Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 291
Filter
1.
Immunol Med ; 47(2): 52-57, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38597289

ABSTRACT

Mixed connective tissue disease (MCTD) is an autoimmune disorder characterized by a combination of clinical features from systemic lupus erythematosus, systemic sclerosis, and inflammatory muscle disease, along with the presence of positive anti-U1-ribonucleoprotein (U1-RNP) antibodies. The exact etiology of the disease remains unclear, but it is believed to involve vascular damage within the context of heightened autoimmune responses. Consequently, Raynaud's phenomenon and pulmonary arterial hypertension are observed in patients with MCTD. While specific biomarkers for MCTD have not yet been identified, the recent study of the utility of anti-survival motor neuron complex (SMN) antibodies in MCTD suggests a promising avenue for further research and the accumulation of additional evidence.


Subject(s)
Autoantibodies , Biomarkers , Mixed Connective Tissue Disease , Humans , Mixed Connective Tissue Disease/immunology , Mixed Connective Tissue Disease/diagnosis , Biomarkers/blood , Autoantibodies/blood , Severity of Illness Index , Motor Neurons/immunology , Ribonucleoprotein, U1 Small Nuclear/immunology
2.
Int J Mol Sci ; 22(15)2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34360808

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a progressive disease leading to the degeneration of motor neurons (MNs). Neuroinflammation is involved in the pathogenesis of ALS; however, interactions of specific immune cell types and MNs are not well studied. We recently found a shift toward T helper (Th)1/Th17 cell-mediated, pro-inflammatory immune responses in the peripheral immune system of ALS patients, which positively correlated with disease severity and progression. Whether Th17 cells or their central mediator, Interleukin-17 (IL-17), directly affects human motor neuron survival is currently unknown. Here, we evaluated the contribution of Th17 cells and IL-17 on MN degeneration using the co-culture of iPSC-derived MNs of fused in sarcoma (FUS)-ALS patients and isogenic controls with Th17 lymphocytes derived from ALS patients, healthy controls, and multiple sclerosis (MS) patients (positive control). Only Th17 cells from MS patients induced severe MN degeneration in FUS-ALS as well as in wildtype MNs. Their main effector, IL-17A, yielded in a dose-dependent decline of the viability and neurite length of MNs. Surprisingly, IL-17F did not influence MNs. Importantly, neutralizing IL-17A and anti-IL-17 receptor A treatment reverted all effects of IL-17A. Our results offer compelling evidence that Th17 cells and IL-17A do directly contribute to MN degeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Induced Pluripotent Stem Cells/immunology , Interleukin-17/immunology , Motor Neurons/immunology , RNA-Binding Protein FUS/immunology , Th17 Cells/immunology , Amyotrophic Lateral Sclerosis/pathology , Cell Survival/immunology , Humans , Induced Pluripotent Stem Cells/pathology , Motor Neurons/pathology , Th17 Cells/pathology
3.
Cell Death Dis ; 12(7): 625, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34135312

ABSTRACT

Motoneuronal loss is the main feature of amyotrophic lateral sclerosis, although pathogenesis is extremely complex involving both neural and muscle cells. In order to translationally engage the sonic hedgehog pathway, which is a promising target for neural regeneration, recent studies have reported on the neuroprotective effects of clobetasol, an FDA-approved glucocorticoid, able to activate this pathway via smoothened. Herein we sought to examine functional, cellular, and metabolic effects of clobetasol in a neurotoxic mouse model of spinal motoneuronal loss. We found that clobetasol reduces muscle denervation and motor impairments in part by restoring sonic hedgehog signaling and supporting spinal plasticity. These effects were coupled with reduced pro-inflammatory microglia and reactive astrogliosis, reduced muscle atrophy, and support of mitochondrial integrity and metabolism. Our results suggest that clobetasol stimulates a series of compensatory processes and therefore represents a translational approach for intractable denervating and neurodegenerative disorders.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Clobetasol/pharmacology , Glucocorticoids/pharmacology , Hedgehog Proteins/metabolism , Motor Activity/drug effects , Motor Neurons/drug effects , Muscle, Skeletal/innervation , Neuronal Plasticity/drug effects , Neuroprotective Agents/pharmacology , Spine/drug effects , Amyotrophic Lateral Sclerosis/chemically induced , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/metabolism , Animals , Case-Control Studies , Cholera Toxin , Databases, Genetic , Disease Models, Animal , Energy Metabolism/drug effects , Humans , Inflammation Mediators/metabolism , Male , Mice, 129 Strain , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Motor Neurons/immunology , Motor Neurons/metabolism , Open Field Test , Saporins , Signal Transduction , Smoothened Receptor/agonists , Smoothened Receptor/metabolism , Spine/immunology , Spine/metabolism , Spine/physiopathology
4.
Nat Rev Neurol ; 17(6): 333-348, 2021 06.
Article in English | MEDLINE | ID: mdl-33927394

