Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 282
Filter
1.
Exp Eye Res ; 237: 109708, 2023 12.
Article in English | MEDLINE | ID: mdl-37913917

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE), induced by the immunization of myelin oligodendrocyte glycoprotein (MOG), is related to human MOG antibody-associated disease (MOGAD). Neuroinflammation and demyelination of the optic nerve can lead to retinal ganglion cell (RGC) death and axonal damage in MOGAD. Here, we aimed to evaluate the structural changes in RGCs longitudinally by in vivo imaging in mice with RGCs expressing yellow fluorescent protein along the course of EAE. Successful induction of EAE was confirmed by the neurological function scores and histology analyses. The changes in the thickness of ganglion cell complex (GCC) layer and RGC survival and dendrites were monitored longitudinally along the course of EAE. Before the onset of EAE, there were no significant changes in the number and morphology of RGCs and the thickness of the GCC layer as compared to the mice without EAE induction. After the onset of EAE, the thickness of the GCC layer and the RGC number and dendritic network all gradually decreased along the course of EAE. Notably, dendritic shrinkage could be detected earlier than the thinning of the GCC layer. In summary, this study delineated the longitudinal profile of RGC structural changes in EAE mice, providing an assessment platform for monitoring outcomes of RGC treatments.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Retinal Ganglion Cells , Humans , Mice , Animals , Retinal Ganglion Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Retina/pathology , Optic Nerve/pathology , Dendrites , Mice, Inbred C57BL
2.
Am J Ther ; 30(3): e220-e231, 2023 May 01.
Article in English | MEDLINE | ID: mdl-37278703

ABSTRACT

BACKGROUND: As far as 80% of people diagnosed with multiple sclerosis (MS) experience disabling symptoms in the course of the disease, such as spasticity and neuropathic pain. As first-line symptomatic therapy is associated with important adverse reactions, cannabinoids have become increasingly popular among patients with MS. This review intends to provide an overview of the evidence of the role of cannabinoids in treating symptoms related to MS and to encourage further research on this matter. AREAS OF UNCERTAINTY: To date, the evidence supporting the role of cannabis and its derivatives in alleviating the MS-related symptoms comes only from studies on experimental models of demyelination. To the best of our knowledge, relatively few clinical trials inquired about the therapeutic effects of cannabinoids on patients with MS, with variable results. DATA SOURCES: We conducted a literature search through PubMed and Google Scholar from the beginning until 2022. We included articles in English describing the latest findings regarding the endocannabinoid system, the pharmacology of cannabinoids, and their therapeutic purpose in MS. RESULTS: Evidence from preclinical studies showed that cannabinoids can limit the demyelination process, promote remyelination, and have anti-inflammatory properties by reducing immune cell infiltration of the central nervous system in mice with experimental autoimmune encephalomyelitis. Moreover, it has been established that experimental autoimmune encephalomyelitis mice treated with cannabinoids experienced a significant reduction of symptoms and slowing of the disease progression. Given the complexity of human immune and nervous systems, cannabinoids did not have the anticipated effects on human subjects. However, data obtained from clinical trials showed some beneficial results of cannabinoids as a single or as add-on therapy in reducing the spasticity and pain related to MS. CONCLUSION: Considering their various mechanisms of action and good tolerability, cannabinoids remain an interesting therapy for spasticity and chronic pain related to MS.


Subject(s)
Cannabinoids , Cannabis , Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Neuralgia , Humans , Animals , Mice , Cannabinoids/adverse effects , Multiple Sclerosis/drug therapy , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/complications , Cannabinoid Receptor Agonists , Neuralgia/drug therapy , Neuralgia/etiology , Muscle Spasticity/drug therapy , Muscle Spasticity/etiology , Models, Theoretical
3.
Exp Neurol ; 357: 114201, 2022 11.
Article in English | MEDLINE | ID: mdl-35963325

ABSTRACT

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system, presenting with optic neuritis in about 20-30% of cases. Optic nerve demyelination, associated with delay of visual evoked potentials (VEPs), is also observed prior to motor signs in the preclinical MS model Experimental Autoimmune Encephalomyelitis (EAE). Transcranial direct current stimulation (tDCS), inducing polarity-dependent changes in neuronal excitability, is widely used to promote neuroplasticity in several neurological disorders. However, its potential effects on inflammation and demyelination are largely unknown. We tested the effectiveness of a preventive, 5-day tDCS treatment started 3 days post-immunization, in reducing the severity of VEP delays observed in early EAE. In mice undergoing cathodal tDCS (n = 6/26 eyes) VEPs were significantly less delayed compared with eyes from EAE-Sham (n = 24/32 eyes) and EAE-Anodal (n = 22/32 eyes). Optic nerve immunohistochemistry revealed a significantly lower cell density of microglia/macrophages, and less axonal loss in EAE-Cathodal vs EAE-Sham and EAE-Anodal, while the percent demyelination with Luxol-fast blue staining was comparable among EAE groups. Considering the latter result, immunofluorescence paranodal staining was performed, revealing a significantly higher number of complete paranode domains in EAE-Cathodal, closer to healthy mice, compared with EAE-Sham and EAE-Anodal groups. These results were reflected by the negative correlation between the number of complete paranode domains and VEP latency increase with respect to pre-immunization. Finally, cathodal tDCS was associated with a lower number, closer to healthy, of single paranodes in contrast to EAE-Sham. The effects of cathodal stimulation in preventing VEPs delays and optic nerve myelin damage were already observed in the pre-motor onset EAE stage, and were associated with a lower density of inflammatory cells. These findings suggest that tDCS may exert an anti-inflammatory effect with potential therapeutic application to be further explored in autoimmune demyelinating diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Optic Neuritis , Transcranial Direct Current Stimulation , Animals , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Evoked Potentials, Visual , Mice , Multiple Sclerosis/complications , Multiple Sclerosis/therapy , Optic Neuritis/therapy
4.
J Neuroimmunol ; 367: 577859, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35395486

