Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.112
Filter
1.
NMR Biomed ; 37(6): e5127, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38450807

ABSTRACT

Multiple sclerosis (MS) is an autoimmune degenerative disease targeting white matter in the central nervous system. The most common animal model that mimics MS is experimental autoimmune encephalomyelitis (EAE) and it plays a crucial role in pharmacological research, from the identification of a therapeutic target to the in vivo validation of efficacy. Magnetic resonance imaging (MRI) is largely used to detect MS lesions, and resting-state functional MRI (rsfMRI) to investigate alterations in the brain functional connectivity (FC). MRI was mainly used in EAE studies to detect lesions in the spinal cord and brain. The current longitudinal MRI study aims to validate rsfMRI as a biomarker of the disease progression in the myelin oligodendrocyte glycoprotein 35-55 induced EAE animal model of MS. MR images were acquired 14, 25, and 50 days postimmunization. Seed-based analysis was used to investigate the whole-brain FC with some predefined areas, such as the thalamic regions, cerebellum, motor and somatosensory cortex. When compared with the control group, the EAE group exhibited a slightly altered FC and a decreasing trend in the total number of activated voxels along the disease progression. The most interesting result regards the whole-brain FC with the cerebellum. A hyperconnectivity behavior was found at an early phase and a significant reduced connectivity at a late phase. Moreover, we found a negative correlation between the total number of activated voxels during the late phase and the cumulative disease index. The results obtained provide a clinically relevant experimental platform that may be pivotal for the elucidation of the key mechanisms of accumulation of irreversible disability, as well as the development of innovative therapies for MS. Moreover, the negative correlation between the disease severity and the size of the activated area suggests a possible research pathway to follow for the resolution of the clinico-radiological paradox.


Subject(s)
Brain , Encephalomyelitis, Autoimmune, Experimental , Magnetic Resonance Imaging , Rest , Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Animals , Female , Brain/diagnostic imaging , Brain/physiopathology , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/physiopathology , Disease Models, Animal
2.
Sci Rep ; 12(1): 20995, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36470947

ABSTRACT

Multiple Sclerosis (MS) is an autoimmune disease with notable sex differences. Women are not only more likely to develop MS but are also more likely than men to experience neuropathic pain in the disease. It has been postulated that neuropathic pain in MS can originate in the peripheral nervous system at the level of the dorsal root ganglia (DRG), which houses primary pain sensing neurons (nociceptors). These nociceptors become hyperexcitable in response to inflammation, leading to peripheral sensitization and eventually central sensitization, which maintains pain long-term. The mouse model experimental autoimmune encephalomyelitis (EAE) is a good model for human MS as it replicates classic MS symptoms including pain. Using EAE mice as well as naïve primary mouse DRG neurons cultured in vitro, we sought to characterize sex differences, specifically in peripheral sensory neurons. We found sex differences in the inflammatory profile of the EAE DRG, and in the TNFα downstream signaling pathways activated intracellularly in cultured nociceptors. We also found increased cell death with TNFα treatment. Given that TNFα signaling has been shown to initiate intrinsic apoptosis through mitochondrial disruption, this led us to investigate sex differences in the mitochondria's response to TNFα. Our results demonstrate that male sensory neurons are more sensitive to mitochondrial stress, making them prone to neuronal injury. In contrast, female sensory neurons appear to be more resistant to mitochondrial stress and exhibit an inflammatory and regenerative phenotype that may underlie greater nociceptor hyperexcitability and pain. Understanding these sex differences at the level of the primary sensory neuron is an important first step in our eventual goal of developing sex-specific treatments to halt pain development in the periphery before central sensitization is established.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Ganglia, Spinal , Multiple Sclerosis , Neuralgia , Sex Characteristics , Animals , Female , Humans , Male , Mice , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Ganglia, Spinal/physiopathology , Multiple Sclerosis/physiopathology , Neuralgia/etiology , Neuralgia/physiopathology , Nociceptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
Front Immunol ; 13: 824411, 2022.
Article in English | MEDLINE | ID: mdl-35211120

ABSTRACT

Multiple Sclerosis (MS) is a neurodegenerative autoimmune disorder of the central nervous system (CNS) characterized by the recruitment of self-reactive T lymphocytes, mainly inflammatory T helper (Th) cell subsets. Once recruited within the CNS, inflammatory Th cells produce several inflammatory cytokines and chemokines that activate resident glial cells, thus contributing to the breakdown of blood-brain barrier (BBB), demyelination and axonal loss. Astrocytes are recognized as key players of MS immunopathology, which respond to Th cell-defining cytokines by acquiring a reactive phenotype that amplify neuroinflammation into the CNS and contribute to MS progression. In this review, we summarize current knowledge of the astrocytic changes and behaviour in both MS and experimental autoimmune encephalomyelitis (EAE), and the contribution of pathogenic Th1, Th17 and Th1-like Th17 cell subsets, and CD8+ T cells to the morphological and functional modifications occurring in astrocytes and their pathological outcomes.