ABSTRACT

The prevailing motor neuron-centric view of amyotrophic lateral sclerosis (ALS) pathogenesis could be an important factor in the failure to identify disease-modifying therapy for this neurodegenerative disorder. Non-neuronal cells have crucial homeostatic functions within the CNS and evidence of involvement of these cells in the pathophysiology of several neurodegenerative disorders, including ALS, is accumulating. Microglia and astrocytes, in crosstalk with peripheral immune cells, can exert both neuroprotective and adverse effects, resulting in a highly nuanced range of neuronal and non-neuronal cell interactions. This Review provides an overview of the diverse roles of non-neuronal cells in relation to the pathogenesis of ALS and the emerging potential of non-neuronal cell biomarkers to advance therapeutic development.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/immunology , Astrocytes/metabolism , Microglia/immunology , Microglia/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Biomarkers/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Humans , Motor Neurons/immunology , Motor Neurons/pathology
5.
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
6.
Nat Neurosci ; 23(11): 1339-1351, 2020 11.
Article in English | MEDLINE | ID: mdl-33077946

ABSTRACT

Microglia and peripheral macrophages have both been implicated in amyotrophic lateral sclerosis (ALS), although their respective roles have yet to be determined. We now show that macrophages along peripheral motor neuron axons in mouse models and patients with ALS react to neurodegeneration. In ALS mice, peripheral myeloid cell infiltration into the spinal cord was limited and depended on disease duration. Targeted gene modulation of the reactive oxygen species pathway in peripheral myeloid cells of ALS mice, using cell replacement, reduced both peripheral macrophage and microglial activation, delayed symptoms and increased survival. Transcriptomics revealed that sciatic nerve macrophages and microglia reacted differently to neurodegeneration, with abrupt temporal changes in macrophages and progressive, unidirectional activation in microglia. Modifying peripheral macrophages suppressed proinflammatory microglial responses, with a shift toward neuronal support. Thus, modifying macrophages at the periphery has the capacity to influence disease progression and may be of therapeutic value for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Axons/immunology , Macrophages/immunology , Microglia/immunology , Motor Neurons/immunology , Sciatic Nerve/immunology , Adult , Aged , Amyotrophic Lateral Sclerosis/metabolism , Animals , Female , Humans , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Middle Aged , Motor Neurons/metabolism , Sciatic Nerve/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism
7.
FASEB J ; 34(6): 7225-7233, 2020 06.
Article in English | MEDLINE | ID: mdl-32307753

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressing motor neuron disease without effective treatment. Although the precise mechanisms leading to ALS are yet to be determined, there is now increasing evidence implicating the defective energy metabolism and components of the innate immune complement system in the onset and progression of its motor phenotypes. This review will survey the mechanisms by which the energy metabolism and the complement system are altered during the disease progression of ALS and how it can contribute to disease. Furthermore, it will also examine how complement activation can modify the energy metabolism in metabolic disorders, in order to highlight how the complement system and energy metabolism may be linked in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Complement Activation/immunology , Complement System Proteins/immunology , Energy Metabolism/immunology , Immunity, Innate/immunology , Animals , Disease Progression , Humans , Motor Neurons/immunology
8.
Pharmacol Res ; 156: 104792, 2020 06.
Article in English | MEDLINE | ID: mdl-32278047

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating and rapidly progressing neurodegenerative disorder with no effective disease-modifying treatment up to date. The underlying molecular mechanisms of ALS are not yet completely understood. However, the critical role of the innate immune system and neuroinflammation in ALS pathogenesis has gained increased attention. High mobility group box 1 (HMGB1) is a typical damage-associated molecular pattern (DAMP) molecule, acting as a pro-inflammatory cytokine mainly through activation of its principal receptors, the receptor for advanced glycation end products (RAGE) and toll-like receptor 4 (TLR4) which are crucial components of the innate immune system. HMGB1 is an endogenous ligand for both RAGE and TLR4 that mediate its biological effects. Herein, on the ground of pre-clinical findings we unravel the underlying mechanisms behind the plausible contribution of HMGB1 and its receptors (RAGE and TLR4) in the ALS pathogenesis. Furthermore, we provide an account of the therapeutic outcomes associated with inhibition/blocking of HMGB1 receptor signalling in preventing motor neuron's death and delaying disease progression in ALS experimental models. There is strong evidence that HMGB1, RAGE and TLR4 signaling axes might present potential targets against ALS, opening a novel headway in ALS research that could plausibly bridge the current treatment gap.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Brain/metabolism , HMGB1 Protein/metabolism , Motor Neurons/metabolism , Receptor for Advanced Glycation End Products/metabolism , Spinal Cord/metabolism , Toll-Like Receptor 4/metabolism , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/immunology , Brain/pathology , Humans , Immunity, Innate , Ligands , Motor Neurons/immunology , Motor Neurons/pathology , Signal Transduction , Spinal Cord/immunology , Spinal Cord/pathology
9.
Nat Commun ; 11(1): 1773, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32286313

