memory CD4+ and Th1/17 cells decreased, while CD4+ naive cells increased in the TUDCA arm compared to placebo. Changes in the composition and function of gut microbiota were also noted in the TUDCA arm compared to placebo. Conclusion: Bile acid metabolism in MS is linked with brain and retinal atrophy. TUDCA supplementation in PMS is safe, tolerable and has measurable biological effects that warrant further evaluation in larger trials with a longer treatment duration.


Serum MIF levels at baseline by sex and by PMS subtype. Boxplots overlapped by swarm plots of standardized log-transformed baseline serum MIF levels in PPMS (a) and SPMS (b) cohorts. Bounds of the box represent interquartile range (IQR), while horizontal central lines denote the median and minimum and maximum whiskers correspond to the Q1 − 1.5 × IQR (or the minimum value, if larger) and Q3 + 1.5 × IQR (or the maximum value, if smaller), respectively. ***p < 0.001.
Association of baseline MIF levels with brain atrophy. Relationship between the annualized brain atrophy rate (y-axis) and scaled baseline MIF levels (x-axis) by PMS subtype. Solid lines are derived from linear mixed-effects model including a three-way interaction of time, PMS subtype, and baseline MIF levels and a three-way interaction between time, PMS subtype, and treatment group with BPF as the outcome variable. Shaded areas denote the 95% CI.
Demographics and clinical characteristics at baseline by treatment group.
Baseline MIF as a predictor of brain atrophy.
MIF rate of change.
Serum macrophage migration inhibitory factor levels predict brain atrophy in people with primary progressive multiple sclerosis

November 2023

·

14 Reads

·

1 Citation

Background Macrophage migration inhibitory factor (MIF) is a cytokine linked to multiple sclerosis (MS) progression that is thought to be inhibited by ibudilast. SPRINT-MS was a phase 2 placebo-controlled trial of ibudilast in progressive multiple sclerosis (PMS). Objective To determine whether baseline MIF levels predict imaging outcomes and assess the effects of ibudilast on serum and cerebrospinal fluid (CSF) MIF levels in people with PMS treated with ibudilast. Methods Participants in the SPRINT-MS trial were treated with either ibudilast or placebo and underwent brain magnetic resonance imaging (MRI) every 24 weeks over a duration of 96 weeks. MIF was measured in serum and CSF. Results MIF levels were compared with imaging outcomes in 223 participants from the SPRINT-MS study. In the primary progressive multiple sclerosis (PPMS) cohort, males had higher serum (p < 0.001) and CSF (p = 0.01) MIF levels, as compared with females. Higher baseline serum MIF levels in PPMS were associated with faster brain atrophy (beta = −0.113%, 95% confidence interval (CI): −0.204% to −0.021%; p = 0.016). These findings were not observed in secondary progressive multiple sclerosis (SPMS). Ibudilast did not affect either serum or CSF MIF levels. Conclusions Serum MIF levels were associated with male sex and predicted brain atrophy in PPMS, but not SPMS. Ibudilast did not demonstrate an effect on MIF levels, as compared with placebo, although we cannot exclude a functional effect.


Fig. 1. Representation of Included Countries Stratified by Income Level. World Health Organization. A majority of the evidence focused on the impact of cerebrovascular disease (17/39; 43.6%) and dementia (5/39; 12.8%) on COVID-19 severity and mortality. 92.3% of the articles (36/39) included for analysis suggested a significant association between neurological conditions and increased risk of severe COVID-19 and mortality.
Selected articles assessing relationship between Parkinson's disease and COVID-19 infection severity.
Selected Articles Assessing Relationship Between Multiple Sclerosis and COVID-19 Infection Severity.
Pre-existing neurological conditions and COVID-19 co-infection: Data from systematic reviews, meta-analyses, and scoping reviews

