Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 182
Filter
1.
Front Immunol ; 15: 1358886, 2024.
Article in English | MEDLINE | ID: mdl-38660303

ABSTRACT

Background: Bickerstaff brainstem encephalitis (BBE) is a rare disease considered caused by acute demyelination of the brainstem, most often resulting from secondary autoimmune responses. To our knowledge, this is the first probable case report of shingles-associated BBE with anti-sulfatide IgM positivity. Case presentation: We report the case of an 83-year-old woman with symptoms of progressive limb weakness, difficulty swallowing food, and disturbed consciousness that occurred 4 weeks following herpes zoster infection. Autoimmune anti-sulfatide antibodies were positive and fluid-attenuated inversion recovery (FLAIR) sequences revealed clear high signal intensity in pons and bilateral thalamus. Our patient's condition improved markedly with glucocorticoid treatment. After 2 months of treatment, our patient was fully recovered. We considered that for her case, BBE is the most appropriate diagnosis. Conclusions: We emphasize the importance of a careful medical history and assessment of clinical symptoms, performing MRI, testing autoimmune antibodies for rapid diagnosis, and ruling out differential diagnoses. Further studies involving more patients with BBE with IgM anti-sulfatide autoantibodies will increase the understanding of the clinical characteristics and advance the diagnosis and treatment of this syndrome. Meanwhile, it is crucial for dermatologists to know about this severe neurological complication following shingles.


Subject(s)
Autoantibodies , Brain Stem , Encephalitis , Immunoglobulin M , Sulfoglycosphingolipids , Humans , Female , Brain Stem/immunology , Aged, 80 and over , Immunoglobulin M/immunology , Immunoglobulin M/blood , Autoantibodies/immunology , Autoantibodies/blood , Encephalitis/diagnosis , Encephalitis/immunology , Encephalitis/drug therapy , Sulfoglycosphingolipids/immunology , Magnetic Resonance Imaging , Glucocorticoids/therapeutic use
2.
Nature ; 619(7969): 323-331, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37380766

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) that results in significant neurodegeneration in the majority of those affected and is a common cause of chronic neurological disability in young adults1,2. Here, to provide insight into the potential mechanisms involved in progression, we conducted a genome-wide association study of the age-related MS severity score in 12,584 cases and replicated our findings in a further 9,805 cases. We identified a significant association with rs10191329 in the DYSF-ZNF638 locus, the risk allele of which is associated with a shortening in the median time to requiring a walking aid of a median of 3.7 years in homozygous carriers and with increased brainstem and cortical pathology in brain tissue. We also identified suggestive association with rs149097173 in the DNM3-PIGC locus and significant heritability enrichment in CNS tissues. Mendelian randomization analyses suggested a potential protective role for higher educational attainment. In contrast to immune-driven susceptibility3, these findings suggest a key role for CNS resilience and potentially neurocognitive reserve in determining outcome in MS.


Subject(s)
Brain , Cognitive Reserve , Educational Status , Genome-Wide Association Study , Multiple Sclerosis , Protective Factors , Humans , Young Adult , Aging , Brain/immunology , Brain/pathology , Brain/physiopathology , Brain Stem/immunology , Brain Stem/pathology , Brain Stem/physiopathology , Case-Control Studies , Disease Progression , Homozygote , Mobility Limitation , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Multiple Sclerosis/psychology , Time Factors
4.
Respir Physiol Neurobiol ; 296: 103800, 2022 02.
Article in English | MEDLINE | ID: mdl-34626831

ABSTRACT

Infants born with neonatal opioid withdrawal syndrome (NOWS) can display abnormal cardiorespiratory patterns including tachypnea, tachycardia, and impaired ventilatory responses to hypoxia (HVR) and hypercapnia (HCVR). Chronic morphine exposure is associated with increased midbrain microglial expression. Using a rat model of pre- and post-natal morphine exposure, we assessed cardiorespiratory features of NOWS (resting tachycardia and tachypnea) including the attenuated HVR and HCVR and whether they are associated with increased brainstem microglia expression. Pregnant rats (dams) received twice-daily subcutaneous injections of morphine (5 mg/kg) during the third (last) week of pregnancy to simulate 3rd trimester in utero opioid exposure. Offspring then received once-daily subcutaneous injections of morphine (0.5 mg/kg) until postnatal (P) day P10 days of age to simulate postnatal morphine therapy. Cardiorespiratory responses were assessed 24 h later (P11 days) following spontaneous withdrawal. Compared to saline-treated pups, morphine-exposed offspring exhibited tachycardia and tachypnea as well as an attenuated HVR and HCVR. Microglial cell counts were increased in the nucleus tractus solitarius (nTS), dorsal motor nucleus of the vagus (DMNV) and nucleus ambiguous (NAamb), but not the retrapezoid nucleus (RTN) or the non-cardiorespriatory region, the cuneate nucleus (CN). These data suggest that the cardiorespiratory features and autonomic dysregulation in NOWS infants may be associated with altered microglial function in specific brainstem cardiorespiratory control regions.