Subject(s)
Astrocytes/physiology , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Humans , Inflammation/immunology , T-Lymphocytes, Helper-Inducer/classification
4.
Front Immunol ; 13: 755900, 2022.
Article in English | MEDLINE | ID: mdl-35185870

ABSTRACT

The key role of B cells in the pathophysiology of multiple sclerosis (MS) is supported by the presence of oligoclonal bands in the cerebrospinal fluid, by the association of meningeal ectopic B cell follicles with demyelination, axonal loss and reduction of astrocytes, as well as by the high efficacy of B lymphocyte depletion in controlling inflammatory parameters of MS. Here, we use a spontaneous model of experimental autoimmune encephalomyelitis (EAE) to study the clonality of the B cell response targeting myelin oligodendrocyte glycoprotein (MOG). In particular, 94% of SJL/j mice expressing an I-As: MOG92-106 specific transgenic T cell receptor (TCR1640) spontaneously develop a chronic paralytic EAE between the age of 60-500 days. The immune response is triggered by the microbiota in the gut-associated lymphoid tissue, while there is evidence that the maturation of the autoimmune demyelinating response might occur in the cervical lymph nodes owing to local brain drainage. Using MOG-protein-tetramers we tracked the autoantigen-specific B cells and localized their enrichment to the cervical lymph nodes and among the brain immune infiltrate. MOG-specific IgG1 antibodies were detected in the serum of diseased TCR1640 mice and proved pathogenic upon adoptive transfer into disease-prone recipients. The ontogeny of the MOG-specific humoral response preceded disease onset coherent with their contribution to EAE initiation. This humoral response was, however, not sufficient for disease induction as MOG-antibodies could be detected at the age of 69 days in a model with an average age of onset of 197 days. To assess the MOG-specific B cell repertoire we FACS-sorted MOG-tetramer binding cells and clonally expand them in vitro to sequence the paratopes of the IgG heavy chain and kappa light chains. Despite the fragility of clonally expanding MOG-tetramer binding effector B cells, our results indicate the selection of a common CDR-3 clonotype among the Igk light chains derived from both disease-free and diseased TCR1640 mice. Our study demonstrates the pre-clinical mobilization of the MOG-specific B cell response within the brain-draining cervical lymph nodes, and reiterates that MOG antibodies are a poor biomarker of disease onset and progression.


Subject(s)
B-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Autoantibodies/immunology , Autoantigens/immunology , B-Lymphocytes/cytology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein/genetics
5.
Cell Rep ; 37(10): 110094, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34879272

ABSTRACT

Cognitive impairment (CI) is a disabling concomitant of multiple sclerosis (MS) with a complex and controversial pathogenesis. The cytokine interleukin-17A (IL-17A) is involved in the immune pathogenesis of MS, but its possible effects on synaptic function and cognition are still largely unexplored. In this study, we show that the IL-17A receptor (IL-17RA) is highly expressed by hippocampal neurons in the CA1 area and that exposure to IL-17A dose-dependently disrupts hippocampal long-term potentiation (LTP) through the activation of its receptor and p38 mitogen-activated protein kinase (MAPK). During experimental autoimmune encephalomyelitis (EAE), IL-17A overexpression is paralleled by hippocampal LTP dysfunction. An in vivo behavioral analysis shows that visuo-spatial learning abilities are preserved when EAE is induced in mice lacking IL-17A. Overall, this study suggests a key role for the IL-17 axis in the neuro-immune cross-talk occurring in the hippocampal CA1 area and its potential involvement in synaptic dysfunction and MS-related CI.


Subject(s)
Behavior, Animal , CA1 Region, Hippocampal/metabolism , Cognition , Encephalomyelitis, Autoimmune, Experimental/metabolism , Interleukin-17/metabolism , Neuronal Plasticity , Receptors, Interleukin-17/metabolism , Synapses/metabolism , Animals , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Encephalomyelitis, Autoimmune, Experimental/psychology , Interleukin-17/genetics , Long-Term Potentiation , Male , Mice, Biozzi , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-17/genetics , Signal Transduction , Spatial Learning , Synapses/pathology , p38 Mitogen-Activated Protein Kinases
6.
Toxins (Basel) ; 13(11)2021 11 22.
Article in English | MEDLINE | ID: mdl-34822611