ABSTRACT

Retinal ganglion cells (RGC) are lost as a sequela of optic nerve inflammation in myelin oligodendrocyte glycoprotein antibody associated disease (MOGAD), but the mechanisms of injury remain incompletely understood and there remains a need to characterize the murine model of MOGAD. Several studies have shown that RGC loss occurs in association with optic neuritis in MOG35-55 experimental autoimmune encephalomyelitis (EAE), but retinal pathology has not been studied in the double transgenic opticospinal EAE (OSE) model, in which animals develop spontaneous disease associated with MOG35-55 peptide specific T cells and B cells producing MOG-specific antibodies. Herein, we show that at 8-weeks OSE mice develop optic nerve inflammation, reactive astrogliosis, and RGC loss. By 10-weeks of age, affected mice have a 50% reduction in RGCs as compared to age matched wild type mice without EAE. The retinal pathology that ensues from spontaneous optic neuritis in OSE mice mirrors that seen following human optic neuritis and may be a useful model for screening neuroprotective compounds for MOGAD and other diseases with optic neuritis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Optic Neuritis , Retina , Animals , Encephalomyelitis, Autoimmune, Experimental/complications , Hashimoto Disease , Inflammation/pathology , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein , Optic Neuritis/complications , Retina/pathology
5.
Article in English | MEDLINE | ID: mdl-35027475

ABSTRACT

BACKGROUND AND OBJECTIVES: Myelin oligodendrocyte glycoprotein antibody-associated disorder (MOGAD) is a rare, autoimmune demyelinating CNS disorder, distinct from multiple sclerosis and neuromyelitis optica spectrum disorder. Characterized by pathogenic immunoglobulin G (IgG) antibodies against MOG, a potential treatment strategy for MOGAD is to reduce circulating IgG levels, e.g., by interference with the IgG recycling pathway mediated by the neonatal Fc receptor (FcRn). Although the optic nerve is often detrimentally involved in MOGAD, the effect of FcRn blockade on the visual pathway has not been assessed. Our objective was to investigate effects of a monoclonal anti-FcRn antibody in murine MOG-IgG-associated experimental autoimmune encephalomyelitis (EAE). METHODS: We induced active MOG35-55 EAE in C57Bl/6 mice followed by the application of a monoclonal MOG-IgG (8-18C5) 10 days postimmunization (dpi). Animals were treated with either a specific monoclonal antibody against FcRn (α-FcRn, 4470) or an isotype-matched control IgG on 7, 10, and 13 dpi. Neurologic disability was scored daily on a 10-point scale. Visual acuity was assessed by optomotor reflex. Histopathologic hallmarks of disease were assessed in the spinal cord, optic nerve, and retina. Immune cell infiltration was visualized by immunohistochemistry, demyelination by Luxol fast blue staining and complement deposition and number of retinal ganglion cells by immunofluorescence. RESULTS: In MOG-IgG-augmented MOG35-55 EAE, anti-FcRn treatment significantly attenuated neurologic disability over the course of disease (mean area under the curve and 95% confidence intervals (CIs): α-FcRn [n = 27], 46.02 [37.89-54.15]; isotype IgG [n = 24], 66.75 [59.54-73.96], 3 independent experiments), correlating with reduced amounts of demyelination and macrophage infiltration into the spinal cord. T- and B-cell infiltration and complement deposition remained unchanged. Compared with isotype, anti-FcRn treatment prevented reduction of visual acuity over the course of disease (median cycles/degree and interquartile range: α-FcRn [n = 16], 0.50 [0.48-0.55] to 0.50 [0.48-0.58]; isotype IgG [n = 17], 0.50 [0.49-0.54] to 0.45 [0.39-0.51]). DISCUSSION: We show preserved optomotor response and ameliorated course of disease after anti-FcRn treatment in an experimental model using a monoclonal MOG-IgG to mimic MOGAD. Selectively targeting FcRn might represent a promising therapeutic approach in MOGAD.