Subject(s)
Brain Stem , Infant, Newborn, Diseases , Microglia , Opioid-Related Disorders , Substance Withdrawal Syndrome , Tachycardia , Tachypnea , Animals , Animals, Newborn , Brain Stem/immunology , Brain Stem/physiopathology , Disease Models, Animal , Female , Humans , Hypercapnia/immunology , Hypercapnia/physiopathology , Hypoxia/immunology , Hypoxia/physiopathology , Infant, Newborn , Infant, Newborn, Diseases/etiology , Infant, Newborn, Diseases/immunology , Infant, Newborn, Diseases/physiopathology , Microglia/immunology , Opioid-Related Disorders/complications , Opioid-Related Disorders/immunology , Opioid-Related Disorders/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/physiopathology , Rats , Substance Withdrawal Syndrome/complications , Substance Withdrawal Syndrome/immunology , Substance Withdrawal Syndrome/physiopathology , Tachycardia/etiology , Tachycardia/immunology , Tachycardia/physiopathology , Tachypnea/etiology , Tachypnea/immunology , Tachypnea/physiopathology
5.
Front Immunol ; 12: 772763, 2021.
Article in English | MEDLINE | ID: mdl-34858431

ABSTRACT

Anti-contactin-associated protein-like 2 (CASPR2) antibody-associated autoimmune encephalitis is commonly characterized by limbic encephalitis with clinical symptoms of mental and behavior disorders, cognitive impairment, deterioration of memory, and epilepsy. The classical lesions reported are located at the medial temporal lobe or hippocampus, whereas prominent brainstem lesions have not been addressed to date. Herein, we reported two patients mimicking progressive brainstem infarction with severe neurological manifestations. On brain magnetic resonance imaging (MRI), prominent brainstem lesions were noted, although multifocal lesions were also shown in the juxtacortical and subcortical white matters, basal ganglia, hippocampus, and cerebellar hemisphere. Unexpectedly and interestingly, both cases had detectable CASPR2 antibodies in sera, and an exclusive IgG1 subclass was documented in the further analysis. They were treated effectively with aggressive immunosuppressive therapies including corticosteroids, intravenous immunoglobulin G, and rituximab, with the first case achieving a rapid remission and the other undergoing a slow but gradual improvement. To the best of our knowledge, this is the first report on prominent brainstem involvement with definite MRI lesions in anti-CASPR2 antibody-associated autoimmune encephalitis, which helps to expand the clinical spectrum of this rare autoimmune disease and update the lesion patterns in the CNS.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , Brain Stem/immunology , Encephalitis/immunology , Autoimmune Diseases/diagnosis , Brain Stem/diagnostic imaging , Encephalitis/diagnosis , Female , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Reproducibility of Results , Sensitivity and Specificity
6.
Nat Commun ; 12(1): 5809, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34608167

ABSTRACT

SARS-CoV-2 has caused a global pandemic of COVID-19 since its emergence in December 2019. The infection causes a severe acute respiratory syndrome and may also spread to central nervous system leading to neurological sequelae. We have developed and characterized two new organotypic cultures from hamster brainstem and lung tissues that offer a unique opportunity to study the early steps of viral infection and screening antivirals. These models are not dedicated to investigate how the virus reaches the brain. However, they allow validating the early tropism of the virus in the lungs and demonstrating that SARS-CoV-2 could infect the brainstem and the cerebellum, mainly by targeting granular neurons. Viral infection induces specific interferon and innate immune responses with patterns specific to each organ, along with cell death by apoptosis, necroptosis, and pyroptosis. Overall, our data illustrate the potential of rapid modeling of complex tissue-level interactions during infection by a newly emerged virus.