ABSTRACT

Multiple sclerosis (MS) is a demyelinating disease of inflammatory and autoimmune origin, which induces sensory and progressive motor impairments, including pain. Cells of the immune system actively participate in the pathogenesis and progression of MS by inducing neuroinflammation, tissue damage, and demyelination. Crotalphine (CRO), a structural analogue to a peptide firstly identified in Crotalus durissus terrificus snake venom, induces analgesia by endogenous opioid release and type 2 cannabinoid receptor (CB2) activation. Since CB2 activation downregulates neuroinflammation and ameliorates symptoms in mice models of MS, it was presently investigated whether CRO has a beneficial effect in the experimental autoimmune encephalomyelitis (EAE). CRO was administered on the 5th day after immunization, in a single dose, or five doses starting at the peak of disease. CRO partially reverted EAE-induced mechanical hyperalgesia and decreased the severity of the clinical signs. In addition, CRO decreases the inflammatory infiltrate and glial cells activation followed by TNF-α and IL-17 downregulation in the spinal cord. Peripherally, CRO recovers the EAE-induced impairment in myelin thickness in the sciatic nerve. Therefore, CRO interferes with central and peripheral neuroinflammation, opening perspectives to MS control.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Neuroinflammatory Diseases/drug therapy , Pain/drug therapy , Peptides/pharmacology , Analgesics/pharmacology , Animals , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Hyperalgesia/drug therapy , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Multiple Sclerosis/physiopathology , Receptor, Cannabinoid, CB2/drug effects , Receptor, Cannabinoid, CB2/metabolism
7.
Amino Acids ; 53(11): 1749-1761, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34642824

ABSTRACT

Muscle weakness and fatigue are primary manifestations of multiple sclerosis (MS), a chronic disease of the central nervous system. Interventions that enhance muscle function may improve overall physical well-being of MS patients. Recently, we described that levels of carnosine, an endogenous muscle dipeptide involved in contractile function and fatigue-resistance, are reduced in muscle tissue from MS patients and a monophasic rodent MS model (experimental autoimmune encephalomyelitis, EAE). In the present study, we aimed to (1) confirm this finding in a chronic EAE model, along with the characterization of structural and functional muscle alterations, and (2) investigate the effect of carnosine supplementation to increase/restore muscle carnosine levels and improve muscle function in EAE. We performed muscle immunohistochemistry and ex vivo contractility measurements to examine muscle structure and function at different stages of EAE, and following nutritional intervention (oral carnosine: 3, 15 or 30 g/L in drinking water). Immunohistochemistry revealed progressively worsening muscle fiber atrophy and a switch towards a fast-twitch muscle phenotype during EAE. Using ex vivo muscle contractility experiments, we observed reductions in muscle strength and contraction speed, but no changes in muscle fatigability of EAE mice. However, carnosine levels were unaltered during all stages of EAE, and even though oral carnosine supplementation dose-dependently increased muscle carnosine levels up to + 94% after 56 days EAE, this did not improve muscle function of EAE mice. In conclusion, EAE mice display significant, yet time-dependent, muscular alterations, and carnosine intervention does not improve muscle function in EAE.


Subject(s)
Carnosine/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Multiple Sclerosis/metabolism , Muscle, Skeletal/physiopathology , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Humans , Mice, Inbred C57BL , Multiple Sclerosis/physiopathology , Muscle Contraction
8.
Cell Death Dis ; 12(10): 907, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34611127

ABSTRACT

Cholesterols are the main components of myelin, and are mainly synthesized in astrocytes and transported to oligodendrocytes and neurons in the adult brain. It has been reported that Hippo/yes-associated protein (YAP) pathways are involved in cholesterol synthesis in the liver, however, it remains unknown whether YAP signaling can prevent the demyelination through promoting cholesterol synthesis in experimental autoimmune encephalomyelitis (EAE), a commonly used animal model of multiple sclerosis characterized by neuroinflammation and demyelination. Here, we found that YAP was upregulated and activated in astrocytes of spinal cords of EAE mice through suppression of the Hippo pathway. YAP deletion in astrocytes aggravated EAE with earlier onset, severer inflammatory infiltration, demyelination, and more loss of neurons. Furthermore, we found that the neuroinflammation was aggravated and the proliferation of astrocytes was decreased in YAPGFAP-CKO EAE mice. Mechanically, RNA-seq revealed that the expression of cholesterol-synthesis pathway genes such as HMGCS1 were decreased in YAP-/- astrocytes. qPCR, western blot, and immunostaining further confirmed the more significant reduction of HMGCS1 in spinal cord astrocytes of YAPGFAP-CKO EAE mice. Interestingly, upregulation of cholesterol-synthesis pathways by diarylpropionitrile (DPN) (an ERß-ligand, to upregulate the expression of HMGCS1) treatment partially rescued the demyelination deficits in YAPGFAP-CKO EAE mice. Finally, activation of YAP by XMU-MP-1 treatment promoted the expression of HMGCS1 in astrocytes and partially rescued the demyelination and inflammatory infiltration deficits in EAE mice. These findings identify unrecognized functions of astrocytic YAP in the prevention of demyelination through promoting cholesterol synthesis in EAE, and reveal a novel pathway of YAP/HMGCS1 for cholesterol synthesis in EAE pathology.