Subject(s)
Antibodies, Monoclonal/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Histocompatibility Antigens Class I/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Receptors, Fc/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/etiology , Female , Mice , Mice, Inbred C57BL , Vision Disorders
6.
Mol Neurobiol ; 59(3): 1744-1765, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35018577

ABSTRACT

Multiple sclerosis (MS) is an autoimmune and demyelinating disease of the central nervous system (CNS) caused by CNS infiltration of peripheral immune cells, immune-mediated attack of the myelin sheath, neuroinflammation, and/or axonal/neuronal dysfunctions. Some drugs are available to cope with relapsing-remitting MS (RRMS) but there is no therapy for the primary progressive MS (PPMS). Because growing evidence supports a regulatory role of the translocator protein (TSPO) in neuroinflammatory, demyelinating, and neurodegenerative processes, we investigated the therapeutic potential of phenylindolyilglyoxylamydes (PIGAs) TSPO ligands in myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) mice mimicking the human PPMS. MOG-EAE C57Bl/6-mice were treated by TSPO ligands PIGA839, PIGA1138, or the vehicle. Several methods were combined to evaluate PIGAs-TSPO ligand effects on MOG-EAE symptoms, CNS infiltration by immune cells, demyelination, and axonal damages. PIGA1138 (15 mg/kg) drastically reduced MOG-EAE mice clinical scores, ameliorated motor dysfunctions assessed with the Catwalk device, and counteracted MOG-EAE-induced demyelination by preserving Myelin basic protein (MBP) expression in the CNS. Furthermore, PIGA1138-treatment prevented EAE-evoked decreased neurofilament-200 expression in spinal and cerebellar axons. Moreover, PIGA1138 inhibited peripheral immune-CD45 + cell infiltration in the CNS, suggesting that it may control inflammatory mechanisms involved in PPMS. Concordantly, PIGA1138 enhanced anti-inflammatory interleukin-10 serum level in MOG-EAE mice. PIGA1138-treatment, which increased neurosteroid allopregnanolone production, ameliorated all pathological biomarkers, while PIGA839, unable to activate neurosteroidogenesis in vivo, exerted only moderate/partial effects in MOG-EAE mice. Altogether, our results suggest that PIGA1138-based treatment may represent an interesting possibility to be explored for the innovation of effective therapies against PPMS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis, Chronic Progressive , Multiple Sclerosis , Animals , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Ligands , Mice , Mice, Inbred C57BL , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein
7.
Pain ; 163(7): 1346-1355, 2022 07 01.
Article in English | MEDLINE | ID: mdl-34711761

ABSTRACT

ABSTRACT: Primary headache conditions are frequently associated with multiple sclerosis (MS), but the mechanism that triggers or worsens headaches in patients with MS is poorly understood. We previously showed that the proalgesic transient receptor potential ankyrin 1 (TRPA1) mediates hind paw mechanical and cold allodynia in a relapsing-remitting experimental autoimmune encephalomyelitis (RR-EAE) model in mice. Here, we investigated the development of periorbital mechanical allodynia (PMA) in RR-EAE, a hallmark of headache, and if TRPA1 contributed to this response. RR-EAE induction by injection of the myelin oligodendrocyte peptide fragment35-55 (MOG35-55) and Quillaja A adjuvant (Quil A) in C57BL/6J female mice elicited a delayed and sustained PMA. The PMA at day 35 after induction was reduced by the calcitonin gene-related peptide receptor antagonist (olcegepant) and the serotonin 5-HT1B/D receptor agonist (sumatriptan), 2 known antimigraine agents. Genetic deletion or pharmacological blockade of TRPA1 attenuated PMA associated with RR-EAE. The levels of oxidative stress biomarkers (4-hydroxynonenal and hydrogen peroxide, known TRPA1 endogenous agonists) and superoxide dismutase and NADPH oxidase activities were increased in the trigeminal ganglion of RR-EAE mice. Besides, the treatment with antioxidants (apocynin or α-lipoic acid) attenuated PMA. Thus, the results of this study indicate that TRPA1, presumably activated by endogenous agonists, evokes PMA in a mouse model of relapsing-remitting MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis, Relapsing-Remitting , Multiple Sclerosis , Animals , Ankyrins , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/complications , Female , Headache/complications , Hyperalgesia/complications , Hyperalgesia/etiology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/complications , Multiple Sclerosis, Relapsing-Remitting/complications , Multiple Sclerosis, Relapsing-Remitting/drug therapy , TRPA1 Cation Channel/genetics
8.
Pain ; 163(1): e49-e61, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-33863858