Subject(s)
Brain Stem/virology , Lung/virology , Models, Biological , SARS-CoV-2/pathogenicity , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Alveolar Epithelial Cells/virology , Animals , Antiviral Agents/pharmacology , Brain Stem/cytology , Brain Stem/immunology , Brain Stem/pathology , Cricetinae , Immunity, Innate , Inflammation , Lung/cytology , Lung/immunology , Lung/pathology , Neurons/virology , Organ Culture Techniques , Regulated Cell Death , SARS-CoV-2/drug effects , Viral Tropism
7.
Front Immunol ; 12: 700903, 2021.
Article in English | MEDLINE | ID: mdl-34566960

ABSTRACT

The activation of the sympathetic nervous system, release of norepinephrine (NE), and adrenergic receptor signaling participate in and regulate the complicated enterovirus 71 (EV71) brainstem encephalitis (BE). The neurotoxin 6-hydroxydopamine (6-OHDA) selectively ablates sympathetic nerves and markedly depletes NE in innervated organs. Changes in the plasma levels of NE, severity score, cytokine profiles, and percentages of immunophenotype expression in 7-day-old Bltw : CD1 (ICR) mice infected with EV71, with or without 6-OHDA treatment, were compared. The survival rate (76.9%) of EV71-infected and 6-OHDA (30 µg/g)-treated mice was increased significantly. The clinical scores were decreased markedly on days 8-12 in MP4-infected and 6-OHDA-treated mice compared to those without treatment. The results showed that the plasma levels of NE, epinephrine, and dopamine were decreased on days 4-8 after 6-OHDA treatment and at most on day 8. The plasma levels of interleukin (IL)-12p70, tumor necrosis factor, IL-6, and IL-10 did not change significantly after 6-OHDA treatment. Interferon-γ levels decreased evidently on days 4, 6, and 8 after 6-OHDA treatment. The absolute events of CD3+CD4+, CD3+CD8+, and CD3+NK1.1+ cells of peripheral blood mononuclear cells were increased significantly in MP4-infected and 6-OHDA-treated mice compared to those without treatment. In splenocytes, the absolute cells of CD3-NK1.1+, CD3+NK1.1+ and CD11b+Gr-1+ cells of EV71-infected mice were increased significantly after 6-OHDA treatment. These findings suggested that 6-OHDA may be used a probe to explore clinical improvements and immune responses in the complicated EV71 infection. Taken together, peripheral chemical sympathectomy contribute to further understand the immunopathogenesis of EV71 BE with autonomic nervous system dysregulation.


Subject(s)
Encephalitis, Viral/immunology , Enterovirus Infections/immunology , Sympathectomy, Chemical/methods , Animals , Brain Stem/immunology , Brain Stem/pathology , Encephalitis, Viral/pathology , Enterovirus A, Human , Enterovirus Infections/pathology , Mice , Mice, Inbred ICR , Oxidopamine
8.
Anticancer Res ; 41(5): 2583-2589, 2021 May.
Article in English | MEDLINE | ID: mdl-33952487

ABSTRACT

BACKGROUND/AIM: High-grade gliomas have a poor prognosis despite standard treatment. The aim of the study was to identify new prognostic factors to select patients who need more intense treatment. PATIENTS AND METHODS: Forty-three consecutive patients underwent surgery plus chemoradiotherapy for pathologically diagnosed high-grade gliomas (grade III, IV). RESULTS: The median survival time was 989 days, and the 1-year survival rate was 87.6%. Among patients with grade IV disease, the median survival time, 1-year, and 2-year survival rate were 814 days, 82.6%, and 58.7%, respectively. In the univariate analysis, unmethylated MGMT promoter (p=0.0495), brainstem infiltration (p=0.0004), basal ganglia as the primary lesion site (p=0.0056), 3-dimensional conformal radiotherapy (p=0.0286), and <50 Gy (p=0.0049) were associated with a poor prognosis. In the multivariate analysis, only brainstem infiltration retained significance (HR for death, 0.21; 95% CI=0.06-0.70; p=0.011). CONCLUSION: Brainstem infiltration is a novel prognostic factor for poor prognosis in patients with high-grade gliomas.


Subject(s)
Brain Stem/immunology , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Glioma/drug therapy , Glioma/radiotherapy , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Basal Ganglia/immunology , Basal Ganglia/pathology , Brain Stem/pathology , Chemoradiotherapy/adverse effects , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/immunology , Glioma/immunology , Glioma/pathology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Neoplasm Grading , Promoter Regions, Genetic/genetics
9.
Neuropathol Appl Neurobiol ; 47(2): 316-327, 2021 02.
Article in English | MEDLINE | ID: mdl-32910464