Subject(s)
Astrocytes/metabolism , Cholesterol/biosynthesis , Demyelinating Diseases/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Gene Expression Regulation , Animals , Astrocytes/pathology , Body Weight , Cell Proliferation , Down-Regulation/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Hippo Signaling Pathway , Inflammation/pathology , Mice, Knockout , Models, Biological , Neurons/metabolism , Neurons/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recovery of Function , Spinal Cord/pathology , Spinal Cord/ultrastructure , Up-Regulation/genetics , YAP-Signaling Proteins/deficiency , YAP-Signaling Proteins/metabolism
9.
J Neuroimmunol ; 361: 577730, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34628133

ABSTRACT

Motor disability in multiple sclerosis is often modeled using experimental autoimmune encephalomyelitis (EAE) and assessed using the clinical score (CS), an observer-dependent tool that can lead to potential bias. The Advanced Dynamic Weight Bearing (ADWB) system was evaluated as an observer-independent measurement of EAE symptoms. ADWB detected weight shifts onto the front paws as mice develop hindlimb motor disability. CS and ADWB were strongly correlated, indicated that these measures are comparable and suggesting that ADWB may be an appropriate observer-independent tool for the assessment of EAE progression.


Subject(s)
Disability Evaluation , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Weight-Bearing , Animals , Disease Progression , Female , Hindlimb/physiopathology , Mice , Mice, Inbred C57BL , Motor Activity , Severity of Illness Index , Single-Blind Method
10.
J Neuroimmunol ; 360: 577700, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34482269

ABSTRACT

Gut microbiota composition may affect the central nervous system (CNS) and immune function. Several studies have recently examined the possible link between gut microbiota composition and multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Most of these studies agree that patients with MS suffer from dysbiosis. Moreover, an altered proportion of certain phyla of bacteria was detected in the digestive tracts of these patients compared to healthy individuals. This review article gathers information from research papers that have examined the relationship between gut microbiota composition and MS and its possible mechanisms.


Subject(s)
Brain-Gut Axis , Dysbiosis/complications , Encephalomyelitis, Autoimmune, Experimental/microbiology , Gastrointestinal Microbiome , Multiple Sclerosis/microbiology , Animals , Brain-Gut Axis/immunology , Brain-Gut Axis/physiology , Disease Models, Animal , Dysbiosis/physiopathology , Dysbiosis/therapy , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Fecal Microbiota Transplantation , Female , Humans , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Multiple Sclerosis/etiology , Multiple Sclerosis/physiopathology , Multiple Sclerosis/therapy , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/microbiology , Probiotics , Rats , Vitamin D/therapeutic use
11.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34502405