ABSTRACT

ABSTRACT: Chronic pain is a highly prevalent symptom associated with the autoimmune disorder multiple sclerosis (MS). The central nucleus of the amygdala plays a critical role in pain processing and modulation. Neuropathic pain alters nociceptive signaling in the central amygdala, contributing to pain chronicity and opioid tolerance. Here, we demonstrate that activated microglia within the central amygdala disrupt nociceptive sensory processing and contribute to pain hypersensitivity in experimental autoimmune encephalomyelitis (EAE), the most frequently used animal model of MS. Male and female mice with EAE exhibited differences in microglial morphology in the central amygdala, which was associated with heat hyperalgesia, impaired morphine reward, and reduced morphine antinociception in females. Animals with EAE displayed a lack of morphine-evoked activity in cells expressing somatostatin within the central amygdala, which drive antinociception. Induction of focal microglial activation in naïve mice via injection of lipopolysaccharide into the central amygdala produced a loss of morphine analgesia in females, similar to as observed in EAE animals. Our data indicate that activated microglia within the central amygdala may contribute to the sexually dimorphic effects of morphine and may drive neuronal adaptations that lead to pain hypersensitivity in EAE. Our results provide a possible mechanism underlying the decreased efficacy of opioid analgesics in the management of MS-related pain, identifying microglial activation as a potential therapeutic target for pain symptoms in this patient population.


Subject(s)
Analgesia , Central Amygdaloid Nucleus , Encephalomyelitis, Autoimmune, Experimental , Neuralgia , Analgesics, Opioid/therapeutic use , Animals , Drug Tolerance , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Humans , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Inflammation , Male , Mice , Morphine/therapeutic use , Neuralgia/drug therapy , Neuralgia/etiology
9.
Article in English | MEDLINE | ID: mdl-34911793

ABSTRACT

BACKGROUND AND OBJECTIVES: To investigate whether the formation or retention of meningeal ectopic lymphoid tissue (mELT) can be inhibited by the sphingosine 1-phosphate receptor 1,5 modulator siponimod (BAF312) in a murine model of multiple sclerosis (MS). METHODS: A murine spontaneous chronic experimental autoimmune encephalomyelitis (EAE) model, featuring meningeal inflammatory infiltrates resembling those in MS, was used. To prevent or treat EAE, siponimod was administered daily starting either before EAE onset or at peak of disease. The extent and cellular composition of mELT, the spinal cord parenchyma, and the spleen was assessed by histology and immunohistochemistry. RESULTS: Siponimod, when applied before disease onset, ameliorated EAE. This effect was also present, although less prominent, when treatment started at peak of disease. Treatment with siponimod resulted in a strong reduction of the extent of mELT in both treatment paradigms. Both B and T cells were diminished in the meningeal compartment. DISCUSSION: Beneficial effects on the disease course correlated with a reduction in mELT, suggesting that inhibition of mELT may be an additional mechanism of action of siponimod in the treatment of EAE. Further studies are needed to establish causality and confirm this observation in MS.


Subject(s)
Azetidines/pharmacology , Benzyl Compounds/pharmacology , Encephalomyelitis, Autoimmune, Experimental , Meninges/drug effects , Multiple Sclerosis , Sphingosine 1 Phosphate Receptor Modulators/pharmacology , Tertiary Lymphoid Structures , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Humans , Meninges/immunology , Mice , Multiple Sclerosis/complications , Multiple Sclerosis/drug therapy , Multiple Sclerosis/prevention & control , Tertiary Lymphoid Structures/drug therapy , Tertiary Lymphoid Structures/etiology , Tertiary Lymphoid Structures/prevention & control
10.
Immunohorizons ; 5(10): 844-854, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702761

ABSTRACT

We recently demonstrated how sepsis influences the subsequent development of experimental autoimmune encephalomyelitis (EAE) presented a conceptual advance in understanding the postsepsis chronic immunoparalysis state. However, the reverse scenario (autoimmunity prior to sepsis) defines a high-risk patient population whose susceptibility to sepsis remains poorly defined. In this study, we present a retrospective analysis of University of Iowa Hospital and Clinics patients demonstrating increased sepsis prevalence among multiple sclerosis (MS), relative to non-MS, patients. To interrogate how autoimmune disease influences host susceptibility to sepsis, well-established murine models of MS and sepsis and EAE and cecal ligation and puncture, respectively, were used. EAE, relative to non-EAE, mice were highly susceptible to sepsis-induced mortality with elevated cytokine storms. These results were further recapitulated in LPS and Streptococcus pneumoniae sepsis models. This work highlights both the relevance of identifying highly susceptible patient populations and expands the growing body of literature that host immune status at the time of septic insult is a potent mortality determinant.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/complications , Multiple Sclerosis/complications , Pneumococcal Infections/immunology , Sepsis/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Datasets as Topic , Disease Susceptibility/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/mortality , Female , Humans , Male , Mice , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/mortality , Pneumococcal Infections/epidemiology , Pneumococcal Infections/microbiology , Prevalence , Retrospective Studies , Risk Factors , Sepsis/epidemiology , Sepsis/microbiology , Streptococcus pneumoniae/immunology , Young Adult
11.
Neurotox Res ; 39(6): 1880-1891, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34487326