ABSTRACT

AIMS: Progressive encephalomyelitis with rigidity and myoclonus (PERM) is a life-threatening condition often associated with highly raised serum antibodies to glycine receptors (GlyRs); these bind to the surface of large neurons and interneurons in rodent brain and spinal cord sections and, in vitro, inhibit function and reduce surface expression of the GlyRs. The effects in vivo have not been reported. METHODS: Purified plasma IgG from a GlyR antibody-positive patient with PERM, and a healthy control (HC), was injected daily into the peritoneal cavity of mice for 12 days; lipopolysaccharide (LPS) to open the blood-brain barrier, was injected on days 3 and 8. Based on preliminary data, behavioural tests were only performed 48 h post-LPS on days 5-7 and 10-12. RESULTS: The GlyR IgG injected mice showed impaired ability on the rotarod from days 5 to 10 but this normalized by day 12. There were no other behavioural differences but, at termination (d13), the GlyR IgG-injected mice had IgG deposits on the neurons that express GlyRs in the brainstem and spinal cord. The IgG was not only on the surface but also inside these large GlyR expressing neurons, which continued to express surface GlyR. CONCLUSIONS: Despite the partial clinical phenotype, not uncommon in passive transfer studies, the results suggest that the antibodies had accessed the GlyRs in relevant brain regions, led to antibody-mediated internalization and increased GlyR synthesis, compatible with the temporary loss of function.


Subject(s)
Autoantibodies/pharmacology , Encephalomyelitis/immunology , Immunoglobulin G/pharmacology , Motor Neurons/metabolism , Muscle Rigidity/immunology , Receptors, Glycine/metabolism , Animals , Autoantibodies/immunology , Autoantigens/immunology , Autoantigens/metabolism , Brain Stem/immunology , Brain Stem/metabolism , Encephalomyelitis/metabolism , Humans , Immunoglobulin G/immunology , Male , Mice , Mice, Inbred C57BL , Motor Neurons/immunology , Muscle Rigidity/metabolism , Myoclonus/immunology , Myoclonus/metabolism , Receptors, Glycine/immunology , Spinal Cord/immunology , Spinal Cord/metabolism
10.
Acupunct Med ; 39(4): 376-384, 2021 08.
Article in English | MEDLINE | ID: mdl-32744055

ABSTRACT

BACKGROUND: Lipopolysaccharide (LPS) endotoxins are activators of innate immunity inducing infection and inflammatory responses. Anti-inflammatory drugs can have undesirable side effects. Acupuncture may be an alternative for the treatment of inflammatory processes. OBJECTIVE: We investigated the potential anti-inflammatory effect of manual acupuncture (MA) at SP6 upon LPS-induced peritonitis in rats. METHODS: Peritonitis was induced in rats with an intraperitoneal injection of LPS (0.002, 0.02, 0.2 or 2 µg/kg) in four experimental groups (n = 6 each). A fifth group was injected with sterile saline solution (saline group, n = 6). Four hours after the procedure, peritoneal fluid was collected to determine total cell counts for inflammatory cells, differential leukocyte counts and peritoneal capillary permeability. The LPS dose of 0.02 µg/kg was used in the subsequent experiments as it most successfully induced peritoneal inflammation. Subsequently, five experimental groups (n = 12 rats each) were used: (1) saline, (2) control (untreated LPS group), (3) indomethacin (LPS group treated with indomethacin), (4) NA (LPS group treated with MA at a location not corresponding to any traditional acupuncture point), and (5) SP6 (LPS group treated with verum MA at SP6). Ten minutes after MA or 30 min after indomethacin treatment, the rats received an intraperitoneal injection of LPS. After 4 h, total leukocyte and differential cell counts, myeloperoxidase (MPO) activity, vascular permeability and cytokine levels were evaluated in the peritoneal fluid. Cytokine levels were additionally evaluated in the brainstem. RESULTS: SP6 MA and indomethacin treatments reduced inflammatory cell infiltration, vascular permeability and MPO activity in the LPS-exposed rats. Pre-treatment with indomethacin and SP6 MA decreased tumor necrosis factor (TNF)-α levels and preserved interleukin (IL)-10 in the peritoneal fluid. Indomethacin also reduced IL-6 in the peritoneal fluid. In the brainstem, indomethacin reduced IL-1ß, IL-6, TNFα and IL-10, whereas SP6 MA reduced only TNFα and IL-6 levels. CONCLUSIONS: This study clearly demonstrates the anti-inflammatory effect of acupuncture, which we believe may involve the activation of anti-inflammatory neural reflexes in the regulation of peritonitis.