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory and degenerative disease of the central nervous system (CNS). MS commonly affects the cerebellum causing acute and chronic symptoms. Cerebellar signs significantly contribute to clinical disability, and symptoms such as tremor, ataxia, and dysarthria are difficult to treat. Fibroblast growth factors (FGFs) and their receptors (FGFRs) are involved in demyelinating pathologies such as MS. In autopsy tissue from patients with MS, increased expression of FGF1, FGF2, FGF9, and FGFR1 was found in lesion areas. Recent research using mouse models has focused on regions such as the spinal cord, and data on the expression of FGF/FGFR in the cerebellum are not available. In recent EAE studies, we detected that oligodendrocyte-specific deletion of FGFRs results in a milder disease course, less cellular infiltrates, and reduced neurodegeneration in the spinal cord. The objective of this study was to characterize the role of FGFR1 in oligodendrocytes in the cerebellum. Conditional deletion of FGFR1 in oligodendrocytes (Fgfr1ind-/-) was achieved by tamoxifen application, EAE was induced using the MOG35-55 peptide. The cerebellum was analyzed by histology, immunohistochemistry, and western blot. At day 62 p.i., Fgfr1ind-/- mice showed less myelin and axonal degeneration compared to FGFR1-competent mice. Infiltration of CD3(+) T cells, Mac3(+) cells, B220(+) B cells and IgG(+) plasma cells in cerebellar white matter lesions (WML) was less in Fgfr1ind-/-mice. There were no effects on the number of OPC or mature oligodendrocytes in white matter lesion (WML). Expression of FGF2 and FGF9 associated with less myelin and axonal degeneration, and of the pro-inflammatory cytokines IL-1ß, IL-6, and CD200 was downregulated in Fgfr1ind-/- mice. The FGF/FGFR signaling protein pAkt, BDNF, and TrkB were increased in Fgfr1ind-/- mice. These data suggest that cell-specific deletion of FGFR1 in oligodendrocytes has anti-inflammatory and neuroprotective effects in the cerebellum in the EAE disease model of MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Oligodendroglia/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Cerebellum/metabolism , Cerebellum/pathology , Demyelinating Diseases/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Fibroblast Growth Factors/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/metabolism , Multiple Sclerosis/physiopathology , Myelin Sheath/metabolism , Myelin-Oligodendrocyte Glycoprotein/pharmacology , Oligodendroglia/pathology , Phosphorylation , Receptor, Fibroblast Growth Factor, Type 1/physiology , Signal Transduction/drug effects , Spinal Cord/metabolism , White Matter/metabolism
12.
Neurobiol Dis ; 159: 105492, 2021 11.
Article in English | MEDLINE | ID: mdl-34478849

ABSTRACT

Repulsive guidance molecule A (RGMa) is a potent inhibitor of axonal growth and a regulator of neuronal cell death. It is up-regulated following neuronal injury and accumulates in chronic neurodegenerative diseases. Neutralizing RGMa has the potential to promote neuroregeneration and neuroprotection. Previously we reported that a rat anti-N terminal RGMa (N-RGMa) antibody r5F9 and its humanized version h5F9 (ABT-207) promote neuroprotection and neuroregeneration in preclinical neurodegenerative disease models. However, due to its cross-reactivity to RGMc/hemojuvelin, ABT-207 causes iron accumulation in vivo, which could present a safety liability. Here we report the generation and characterization of a novel RGMa-selective anti-N-RGMa antibody elezanumab, which is currently under Phase 2 clinical evaluation in multiple disease indications. Elezanumab, a human monoclonal antibody generated by in vitro PROfusion mRNA display technology, competes with ABT-207 in binding to N-RGMa but lacks RGMc cross-reactivity with no impact on iron metabolism. It neutralizes repulsive activity of soluble RGMa in vitro and blocks membrane RGMa mediated BMP signaling. In the optic nerve crush and optic neuritis models, elezanumab promotes axonal regeneration and prevents retinal nerve fiber layer degeneration. In the spinal targeted experimental autoimmune encephalomyelitis (EAE) model, elezanumab promotes axonal regeneration and remyelination, decreases inflammatory lesion area and improves functional recovery. Finally, in the mouse cuprizone model, elezanumab reduces demyelination, which is consistent with its inhibitory effect on BMP signaling. Taken together, these preclinical data demonstrate that elezanumab has neuroregenerative and neuroprotective activities without impact on iron metabolism, thus providing a compelling rationale for its clinical development in neurodegenerative diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , GPI-Linked Proteins , Nerve Regeneration , Nerve Tissue Proteins , Neuroprotection , Optic Nerve Injuries , Optic Nerve , Optic Neuritis , Recovery of Function , Retina , Animals , Mice , Cuprizone/toxicity , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/physiopathology , GPI-Linked Proteins/antagonists & inhibitors , Monoamine Oxidase Inhibitors/toxicity , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Neuroprotection/drug effects , Optic Nerve/drug effects , Optic Nerve/physiology , Optic Nerve Injuries/physiopathology , Optic Neuritis/physiopathology , Recovery of Function/drug effects , Recovery of Function/physiology , Retina/drug effects , Surface Plasmon Resonance
13.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Article in English | MEDLINE | ID: mdl-34479997