ABSTRACT

Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system presented by autoimmune manifestations. This study aimed at investigating the effects of apamin administration on the activated T cell population in an experimental autoimmune encephalomyelitis (EAE) MS model. Thirty mice underwent EAE induction and were then randomly divided into 5 groups. Three groups received 10, 50, and 100 µg/kg apamin; the fourth group received 1 mg/kg dexamethasone; and the fifth group received the equivalent amount of PBS (phosphate-buffered saline) intraperitoneally. Peripheral CD4 + cell and memory T cell distribution was measured with a flow cytometer every week. Also, CD4 + and CD8 + cell infiltration to the brain was assessed with immunohistochemistry. It was observed that the group receiving 50 µg/kg apamin had a lower EAE score in comparison with the groups receiving 100 µg/kg apamin (p 0.014). Also, peripheral blood memory cells with CD44 + , CD62L - , and CD4 + markers were decreased in apamin-administered groups. Regarding the infiltrated CD8 + cells, a significant decrease (p 0.002) was observed in the group receiving 50 µg/kg apamin compared with the control group. These results indicate that 50-µg/kg doses of apamin had an effective treatment over 14 days; it reduced both the severity of symptoms and the infiltration of CD8 + cells into the CNS. Moreover, it increased myelin density and decreased the circulation of CD62L - , CD44L - , and CD44 + memory T cells. So, it appears that apamin plays a critical role in regulating immunity and reducing the complications of autoimmune MS.


Subject(s)
Apamin/therapeutic use , Blood-Brain Barrier , Multiple Sclerosis/drug therapy , T-Lymphocytes/drug effects , Animals , Apamin/pharmacology , Blood-Brain Barrier/drug effects , Dexamethasone/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Multiple Sclerosis/etiology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Myelin Sheath/pathology
12.
J Transl Med ; 19(1): 317, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34301274

ABSTRACT

BACKGROUND: Constipation is a common gastrointestinal dysfunction which has a potential impact on people's immune state and their quality of life. Here we investigated the effects of constipation on experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). METHODS: Constipation was induced by loperamide in female C57BL/6 mice. The alternations of gut microbiota, permeability of intestinal barrier and blood-brain barrier, and histopathology of colon were assessed after constipation induction. EAE was induced in the constipation mice. Fecal microbiota transplantation (FMT) was performed from constipation mice into microbiota-depleted mice. Clinical scores, histopathology of inflammation and demyelination, Treg/Th17 and Treg17/Teff17 imbalance both in the peripheral lymphatic organs and central nervous system, cytokines include TGF-ß, GM-CSF, IL-10, IL-17A, IL-17F, IL-21, IL-22, and IL-23 in serum were assessed in different groups. RESULTS: Compared with the vehicle group, the constipation mice showed gut microbiota dysbiosis, colon inflammation and injury, and increased permeability of intestinal barrier and blood-brain barrier. We found that the clinical and pathological scores of the constipation EAE mice were severer than that of the EAE mice. Compared with the EAE mice, the constipation EAE mice showed reduced percentage of Treg and Treg17 cells, increased percentage of Th17 and Teff17 cells, and decreased ratio of Treg/Th17 and Treg17/Teff17 in the spleen, inguinal lymph nodes, brain, and spinal cord. Moreover, the serum levels of TGF-ß, IL-10, and IL-21 were decreased while the GM-CSF, IL-17A, IL-17F, IL-22, and IL-23 were increased in the constipation EAE mice. In addition, these pathological processes could be transferred via their gut microbiota. CONCLUSIONS: Our results verified that constipation induced gut microbiota dysbiosis exacerbated EAE via aggravating Treg/Th17 and Treg17/Teff17 imbalance and cytokines disturbance in C57BL/6 mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Gastrointestinal Microbiome , Animals , Constipation , Cytokines , Dysbiosis/complications , Encephalomyelitis, Autoimmune, Experimental/complications , Female , Mice , Mice, Inbred C57BL , Quality of Life , Th17 Cells
13.
Article in English | MEDLINE | ID: mdl-33551096

ABSTRACT

Multiple sclerosis (MS), a disease in which the immune system attacks nerve cells, has been associated with both genetic and environmental risk factors. We observed increased micronucleus (MN) formation in SJL/J mouse experimental autoimmune encephalomyelitis (EAE), an animal model of MS. Most of these MN were due to chromosomal loss. Increased activation of MAP kinases, which leads to disruption of the mitotic spindle and improper segregation of chromosomes, is associated with MS. MAP kinase inhibitors, such as PD98059, may therefore be beneficial for MS. In the EAE model, PD98059 treatment reduced adverse effects, including MN formation, lipid peroxidation, and GSH oxidation. Interventions that mitigate chromosomal instability may have therapeutic value in MS.