Subject(s)
Acupuncture Therapy , Brain Stem/immunology , Cytokines/immunology , Peripheral Nerves/immunology , Peritonitis/therapy , Acupuncture Points , Animals , Cytokines/genetics , Humans , Lipopolysaccharides/adverse effects , Male , Peritonitis/genetics , Peritonitis/immunology , Rats , Rats, Wistar
11.
Acta Neurobiol Exp (Wars) ; 80(4): 353-357, 2020.
Article in English | MEDLINE | ID: mdl-33350987

ABSTRACT

We recently identified a novel neuroimmune mechanism in the nasal mucosa, in which activation of neuronal Toll­like receptor (TLR) 7 results in upregulation of epithelial TLRs, via release of substance P. In the present study, we assessed whether intranasal challenge with the TLR7 agonist R­837 additionally activated neurons in the central nervous system. Within one hour, R­837 induced activation of the nucleus of the solitary tract, as well as a small increase in nasal IL­6, but otherwise in the absence of an overt inflammatory response. It is tempting to speculate that it might be a direct interaction of R­837 with trigeminal neurons in order to alert the central nervous system of invading pathogens.


Subject(s)
Brain Stem/physiology , Central Nervous System/immunology , Nasal Mucosa/physiology , Toll-Like Receptor 7/metabolism , Animals , Brain Stem/immunology , Central Nervous System/physiology , Male , Mice, Inbred C57BL , Nasal Mucosa/immunology , Neurons/immunology , Proto-Oncogene Proteins c-fos/metabolism
12.
BMJ Case Rep ; 13(9)2020 Sep 18.
Article in English | MEDLINE | ID: mdl-32948528

ABSTRACT

Bickerstaff's brainstem encephalitis (BBE) is a Guillain-Barré syndrome (GBS) spectrum disorder associated with predominantly central nervous system predilection. Patients exhibit a variable constellation of depressed consciousness, bilateral external ophthalmoplegia, ataxia and long tract signs. Although the pathophysiology is not fully understood, it has been associated with anti-GQ1b antibodies in two-thirds of patients. We present a patient with clinical features consistent with BBE and positive anti-GM1 and anti-GD1a antibodies. A diagnostic approach to the acutely unwell patient with brainstem encephalitis is explored in this clinical context with a literature review of the aforementioned ganglioside antibody significance. Intravenous immunoglobulin therapy is highlighted in BBE using up-to-date evidence-based extrapolation from GBS.


Subject(s)
Ataxia/immunology , Autoantibodies/blood , Brain Stem/immunology , Encephalitis/diagnosis , Ophthalmoplegia/immunology , Adult , Ataxia/blood , Autoantibodies/immunology , Diagnosis, Differential , Electroencephalography , Encephalitis/blood , Encephalitis/complications , Encephalitis/immunology , G(M1) Ganglioside/immunology , Gangliosides/immunology , Glasgow Coma Scale , Humans , Male , Ophthalmoplegia/blood
13.
J Pharmacol Exp Ther ; 375(1): 210-222, 2020 10.
Article in English | MEDLINE | ID: mdl-32661056

ABSTRACT

The neural control system underlying breathing is sexually dimorphic with males being more vulnerable to dysfunction. Microglia also display sex differences, and their role in the architecture of brainstem respiratory rhythm circuitry and modulation of cervical spinal cord respiratory plasticity is becoming better appreciated. To further understand the molecular underpinnings of these sex differences, we performed RNA sequencing of immunomagnetically isolated microglia from brainstem and cervical spinal cord of adult male and female rats. We used various bioinformatics tools (Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, Reactome, STRING, MAGICTRICKS) to functionally categorize identified gene sets, as well as to pinpoint common transcriptional gene drivers that may be responsible for the observed transcriptomic differences. We found few sex differences in the microglial transcriptomes derived from the brainstem, but several hundred genes were differentially expressed by sex in cervical spinal microglia. Comparing brainstem and spinal microglia within and between sexes, we found that the major factor guiding transcriptomic differences was central nervous system (CNS) location rather than sex. We further identified key transcriptional drivers that may be responsible for the transcriptomic differences observed between sexes and CNS regions; enhancer of zeste homolog 2 emerged as the predominant driver of the differentially downregulated genes. We suggest that functional gene alterations identified in metabolism, transcription, and intercellular communication underlie critical microglial heterogeneity and sex differences in CNS regions that contribute to respiratory disorders categorized by dysfunction in neural control. These data will also serve as an important resource data base to advance our understanding of innate immune cell contributions to sex differences and the field of respiratory neural control. SIGNIFICANCE STATEMENT: The contributions of central nervous system (CNS) innate immune cells to sexually dimorphic differences in the neural circuitry controlling breathing are poorly understood. We identify key transcriptomic differences, and their transcriptional drivers, in microglia derived from the brainstem and the C3-C6 cervical spinal cord of healthy adult male and female rats. Gene alterations identified in metabolism, gene transcription, and intercellular communication likely underlie critical microglial heterogeneity and sex differences in these key CNS regions that contribute to the neural control of breathing.