ABSTRACT

Neuroinflammation is a pathophysiological hallmark of multiple sclerosis and has a close mechanistic link to neurodegeneration. Although this link is potentially targetable, robust translatable models to reliably quantify and track neuroinflammation in both mice and humans are lacking. The choroid plexus (ChP) plays a pivotal role in regulating the trafficking of immune cells from the brain parenchyma into the cerebrospinal fluid (CSF) and has recently attracted attention as a key structure in the initiation of inflammatory brain responses. In a translational framework, we here address the integrity and multidimensional characteristics of the ChP under inflammatory conditions and question whether ChP volumes could act as an interspecies marker of neuroinflammation that closely interrelates with functional impairment. Therefore, we explore ChP characteristics in neuroinflammation in patients with multiple sclerosis and in two experimental mouse models, cuprizone diet-related demyelination and experimental autoimmune encephalomyelitis. We demonstrate that ChP enlargement-reconstructed from MRI-is highly associated with acute disease activity, both in the studied mouse models and in humans. A close dependency of ChP integrity and molecular signatures of neuroinflammation is shown in the performed transcriptomic analyses. Moreover, pharmacological modulation of the blood-CSF barrier with natalizumab prevents an increase of the ChP volume. ChP enlargement is strongly linked to emerging functional impairment as depicted in the mouse models and in multiple sclerosis patients. Our findings identify ChP characteristics as robust and translatable hallmarks of acute and ongoing neuroinflammatory activity in mice and humans that could serve as a promising interspecies marker for translational and reverse-translational approaches.


Subject(s)
Choroid Plexus/diagnostic imaging , Multiple Sclerosis/physiopathology , Neuroinflammatory Diseases/diagnostic imaging , Adult , Animals , Blood-Brain Barrier/physiology , Brain/physiology , Choroid Plexus/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Humans , Magnetic Resonance Imaging/methods , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/diagnostic imaging , Proteomics/methods
14.
Int J Mol Sci ; 22(15)2021 Jul 25.
Article in English | MEDLINE | ID: mdl-34360699

ABSTRACT

Reactive astrocytes are a hallmark of neurodegenerative disease including multiple sclerosis. It is widely accepted that astrocytes may adopt alternative phenotypes depending on a combination of environmental cues and intrinsic features in a highly plastic and heterogeneous manner. However, we still lack a full understanding of signals and associated signaling pathways driving astrocyte reaction and of the mechanisms by which they drive disease. We have previously shown in the experimental autoimmune encephalomyelitis mouse model that deficiency of the molecular adaptor Rai reduces disease severity and demyelination. Moreover, using primary mouse astrocytes, we showed that Rai contributes to the generation of a pro-inflammatory central nervous system (CNS) microenvironment through the production of nitric oxide and IL-6 and by impairing CD39 activity in response to soluble factors released by encephalitogenic T cells. Here, we investigated the impact of Rai expression on astrocyte function both under basal conditions and in response to IL-17 treatment using a proteomic approach. We found that astrocytes and astrocyte-derived extracellular vesicles contain a set of proteins, to which Rai contributes, that are involved in the regulation of oligodendrocyte differentiation and myelination, nitrogen metabolism, and oxidative stress. The HIF-1α pathway and cellular energetic metabolism were the most statistically relevant molecular pathways and were related to ENOA and HSP70 dysregulation.


Subject(s)
Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Extracellular Vesicles/metabolism , Interleukin-17/pharmacology , Neuroprotection , Oligodendroglia/physiology , Src Homology 2 Domain-Containing, Transforming Protein 3/genetics , Animals , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath , Proteomics , Src Homology 2 Domain-Containing, Transforming Protein 3/metabolism
15.
Theranostics ; 11(17): 8480-8499, 2021.
Article in English | MEDLINE | ID: mdl-34373754

ABSTRACT

Rationale: Optic neuritis is one of main symptoms in multiple sclerosis (MS) that causes visual disability. Astrocytes are pivotal regulators of neuroinflammation in MS, and astrocytic yes-associated protein (YAP) plays a critical role in neuroinflammation. Meanwhile, YAP signaling is involved in visual impairment, including glaucoma, retinal choroidal atrophy and retinal detachment. However, the roles and underlying mechanisms of astrocytic YAP in neuroinflammation and demyelination of MS-related optic neuritis (MS-ON) remains unclear. Methods: To assess the functions of YAP in MS-ON, experimental autoimmune encephalomyelitis (EAE, a common model of MS) was established, and mice that conditional knockout (CKO) of YAP in astrocytes, YAPGFAP-CKO mice, were successfully generated. Behavior tests, immunostaining, Nissl staining, Hematoxylin-Eosin (HE) staining, TUNEL staining, Luxol Fast Blue (LFB) staining, electron microscopy (EM), quantitative real-time PCR (qPCR), gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) by RNA sequencing were used to examine the function and mechanism of YAP signaling based on these YAPGFAP-CKO mice and EAE model mice. To further explore the potential treatment of YAP signaling in EAE, EAE mice were treated with various drugs, including SRI-011381 that is an agonist of transforming growth factor-ß (TGF-ß) pathway, and XMU-MP-1 which inhibits Hippo kinase MST1/2 to activate YAP. Results: We found that YAP was significantly upregulated and activated in the astrocytes of optic nerve in EAE mice. Conditional knockout of YAP in astrocytes caused more severe inflammatory infiltration and demyelination in optic nerve, and damage of retinal ganglion cells (RGCs) in EAE mice. Moreover, YAP deletion in astrocytes promoted the activation of astrocytes and microglia, but inhibited the proliferation of astrocytes of optic nerve in EAE mice. Mechanically, TGF-ß signaling pathway was significantly down-regulated after YAP deletion in astrocytes. Additionally, both qPCR and immunofluorescence assays confirmed the reduction of TGF-ß signaling pathway in YAPGFAP-CKO EAE mice. Interestingly, SRI-011381 partially rescued the deficits in optic nerve and retina of YAPGFAP-CKO EAE mice. Finally, activation of YAP signaling by XMU-MP-1 relieved the neuroinflammation and demyelination in optic nerve of EAE mice. Conclusions: These results suggest astrocytic YAP may prevent the neuroinflammatory infiltration and demyelination through upregulation of TGF-ß signaling and provide targets for the development of therapeutic strategies tailored for MS-ON.