Subject(s)
Chromosomal Instability/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Flavonoids/pharmacology , Mitogen-Activated Protein Kinases/chemistry , Multiple Sclerosis/drug therapy , Animals , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Mice , Multiple Sclerosis/complications , Multiple Sclerosis/pathology
14.
ASN Neuro ; 13: 1759091421991771, 2021.
Article in English | MEDLINE | ID: mdl-33541127

ABSTRACT

Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system. Recent clinical study suggested that MS patient exhibited acute heart failure. Further, 12-lead electrocardiographic study showed a longer QTc interval in both MS patient and experimental autoimmune encephalomyelitis (EAE) Lewis rat. However, there is limited study regarding the effect of sex on cardiac injury in EAE. To our knowledge, sex effect on cardiac damage in mice with EAE has not yet been published. Herein, we examined the role of the immune system in mediating cardiac dysfunction after EAE in female and male mice. Neurological function was subsequently evaluated and cardiac function was assessed by echocardiography at multiple time points after EAE. EAE mice exhibited severe neurological deficit and significant cardiac dysfunction, including decreased left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS) at 1 and 2 months after EAE induction. Meanwhile male EAE presented increased expression of the oxidative stress (e.g., nicotinamaide adenine dinucleotide phosphate oxidase-2; NOX-2) in heart, as well as cardiac hypertrophy, increased left ventricle (LV) mass and more severe cardiac fibrosis compared with male control mice. In addition, male EAE mice showed significantly increased cardiac canonical inflammatory mediator (e.g., monocyte chemoattractant protein-1; MCP-1, transforming growth factor-ß; TGF-ß and toll-like receptor 2; TLR-2) compared with female EAE mice at 2 months after EAE induction. In conclusion, EAE increases inflammatory factor expression and aggravates cardiac dysfunction in male mice compared with female mice, which may contribute to different cardiac outcome in EAE mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Encephalomyelitis, Autoimmune, Experimental/complications , Female , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred Lew , Stroke Volume , Ventricular Function, Left
15.
PLoS One ; 16(1): e0243862, 2021.
Article in English | MEDLINE | ID: mdl-33406093

ABSTRACT

ST266 is the biological secretome of cultured Amnion-derived Multipotent Progenitor cells containing multiple growth factors and cytokines. While intranasally-administered ST266 improves the phenotype in experimental optic neuritis, specific ST266 components mediating these effects are not known. We compared the effects of ST266 with and without removal of large molecular weight proteins both in vitro and in the multiple sclerosis model experimental autoimmune encephalomyelitis (EAE) in C57BL/6J mice. Mice were treated daily with intranasal vehicle, ST266 or lower molecular weight fraction of ST266. Retinal ganglion cells were counted in isolated retinas, and optic nerves were assessed for inflammation and demyelination. ST266 treatment significantly improved retinal ganglion cell survival and reduced optic nerve demyelination in EAE mice. The lower molecular weight ST266 fraction significantly improved optic nerve demyelination, but only showed a trend towards improved retinal ganglion cell survival. ST266 fractions below 50kDa increased Schwann cell proliferation in vitro, but were less effective than non-fractionated ST266. Demyelination attenuation was partially associated with the lower molecular weight ST266 fraction, but removal of higher molecular weight biomolecules from ST266 diminishes its neuroprotective effects, suggesting at least some high molecular weight proteins play a role in ST266-mediated neuroprotection.


Subject(s)
Amnion/cytology , Multipotent Stem Cells/cytology , Neuroprotection , Animals , Cell Proliferation , Demyelinating Diseases/complications , Demyelinating Diseases/pathology , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Mice, Inbred C57BL , Molecular Weight , Myelin-Oligodendrocyte Glycoprotein , Optic Nerve/pathology , Optic Neuritis/complications , Optic Neuritis/pathology , Peptides , Retinal Ganglion Cells/pathology , Schwann Cells/pathology
16.
Behav Brain Res ; 396: 112896, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32905811

ABSTRACT

Multiple sclerosis (MS) is associated with burdensome memory impairments and preclinical literature suggests that these impairments are linked to neuroinflammation. Previously, we have shown that toll-like receptor 4 (TLR4) antagonists, such as (+)-naltrexone [(+)-NTX], block neuropathic pain and associated spinal inflammation in rats. Here we extend these findings to first demonstrate that (+)-NTX blocks TLR2 in addition to TLR4. Additionally, we examined in two rat strains whether (+)-NTX could attenuate learning and memory disturbances and associated neuroinflammation using a low-dose experimental autoimmune encephalomyelitis (EAE) model of MS. EAE is the most commonly used experimental model for the human inflammatory demyelinating disease, MS. This low-dose model avoided motor impairments that would confound learning and memory measurements. Fourteen days later, daily subcutaneous (+)-NTX or saline injections began and continued throughout the study. Contextual and auditory-fear conditioning were conducted at day 21 to assess hippocampal and amygdalar function. With this low-dose model, EAE impaired long-term, but not short-term, contextual fear memory; both long-term and short-term auditory-cued fear memory were spared. This was associated with increased mRNA for hippocampal interleukin-1ß (IL-1ß), TLR2, TLR4, NLRP3, and IL-17 and elevated expression of the microglial marker Iba1 in CA1 and DG regions of the hippocampus, confirming the neuroinflammation observed in higher-dose EAE models. Importantly, (+)-NTX completely prevented the EAE-induced memory impairments and robustly attenuated the associated proinflammatory effects. These findings suggest that (+)-NTX may exert therapeutic effects on memory function by dampening the neuroinflammatory response in the hippocampus through blockade of TLR2/TLR4. This study suggests that TLR2 and TLR4 antagonists may be effective at treating MS-related memory deficits.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/complications , Hippocampus/drug effects , Hippocampus/immunology , Inflammation/etiology , Inflammation/prevention & control , Memory Disorders/etiology , Memory Disorders/prevention & control , Multiple Sclerosis/complications , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cells, Cultured , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Fear/drug effects , Fear/physiology , Male , Mice , Microglia/drug effects , Microglia/immunology , Naltrexone/administration & dosage , Narcotic Antagonists/administration & dosage , Rats , Rats, Sprague-Dawley , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors
17.
Immunology ; 161(3): 209-229, 2020 11.
Article in English | MEDLINE | ID: mdl-32687611