Subject(s)
Brain Stem/metabolism , Cervical Cord/metabolism , Microglia/metabolism , Respiration/genetics , Sex Characteristics , Transcriptome/genetics , Animals , Brain Stem/immunology , Cervical Cord/immunology , Female , Immunity, Innate/genetics , Male , Microglia/immunology , Rats , Respiration/immunology
14.
Mol Med ; 26(1): 58, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32546125

ABSTRACT

In light of the present therapeutic situation in COVID-19, any measure to improve course and outcome of seriously affected individuals is of utmost importance. We recap here evidence that supports the use of human recombinant erythropoietin (EPO) for ameliorating course and outcome of seriously ill COVID-19 patients. This brief expert review grounds on available subject-relevant literature searched until May 14, 2020, including Medline, Google Scholar, and preprint servers. We delineate in brief sections, each introduced by a summary of respective COVID-19 references, how EPO may target a number of the gravest sequelae of these patients. EPO is expected to: (1) improve respiration at several levels including lung, brainstem, spinal cord and respiratory muscles; (2) counteract overshooting inflammation caused by cytokine storm/ inflammasome; (3) act neuroprotective and neuroregenerative in brain and peripheral nervous system. Based on this accumulating experimental and clinical evidence, we finally provide the research design for a double-blind placebo-controlled randomized clinical trial including severely affected patients, which is planned to start shortly.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/drug therapy , Cytokine Release Syndrome/prevention & control , Erythropoietin/therapeutic use , Neuroprotective Agents/therapeutic use , Pneumonia, Viral/drug therapy , Respiratory System Agents/therapeutic use , Brain Stem/drug effects , Brain Stem/immunology , Brain Stem/virology , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Coronavirus Infections/virology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Cytokine Release Syndrome/virology , Double-Blind Method , Humans , Lung/drug effects , Lung/immunology , Lung/virology , Pandemics , Phrenic Nerve/drug effects , Phrenic Nerve/immunology , Phrenic Nerve/virology , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Proof of Concept Study , Randomized Controlled Trials as Topic , Recombinant Proteins/therapeutic use , Respiratory Muscles/drug effects , Respiratory Muscles/immunology , Respiratory Muscles/virology , SARS-CoV-2 , Severity of Illness Index , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/virology
15.
J Neuroinflammation ; 17(1): 184, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32532298

ABSTRACT

BACKGROUND: In early autoimmune neuroinflammation, interferon (IFN)γ and its upregulation of the immunoproteasome (iP) is pathologic. However, during chronic multiple sclerosis (MS), IFNγ has protective properties. Although dysregulation of the iP has been implicated in neurodegeneration, its function remains to be fully elucidated. Here, we demonstrate that IFNγ signaling in regional astrocytes induces the iP and promotes protection of the CNS during chronic autoimmunity. METHODS: In a multiple sclerosis (MS) brain, we evaluated mRNA expression and labeled postmortem MS brainstem and spinal cord for iP subunits and indicators of oxidative stress. Primary regional human astrocytes were analyzed for iP regulation and function by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), Western blot, OxyBlot, and reactive oxygen species and caspase activity detection assays. Following immunization with myelin oligodendrocyte glycoprotein (MOG)35-55, the role of IFNγ signaling and the iP during chronic experimental autoimmune encephalomyelitis (EAE) were assessed using pharmacologic inhibition of the iP and genetic interruption of IFNγ signaling specifically in astrocytes. Central nervous system (CNS) tissues were analyzed by immunohistochemistry (IHC) and immunofluorescence, and cell-specific colocalization was quantified. RESULTS: In MS tissue, iP expression was enhanced in the spinal cord compared to brainstem lesions, which correlated with a decrease in oxidative stress. In vitro, IFNγ stimulation enhanced iP expression, reduced reactive oxygen species burden, and decreased oxidatively damaged and poly-ubiquitinated protein accumulation preferentially in human spinal cord astrocytes, which was abrogated with the use of the iP inhibitor, ONX 0914. During the chronic phase of an MS animal model, EAE, ONX 0914 treatment exacerbated the disease and led to increased oxidative stress and poly-ubiquitinated protein buildup. Finally, mice with astrocyte-specific loss of the IFNγ receptor exhibited worsened chronic EAE associated with reduced iP expression, enhanced lesion size and oxidative stress, and poly-ubiquitinated protein accumulation in astrocytes. CONCLUSIONS: Taken together, our data reveal a protective role for IFNγ in chronic neuroinflammation and identify a novel function of the iP in astrocytes during CNS autoimmunity.