Subject(s)
Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , YAP-Signaling Proteins/metabolism , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , Neuroinflammatory Diseases , Optic Nerve/physiology , Optic Neuritis/metabolism , Optic Neuritis/physiopathology , Retina/metabolism , Retina/physiology , Retinal Ganglion Cells/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology , YAP-Signaling Proteins/physiology
16.
Front Immunol ; 12: 691590, 2021.
Article in English | MEDLINE | ID: mdl-34349758

ABSTRACT

Multiple sclerosis (MS) is a chronic disease in the central nervous system (CNS), characterized by inflammatory cells that invade into the brain and the spinal cord. Among a bulk of different MS models, the most widely used and best understood rodent model is experimental autoimmune encephalomyelitis (EAE). Arctigenin, a botanical extract from Arctium lappa, is reported to exhibit pharmacological properties, including anti-inflammation and neuroprotection. However, the effects of arctigenin on neural activity attacked by inflammation in MS are still unclear. Here, we use two-photon calcium imaging to observe the activity of somatosensory cortex neurons in awake EAE mice in vivo and found added hyperactive cells, calcium influx, network connectivity, and synchronization, mainly at preclinical stage of EAE model. Besides, more silent cells and decreased calcium influx and reduced network synchronization accompanied by a compensatory rise in functional connectivity are found at the remission stage. Arctigenin treatment not only restricts inordinate individually neural spiking, calcium influx, and network activity at preclinical stage but also restores neuronal activity and communication at remission stage. In addition, we confirm that the frequency of AMPA receptor-mediated spontaneous excitatory postsynaptic current (sEPSC) is also increased at preclinical stage and can be blunted by arctigenin. These findings suggest that excitotoxicity characterized by calcium influx is involved in EAE at preclinical stage. What is more, arctigenin exerts neuroprotective effect by limiting hyperactivity at preclinical stage and ameliorates EAE symptoms, indicating that arctigenin could be a potential therapeutic drug for neuroprotection in MS-related neuropsychological disorders.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Furans/therapeutic use , Lignans/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Calcium/physiology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Excitatory Postsynaptic Potentials/drug effects , Female , Furans/pharmacology , Lignans/pharmacology , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/pharmacology , Somatosensory Cortex/drug effects , Somatosensory Cortex/physiology
17.
Int J Mol Sci ; 22(16)2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34445295

ABSTRACT

Skeletal muscle is affected in experimental autoimmune encephalomyelitis (EAE), which is a model of multiple sclerosis that produces changes including muscle atrophy; histological features of neurogenic involvement, and increased oxidative stress. In this study, we aimed to evaluate the therapeutic effects of transcranial magnetic stimulation (TMS) on the involvement of rat skeletal muscle and to compare them with those produced by natalizumab (NTZ). EAE was induced by injecting myelin oligodendrocyte glycoprotein (MOG) into Dark Agouti rats. Both treatments, NTZ and TMS, were implemented from day 15 to day 35. Clinical severity was studied, and after sacrifice, the soleus and extensor digitorum longus muscles were extracted for subsequent histological and biochemical analysis. The treatment with TMS and NTZ had a beneficial effect on muscle involvement in the EAE model. There was a clinical improvement in functional motor deficits, atrophy was attenuated, neurogenic muscle lesions were reduced, and the level of oxidative stress biomarkers was lower in both treatment groups. Compared to NTZ, the best response was obtained with TMS for all the parameters analyzed. The myoprotective effect of TMS was higher than that of NTZ. Thus, the use of TMS may be an effective strategy to reduce muscle involvement in multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Muscular Atrophy/prevention & control , Transcranial Magnetic Stimulation , Animals , Cell Count , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Male , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/physiopathology , Myelin-Oligodendrocyte Glycoprotein , Natalizumab/pharmacology , Rats
18.
Int J Mol Sci ; 22(10)2021 May 18.
Article in English | MEDLINE | ID: mdl-34070011