ABSTRACT

Nanoparticles (NPs) displaying autoimmune disease-relevant peptide-major histocompatibility complex class II molecules (pMHCII-NPs) trigger cognate T-regulatory type 1 (Tr1)-cell formation and expansion, capable of reversing organ-specific autoimmune responses. These pMHCII-NPs that display epitopes from mitochondrial protein can blunt the progression of both autoimmune hepatitis (AIH) and experimental autoimmune encephalomyelitis (EAE) in mice carrying either disease. However, with co-morbid mice having both diseases, these pMHCII-NPs selectively treat AIH. In contrast, pMHCII-NPs displaying central nervous system (CNS)-specific epitopes can efficiently treat CNS autoimmunity, both in the absence and presence of AIH, without having any effects on the progression of the latter. Here, we develop a compartmentalized population model of T-cells in co-morbid mice to identify the mechanisms by which Tr1 cells mediate organ-specific immunoregulation. We perform time-series simulations and bifurcation analyses to study how varying physiological parameters, including local cognate antigenic load and rates of Tr1-cell recruitment and retention, affect T-cell allocation and Tr1-mediated immunoregulation. Various regimes of behaviour, including 'competitive autoimmunity' where pMHCII-NP-treatment fails against both diseases, are identified and compared with experimental observations. Our results reveal that a transient delay in Tr1-cell recruitment to the CNS, resulting from inflammation-dependent Tr1-cell allocation, accounts for the liver-centric effects of AIH-specific pMHCII-NPs in co-morbid mice as compared with mice exclusively having EAE. They also suggest that cognate autoantigen expression and local Tr1-cell retention are key determinants of effective regulatory-cell function. These results thus provide new insights into the rules that govern Tr1-cell recruitment and their autoregulatory function.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Hepatitis, Autoimmune/immunology , Models, Immunological , Models, Theoretical , Multiple Sclerosis/immunology , Nanomedicine/methods , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation , Autoantigens/immunology , Cell Compartmentation , Encephalomyelitis, Autoimmune, Experimental/complications , Hepatitis, Autoimmune/complications , Humans , Immunomodulation , Lymphocyte Activation , Mice
18.
Mol Neurobiol ; 57(5): 2420-2435, 2020 May.
Article in English | MEDLINE | ID: mdl-32095993

ABSTRACT

Central neuropathic pain is a common untreated symptom in progressive multiple sclerosis (PMS) and is associated with poor quality of life and interference with patients' daily activities. The neuroinflammation process and mitochondrial dysfunction in the PMS lesions generate reactive species. The transient potential receptor ankyrin 1 (TRPA1) has been identified as one of the major mechanisms that contribute to neuropathic pain signaling and can be activated by reactive compounds. Thus, the goal of our study was to evaluate the role of spinal TRPA1 in the central neuropathic pain observed in a PMS model in mice. We used C57BL/6 female mice (20-30 g), and the PMS model was induced by the experimental autoimmune encephalomyelitis (EAE) using mouse myelin oligodendrocyte glycoprotein (MOG35-55) antigen and CFA (complete Freund's adjuvant). Mice developed progressive clinical score, with motor impairment observed after 15 days of induction. This model induced mechanical and cold allodynia and heat hyperalgesia which were measured up to 14 days after induction. The hypersensitivity observed was reduced by the administration of selective TRPA1 antagonists (HC-030031 and A-967079, via intrathecal and intragastric), antioxidants (α-lipoic acid and apocynin, via intrathecal and intragastric), and TRPA1 antisense oligonucleotide (via intrathecal). We also observed an increase in TRPA1 mRNA levels, NADPH oxidase activity, and 4-hydroxinonenal (a TRPA1 agonist) levels in spinal cord samples of PMS-EAE induced animals. In conclusion, these results support the hypothesis of the TRPA1 receptor involvement in nociception observed in a PMS-EAE model in mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/complications , Hyperalgesia/physiopathology , Nerve Tissue Proteins/physiology , Neuralgia/physiopathology , Nociception/physiology , Spinal Cord/physiopathology , TRPA1 Cation Channel/physiology , Acetanilides/pharmacology , Acetanilides/therapeutic use , Acetophenones/pharmacology , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Antipyrine/analogs & derivatives , Antipyrine/pharmacology , Antipyrine/therapeutic use , Dipyrone/pharmacology , Dipyrone/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein/toxicity , NADPH Oxidases/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuralgia/drug therapy , Neuralgia/etiology , Nociception/drug effects , Oligonucleotides, Antisense/pharmacology , Oxidative Stress , Oximes/pharmacology , Oximes/therapeutic use , Peptide Fragments/immunology , Peptide Fragments/toxicity , Pregabalin/pharmacology , Pregabalin/therapeutic use , Purines/pharmacology , Purines/therapeutic use , TRPA1 Cation Channel/antagonists & inhibitors , TRPA1 Cation Channel/biosynthesis , TRPA1 Cation Channel/genetics , Thioctic Acid/pharmacology , Up-Regulation/drug effects
19.
Exp Neurol ; 328: 113241, 2020 06.
Article in English | MEDLINE | ID: mdl-32045597