Subject(s)
Astrocytes/immunology , Autoimmunity/immunology , Interferon-gamma/immunology , Multiple Sclerosis/immunology , Proteasome Endopeptidase Complex/immunology , Signal Transduction/immunology , Aged , Animals , Astrocytes/metabolism , Brain Stem/immunology , Brain Stem/metabolism , Brain Stem/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Oxidative Stress/immunology , Proteasome Endopeptidase Complex/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology
16.
Cardiovasc Pathol ; 47: 107221, 2020.
Article in English | MEDLINE | ID: mdl-32371340

ABSTRACT

Unexpected sudden cardiac death (SCD), sudden infant death syndrome (SIDS) and sudden intrauterine unexplained death (SIUD) are major unsolved, devastating forms of death that occur frequently. Obstructive sleep apnea (OSA) has been associated with increased cardiovascular and cerebrovascular morbidity and mortality, including sudden cardiac death (SCD). This editorial will review the pathology of SCD, including sudden infant death syndrome (SIDS) and sudden intrauterine unexplained death (SIUD); OSA with its cardiovascular consequences; the possible link between SCD and OSA, discussing the potential mechanisms underlying these two frequent, but yet overlooked pathologies. Finally, the possible preventive benefits of treating OSA and identifying patients at common risk for OSA and SCD and SIDS-SIUD to prevent unexpected deaths will be discussed. Post-mortem examination is of great importance in every case of SCD sine materia, with examination of the brainstem and cardiac conduction system on serial sections, when general autopsy fails, but it should be stressed that also the investigations of patients suffering from OSA should focus on the possibility of pathological findings in common with cases of SCD.


Subject(s)
Brain Stem/pathology , Death, Sudden, Cardiac/pathology , Fetal Death/etiology , Heart Conduction System/pathology , Sleep Apnea, Obstructive/pathology , Sudden Infant Death/pathology , Brain Stem/immunology , Death, Sudden, Cardiac/epidemiology , Death, Sudden, Cardiac/prevention & control , Female , Fetal Death/prevention & control , Heart Conduction System/immunology , Humans , Infant , Infant, Newborn , Inflammation Mediators/immunology , Pregnancy , Prognosis , Risk Factors , Sleep Apnea, Obstructive/immunology , Sleep Apnea, Obstructive/mortality , Sleep Apnea, Obstructive/therapy , Sudden Infant Death/epidemiology , Sudden Infant Death/immunology , Sudden Infant Death/prevention & control
17.
J Virol ; 94(1)2019 12 12.
Article in English | MEDLINE | ID: mdl-31597775

ABSTRACT

Demyelinating central nervous system (CNS) disorders like multiple sclerosis (MS) and acute disseminated encephalomyelitis (ADEM) have been difficult to study and treat due to the lack of understanding of their etiology. Numerous cases point to the link between herpes simplex virus (HSV) infection and multifocal CNS demyelination in humans; however, convincing evidence from animal models has been missing. In this work, we found that HSV-1 infection of the cotton rat Sigmodon hispidus via a common route (lip abrasion) can cause multifocal CNS demyelination and inflammation. Remyelination occurred shortly after demyelination in HSV-1-infected cotton rats but could be incomplete, resulting in "scars," further supporting an association between HSV-1 infection and multifocal demyelinating disorders. Virus was detected sequentially in the lip, trigeminal ganglia, and brain of infected animals. Brain pathology developed primarily on the ipsilateral side of the brain stem, in the cerebellum, and contralateral side of the forebrain/midbrain, suggesting that the changes may ascend along the trigeminal lemniscus pathway. Neurologic defects occasionally detected in infected animals (e.g., defective whisker touch and blink responses and compromised balance) could be representative of the brain stem/cerebellum dysfunction. Immunization of cotton rats with a split HSV-1 vaccine protected animals against viral replication and brain pathology, suggesting that vaccination against HSV-1 may protect against demyelinating disorders.IMPORTANCE Our work demonstrates for the first time a direct association between infection with herpes simplex virus 1, a ubiquitous human pathogen generally associated with facial cold sores, and multifocal brain demyelination in an otherwise normal host, the cotton rat Sigmodon hispidus For a long time, demyelinating diseases were considered to be autoimmune in nature and were studied by indirect methods, such as immunizing animals with myelin components or feeding them toxic substances that induce demyelination. Treatment against demyelinating diseases has been elusive, partially because of their unknown etiology. This work provides the first experimental evidence for the role of HSV-1 as the etiologic agent of multifocal brain demyelination in a normal host and suggests that vaccination against HSV-1 can help to combat demyelinating disorders.