ABSTRACT

Dopamine is a neurotransmitter that mediates neuropsychological functions of the central nervous system (CNS). Recent studies have shown the modulatory effect of dopamine on the cells of innate and adaptive immune systems, including Th17 cells, which play a critical role in inflammatory diseases of the CNS. This article reviews the literature data on the role of dopamine in the regulation of neuroinflammation in multiple sclerosis (MS). The influence of dopaminergic receptor targeting on experimental autoimmune encephalomyelitis (EAE) and MS pathogenesis, as well as the therapeutic potential of dopaminergic drugs as add-on pathogenetic therapy of MS, is discussed.


Subject(s)
Dopamine/immunology , Multiple Sclerosis/drug therapy , Receptors, Dopamine/drug effects , Animals , Dopamine/physiology , Dopamine Agents/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Humans , Mice , Models, Immunological , Models, Neurological , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/immunology , Neuroimmunomodulation/physiology , Receptors, Dopamine/immunology , Receptors, Dopamine/physiology , Th17 Cells/drug effects , Th17 Cells/immunology
19.
Front Immunol ; 12: 666961, 2021.
Article in English | MEDLINE | ID: mdl-33936108

ABSTRACT

In multiple sclerosis (MS) and other neuroinflammatory diseases, monocyte-derived cells (MoCs) traffic through distinct central nervous system (CNS) barriers and gain access to the organ parenchyma exerting detrimental or beneficial functions. How and where these MoCs acquire their different functional commitments during CNS invasion remains however unclear, thus hindering the design of MS treatments specifically blocking detrimental MoC actions. To clarify this issue, we investigated the distribution of iNOS+ pro-inflammatory and arginase-1+ anti-inflammatory MoCs at the distinct border regions of the CNS in a mouse model of MS. Interestingly, MoCs within perivascular parenchymal spaces displayed a predominant pro-inflammatory phenotype compared to MoCs accumulating at the leptomeninges and at the intraventricular choroid plexus (ChP). Furthermore, in an in vitro model, we could observe the general ability of functionally-polarized MoCs to migrate through the ChP epithelial barrier, together indicating the ChP as a potential CNS entry and polarization site for MoCs. Thus, pro- and anti-inflammatory MoCs differentially accumulate at distinct CNS barriers before reaching the parenchyma, but the mechanism for their phenotype acquisition remains undefined. Shedding light on this process, we observed that endothelial (BBB) and epithelial (ChP) CNS barrier cells can directly regulate transcription of Nos2 (coding for iNOS) and Arg1 (coding for arginase-1) in interacting MoCs. More specifically, while TNF-α+IFN-γ stimulated BBB cells induced Nos2 expression in MoCs, IL-1ß driven activation of endothelial BBB cells led to a significant upregulation of Arg1 in MoCs. Supporting this latter finding, less pro-inflammatory MoCs could be found nearby IL1R1+ vessels in the mouse spinal cord upon neuroinflammation. Taken together, our data indicate differential distribution of pro- and anti-inflammatory MoCs at CNS borders and highlight how the interaction of MoCs with CNS barriers can significantly affect the functional activation of these CNS-invading MoCs during autoimmune inflammation.


Subject(s)
Arginase/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Macrophages/metabolism , Nitric Oxide Synthase Type II/metabolism , Animals , Blood-Brain Barrier/metabolism , Central Nervous System/metabolism , Central Nervous System/physiology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Endothelial Cells/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , Multiple Sclerosis/physiopathology
20.
Neuron ; 109(10): 1754-1754.e1, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34015268

ABSTRACT

Neuronal function relies on tightly controlled cytoskeleton transport with adaptive cargo trafficking as prerequisite for synaptic transmission. During inflammation in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), axonal transport efficiency declines, followed by neurodegeneration. Furthermore, neuroinflammation causes an imbalance between excitatory and inhibitory transmission, triggering synaptic dysfunction and loss. Recent data suggest that neuronal transport and synaptic deficits during neuroinflammation are functionally interconnected. To view this SnapShot, open or download the PDF.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/physiopathology , Multiple Sclerosis/physiopathology , Neurons/physiology , Animals , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Multiple Sclerosis/immunology , Neurons/immunology , Synaptic Transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...