ABSTRACT

Central neuropathic pain is the main symptom caused by spinal cord lesion in relapsing-remitting multiple sclerosis (RRMS), but its management is still not effective. The transient receptor potential ankyrin 1 (TRPA1) is a pain detecting ion channel involved in neuropathic pain development. Thus, the aim of our study was to evaluate the role of TRPA1 in central neuropathic nociception induced by relapsing-remitting experimental autoimmune encephalomyelitis (RR-EAE) mouse model. In this model, we observed the development of similar clinical conditions of RRMS in C57BL/6 female mice through RR-EAE using MOG35-55 antigen and Quil A adjuvant. At the thirty-fifth day post-induction, C57BL/6 female mice demonstrated alteration in the RR-EAE score without motor impairment, mechanical and cold allodynia. Also, significative changes in demyelinating (Mog and olig-1) and neuroinflammatory (Iba1, Gfap and Tnfa) markers were observed, but this model did not alter Trpa1 RNA expression levels in the spinal cord. The hydrogen peroxide and 4-hydroxynonenal levels (TRPA1 agonists) were increased in RR-EAE induced mice, as well as the NADPH oxidase activity. The intragastric treatment of RR-EAE induced mice with TRPA1 antagonists (HC-030031 and A-967079) and antioxidant (α-lipoic acid and apocynin) caused an antiallodynic effect. Moreover, the intrathecal administration of TRPA1 antisense oligonucleotide, HC-030031, α-lipoic acid, and apocynin transiently attenuated mechanical and cold allodynia. Thus, TRPA1 plays a key role in the induction of neuropathic pain in this model of RR-EAE and can be a possible target for investigating the development of pain in RRMS patients.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Hyperalgesia/metabolism , Neuralgia/metabolism , Nociception/physiology , TRPA1 Cation Channel/metabolism , Animals , Encephalomyelitis, Autoimmune, Experimental/complications , Female , Hyperalgesia/etiology , Mice , Mice, Inbred C57BL , Neuralgia/etiology
20.
Article in English | MEDLINE | ID: mdl-31467038

ABSTRACT

OBJECTIVE: To measure the impact of JHU-083, a novel prodrug of the glutamine antagonist 6-diazo-5-oxo-l-norleucine, on immune cell proliferation and activation, along with physical and cognitive impairments associated with the experimental autoimmune encephalomyelitis (EAE) mouse model of MS. METHODS: Splenic-derived T cells and bone marrow-derived dendritic cells (DCs) were cultured, activated, and treated daily with vehicle or JHU-083. Proliferation and activation were measured via flow cytometry and IncuCyte live cell analysis. C57BL/6 mice were immunized for EAE. Vehicle or JHU-083 was administered orally every other day either from the time of immunization in the prevention paradigm or from the time of disease onset in the treatment paradigm. Disease scores and body weight were monitored. In the treatment paradigm, cognition was evaluated using the Barnes maze test. RESULTS: JHU-083 selectively inhibits T-cell proliferation and decreases T-cell activation, with no effect on DCs. In vivo, orally administered JHU-083 significantly decreases EAE severity in both prevention and treatment paradigms and reverses EAE-induced cognitive impairment. CONCLUSIONS: JHU-083, a well-tolerated, brain penetrable glutamine antagonist, is a promising novel treatment for both the physical and cognitive deficits of MS.


Subject(s)
Azo Compounds/pharmacology , Caproates/pharmacology , Cognitive Dysfunction/drug therapy , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Multiple Sclerosis/drug therapy , Animals , Behavior, Animal/drug effects , Cells, Cultured , Cognitive Dysfunction/etiology , Encephalomyelitis, Autoimmune, Experimental/complications , Female , Glutamine , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...