October 2023

·

52 Reads

·

8 Citations

Journal of the Neurological Sciences

Background Pre-existing neurological diseases have been identified as risk factors for severe COVID-19 infection and death. There is a lack of comprehensive literature review assessing the relationship between pre-existing neurological conditions and COVID-19 outcomes. Identification of high risk groups is critical for optimal treatment and care. Methods A literature review was conducted for systematic reviews, meta-analyses, and scoping reviews published between January 1, 2020 and January 1, 2023. Literature assessing individuals with pre-existing neurological diseases and COVID-19 infection was included. Information regarding infection severity was extracted, and potential limitations were identified. Results Thirty-nine articles met inclusion criteria, with data assessing >3 million patients from 51 countries. 26/51 (50.9%) of countries analyzed were classified as high income, while the remaining represented middle-low income countries (25/51; 49.0%). A majority of evidence focused on the impact of cerebrovascular disease (17/39; 43.5%) and dementia (5/39; 12.8%) on COVID-19 severity and mortality. 92.3% of the articles (36/39) suggested a significant association between neurological conditions and increased risk of severe COVID-19 and mortality. Cerebrovascular disease, dementia, Parkinson's disease, and epilepsy were associated with increased COVID severity and mortality. Conclusion Pre-existing neurological diseases including cerebrovascular disease, Alzheimer's disease and other dementias, epilepsy, and Parkinson's disease are significant risk factors for severity of COVID-19 infection and mortality in the acute infectious period. Given that 61.5% (24/39) of the current evidence only includes data from 2020, further updated literature is crucial to identify the relationship between chronic neurological conditions and clinical characteristics of COVID-19 variants.


The calvaria stands alone: Unique aspects of the skull bone marrow-meninges border

August 2023

·

22 Reads

Cell

Channels connecting the skull bone marrow and the meninges have recently been discovered as a path for immune cell and molecule trafficking. In this issue of Cell, Kolabas, Kuemmerle, Perneczky, Förstera, and colleagues characterize these channels in humans and mice, revealing unique features of skull bone marrow and localized activation in human pathology.


Citations (11)


... Therapies aimed at modifying gut microbiota, such as fecal microbiota transplantation and bile acid derivatives (e.g., tauroursodeoxycholic acid), indirectly influence microglial activation via systemic immune modulation. This strategy highlights the role of systemic inflammation in CNS pathophysiology [50]. ...

Reference:

Microglia/Macrophages in Autoimmune Demyelinating Encephalomyelitis (Multiple Sclerosis/Neuromyelitis Optica)
Bile acid metabolites predict multiple sclerosis progression and supplementation is safe in progressive disease
  • Citing Article
  • October 2024

Med

... Alzheimer's disease MTG Visium (10× Genomics) [132] TH, HY, FB, HIP, CNU, CTXsp, OLF, ENTI, TEP, AUD, SSp, PTL and RSP Spatial transcriptomics (ST) and In situ sequencing (ISS) [23] HIP and OLF Spatial transcriptomics (ST) [153] HIP and NCx Visium (10× Genomics) [134] TH, HY, FB, HIP, CNU, CTXsp, OLF, ENTI and NCx Visium (10× Genomics) [133] Parkinson's disease CC; ACA, MOs, and MOp GeoMx (NanoString) [137] Multiple sclerosis TH, HY, FB, HIP, CNU, OLF and NCx Visium (10× Genomics) [141] CR Spatial transcriptomics (ST) and Laser capture microdissection (LCM) [139,140] CCa and SC In situ sequencing (ISS) and Xenium [142] Amyotrophic lateral sclerosis CB Spatial transcriptomics (ST) [146] SC Spatial transcriptomics (ST) [147] Major depressive disorder TH, HY, FB, HIP, CNU, AMY, CTXsp, OLF, ENTI, TEP, AUD, SSp, PTL and RSP Visium (10× Genomics) [149] dlPFC Visium (10× Genomics) [24] Schizophrenia dlPFC Visium (10× Genomics) [150] Fear AMY Visium (10× Genomics) [151] Sleep deprivation HY, HIP, FB, TH, NCx and CNU Visium (10× Genomics) [152] Note to neuronal loss and functional decline. ST has significantly advanced our understanding of the molecular and cellular mechanisms underlying these pathologies, offering insights into the spatial dynamics of disease progression and neuronal vulnerability and immune responses ( Figure 5). ...

Spatial Transcriptomics of Meningeal Inflammation Reveals Inflammatory Gene Signatures in Adjacent Brain Parenchyma
  • Citing Preprint
  • September 2024

... Classical neurodegenerative diseases include Parkinson's disease (PD) and Alzheimer's disease (AD) (Breijyeh and Karaman, 2020;Marogianni et al., 2020). Furthermore, multiple sclerosis (MS), which is mainly neuroinflammatory disease, can be regarded as a neurodegenerative illness (Garton et al., 2024). Although having distinct pathogenetic processes, all of these disorders share the characteristic of persistent neuroinflammation. ...