Subject(s)
Demyelinating Diseases/prevention & control , Encephalitis/prevention & control , Herpes Simplex Virus Vaccines/administration & dosage , Herpes Simplex/prevention & control , Herpesvirus 1, Human/drug effects , Animals , Brain Stem/drug effects , Brain Stem/immunology , Brain Stem/pathology , Brain Stem/virology , Cerebellum/drug effects , Cerebellum/immunology , Cerebellum/pathology , Cerebellum/virology , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Demyelinating Diseases/virology , Disease Models, Animal , Encephalitis/immunology , Encephalitis/pathology , Encephalitis/virology , Female , Herpes Simplex/immunology , Herpes Simplex/pathology , Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Humans , Male , Prosencephalon/drug effects , Prosencephalon/immunology , Prosencephalon/pathology , Prosencephalon/virology , Sigmodontinae , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/immunology , Trigeminal Ganglion/pathology , Trigeminal Ganglion/virology , Vaccination , Viral Load/drug effects
18.
J Neurophysiol ; 122(4): 1421-1460, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31339807

ABSTRACT

Acoustic overstimulation (AOS) is defined as the stressful overexposure to high-intensity sounds. AOS is a precipitating factor that leads to a glutamate (GLU)-induced Type I auditory neural excitotoxicity and an activation of an immune/inflammatory/oxidative stress response within the inner ear, often resulting in cochlear hearing loss. The dendrites of the Type I auditory neural neurons that innervate the inner hair cells (IHCs), and respond to the IHC release of the excitatory neurotransmitter GLU, are themselves directly innervated by the dynorphin (DYN)-bearing axon terminals of the descending brain stem lateral olivocochlear (LOC) system. DYNs are known to increase GLU availability, potentiate GLU excitotoxicity, and induce superoxide production. DYNs also increase the production of proinflammatory cytokines by modulating immune/inflammatory signal transduction pathways. Evidence is provided supporting the possibility that the GLU-mediated Type I auditory neural dendritic swelling, inflammation, excitotoxicity, and cochlear hearing loss that follow AOS may be part of a brain stem-activated, DYN-mediated cascade of inflammatory events subsequent to a LOC release of DYNs into the cochlea. In support of a DYN-mediated cascade of events are established investigations linking DYNs to the immune/inflammatory/excitotoxic response in other neural systems.


Subject(s)
Dynorphins/immunology , Ear, Inner/immunology , Ear, Inner/physiopathology , Glutamic Acid/immunology , Hearing Loss, Noise-Induced/immunology , Neurons/immunology , Otitis/immunology , Animals , Brain Stem/immunology , Brain Stem/physiopathology , Ear, Inner/innervation , Humans
19.
J Neurovirol ; 25(4): 578-588, 2019 08.
Article in English | MEDLINE | ID: mdl-31119711

ABSTRACT

Despite combination antiretroviral therapies making HIV a chronic rather than terminal condition for many people, the prevalence of HIV-associated neurocognitive disorders (HAND) is increasing. This is especially problematic for children living with HIV. Children diagnosed HAND rarely display the hallmark pathology of HIV encephalitis in adults, namely infected macrophages and multinucleated giant cells in the brain. This finding has also been documented in rhesus macaques infected perinatally with simian immunodeficiency virus (SIV). However, the extent and mechanisms of lack of susceptibility to encephalitis in perinatally HIV-infected children remain unclear. In the current study, we compared brains of macaques infected with pathogenic strains of SIV at different ages to determine neuropathology, correlates of neuroinflammation, and potential underlying mechanisms. Encephalitis was not found in the macaques infected within 24 h of birth despite similar high plasma viral load and high monocyte turnover. Macaques developed encephalitis only when they were infected after 4 months of age. Lower numbers of CCR5-positive cells in the brain, combined with a less leaky blood-brain barrier, may be responsible for the decreased virus infection in the brain and consequently the absence of encephalitis in newborn macaques infected with SIV.


Subject(s)
Blood-Brain Barrier/immunology , Brain Stem/immunology , Disease Resistance , Encephalitis, Viral/immunology , Frontal Lobe/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/pathogenicity , Age Factors , Animals , Animals, Newborn , Blood-Brain Barrier/pathology , Blood-Brain Barrier/virology , Brain Stem/pathology , Brain Stem/virology , Capillary Permeability/immunology , Encephalitis, Viral/genetics , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Frontal Lobe/pathology , Frontal Lobe/virology , Gene Expression , Macaca mulatta/virology , Macrophages/immunology , Macrophages/pathology , Macrophages/virology , Monocytes/immunology , Monocytes/pathology , Monocytes/virology , RNA, Viral/genetics , RNA, Viral/metabolism , Receptors, CCR5/genetics , Receptors, CCR5/immunology , Receptors, Virus/genetics , Receptors, Virus/immunology , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...