Neurodegeneration and demyelination in multiple sclerosis
  • Citing Article
  • June 2024

Neuron

... TUDCA was found to inhibit inflammatory responses in C57BL/6 mice by suppressing astrocyte hyperactivation in the cerebral cortex of EAE mice through the G-protein coupled bile acid receptor Gpbar1/ threonine kinase/ I (TGR5/AKT/NF-KB) signaling pathway (Xu et al., 2023). Another study also found that TUDCA supplementation increased circulating levels of several bile acids, affected peripheral immune phenotypes, and altered gut microbiota (Ladakis et al., 2024). In a mouse model of EAE, agonism of GPBAR1 was found to reduce the number of monocytes and T cells in the CNS and to reduce monocyte and microglia activation in the CNS as well as monocyte activation in the periphery, and a reduction in the severity of the disease in EAE was found to be detected by clinical scores (Lewis et al., 2014). ...

Bile acid metabolites predict multiple sclerosis progression and supplementation is safe in progressive disease

... Elderly individuals have been identified as a population with a high mortality rate of COVID-19, severe COVID-19 symptoms and complications, and the need for invasive mechanical ventilation [3]. In particular, elderly individuals with the diagnosis of a major neurocognitive disorder (MND) have increased COVID-19 severity and mortality rates [4][5][6][7][8][9][10][11]. A study found that large increases in mortality in individuals with MNDs such as Alzheimer's disease as an underlying or contributing cause of death occurred in the first year of the COVID-19 pandemic in the United States [12]. ...

Pre-existing neurological conditions and COVID-19 co-infection: Data from systematic reviews, meta-analyses, and scoping reviews

Journal of the Neurological Sciences

... In the future, a combined approach using radiological and laboratory biomarkers may be crucial in the differential diagnosis of MS and SLE. In the context of MS, there are already highly specific imaging biomarkers, such as the central vein sign (CVS) and paramagnetic rim lesions (PRLs), which show a high specificity for this disease [85]. CVS is an imaging marker detected on MRI that appears as small, round, or oval demyelinating lesions with a clearly visible central vein [85]. ...

Emerging imaging and liquid biomarkers in multiple sclerosis

European Journal of Immunology

... Acute necrotizing encephalopathy (ANE) is one of the most severe forms of acute encephalopathy, accounting for approximately 4% of all AE cases. To date, only a few cases of pediatric ANE with COVID-19 have been reported; however, the majority of cases have resulted in devastating neurological sequelae [2][3][4]. Here, we report a case of severe ANE associated with COVID-19 with favorable outcomes after early immunomodulatory therapy and therapeutic plasma exchange (TPE). ...

Acute Necrotizing Encephalitis as an Early Manifestation of COVID-19

Cureus

... With this study we investigated cerebrospinal uid (CSF) cells of monocyte origin expressing dual immunoglobulin domain-containing cell adhesion molecule (DICAM; with the alternative names MXRA8 and limitrin). DICAM is a membrane-bound receptor with proposed BBB-migratory abilities [6], likely through its homophilic interaction with DICAM and heterophilic interaction with aVb3 on endothelial cells [7]. Also, DICAM is likely implicated in intracellular signalling pathways affecting various functions of cells including proliferation [8], angiogenesis [9], apoptosis [9] and expression of in ammatory molecules, including IL-1b, TNFa, IL-6 and integrin b1 and b3 [10]. ...

DICAM, a molecular passport for T H 17 cell entry into the brain
  • Citing Article
  • January 2022

Science Translational Medicine

... However, since the beginning of COVID-19 vaccination campaign, it has been evident that MS patients receiving disease-modifying therapies (DMTs) might be unable to mount a complete immune response against breakthrough COVID-19 infections. This concern was particularly relevant for some DMTs, especially aCD20 agents, which can significantly impact antibody production and reduce the amount of antigen-specific B cells [21,[23][24][25][26][27]. ...

Discordant humoral and T cell immune responses to SARS-CoV-2 vaccination in people with multiple sclerosis on anti-CD20 therapy

EBioMedicine

... Several questions have been posed, regarding the effect on humoral and cellular responses to vaccination based on the effect of rituximab (RTX) and ocrelizumab (OCR) on CD20 pos B-cells and T cells. A limited humoral response upon vaccination has already been demonstrated in several studies focusing on neurological patients (6)(7)(8)(9)(10). On the other hand, several studies have also shown evidence for the preserved cellular response of MS patients treated with RTX or OCR to vaccination against SARS-CoV-2, but there is a paucity of information about the longevity of these responses (11)(12)(13)(14). ...

Discordant humoral and T cell immune responses to SARS-CoV-2 vaccination in people with multiple sclerosis on anti-CD20 therapy