ABSTRACT

In amyotrophic lateral sclerosis (ALS), immune cells and glia contribute to motor neuron (MN) degeneration. We report the presence of NK cells in post-mortem ALS motor cortex and spinal cord tissues, and the expression of NKG2D ligands on MNs. Using a mouse model of familial-ALS, hSOD1G93A, we demonstrate NK cell accumulation in the motor cortex and spinal cord, with an early CCL2-dependent peak. NK cell depletion reduces the pace of MN degeneration, delays motor impairment and increases survival. This is confirmed in another ALS mouse model, TDP43A315T. NK cells are neurotoxic to hSOD1G93A MNs which express NKG2D ligands, while IFNγ produced by NK cells instructs microglia toward an inflammatory phenotype, and impairs FOXP3+/Treg cell infiltration in the spinal cord of hSOD1G93A mice. Together, these data suggest a role of NK cells in determining the onset and progression of MN degeneration in ALS, and in modulating Treg recruitment and microglia phenotype.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Killer Cells, Natural/metabolism , Motor Neurons/metabolism , Adult , Aged , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Disease Progression , Female , Humans , Killer Cells, Natural/immunology , Male , Mice , Middle Aged , Motor Cortex/immunology , Motor Cortex/metabolism , Motor Cortex/pathology , Motor Neurons/immunology , Motor Neurons/pathology , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology
10.
J Peripher Nerv Syst ; 25(2): 143-151, 2020 06.
Article in English | MEDLINE | ID: mdl-32250537

ABSTRACT

In mouse models of acute motor axonal neuropathy, anti-ganglioside antibodies (AGAbs) bind to motor axons, notably the distal nerve, and activate the complement cascade. While complement activation is well studied in this model, the role of inflammatory cells is unknown. Herein we aimed to investigate the contribution of phagocytic cells including macrophages, neutrophils and perisynaptic Schwann cells (pSCs) to distal nerve pathology. To observe this, we first created a subacute injury model of sufficient duration to allow inflammatory cell recruitment. Mice were injected intraperitoneally with an anti-GD1b monoclonal antibody that binds strongly to mouse motor nerve axons. Subsequently, mice received normal human serum as a source of complement. Dosing was titrated to allow humane survival of mice over a period of 3 days, yet still induce the characteristic neurological impairment. Behaviour and pathology were assessed in vivo using whole-body plethysmography and post-sacrifice by immunofluorescence and flow cytometry. ex vivo nerve-muscle preparations were used to investigate the acute phagocytic role of pSCs following distal nerve injury. Following complement activation at distal intramuscular nerve sites in the diaphragm macrophage localisation or numbers are not altered, nor do they shift to a pro- or anti-inflammatory phenotype. Similarly, neutrophils are not significantly recruited. Instead, ex vivo nerve-muscle preparations exposed to AGAb plus complement reveal that pSCs rapidly become phagocytic and engulf axonal debris. These data suggest that pSCs, rather than inflammatory cells, are the major cellular vehicle for axonal debris clearance following distal nerve injury, in contrast to larger nerve bundles where macrophage-mediated clearance predominates.


Subject(s)
Antibodies, Monoclonal/pharmacology , Gangliosides/immunology , Guillain-Barre Syndrome , Motor Neurons , Neuromuscular Junction , Phagocytosis/physiology , Presynaptic Terminals , Schwann Cells/physiology , Animals , Antibodies, Monoclonal/administration & dosage , Behavior, Animal/physiology , Complement Activation/immunology , Disease Models, Animal , Female , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/pathology , Humans , Male , Mice , Mice, Transgenic , Motor Neurons/immunology , Motor Neurons/pathology , Neuromuscular Junction/immunology , Neuromuscular Junction/pathology , Presynaptic Terminals/immunology , Presynaptic Terminals/pathology
11.
J Cell Physiol ; 235(6): 5030-5040, 2020 06.
Article in English | MEDLINE | ID: mdl-31788795

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by neuronal degeneration and inflammation in the nerves. The role of the immune system has been concentrated by researchers in the etiopathogenesis of the disease. Given the inhibitory roles of regulatory T cells (Tregs), it is expected that increasing or activating their populations in patients with ALS can have significant therapeutic effects. Here we searched databases, including CENTRAL, MEDLINE, CINAHL Plus, clinicaltrials.gov, and ICTRP for randomized clinical trials (RCTs) and non-RCTs until March 2019. For preclinical studies, we searched PubMed, Scopus, and Google Scholar up to June 2019. We also included preclinical studies, due to the lack of clinical information available, which used Tregs (or directly targeting them) for treating mice models of ALS. We identified 29 records (CENTRAL 7, MEDLINE 4, CINAHL Plus 8, and clinicaltrials.gov 10) and removed 10 duplicated publications. After screening, we identified one RCT which had been published as an abstract, three non-RCTs, and four ongoing studies. We also identified 551 records (PubMed 446, Google Scholar 68, and Scopus 37) for preclinical studies and performed a meta-analysis. Finally, we found three papers that matched our inclusion criteria for preclinical studies. Results indicated the effectiveness of the application of Tregs in the treatment of ALS. Our meta-analysis on preclinical studies revealed that Tregs significantly prolonged survival in mice models of ALS. Overall, our analysis testified that exertion of Tregs in the treatment of ALS is a promising approach, that notwithstanding, requires further evaluations.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Inflammation/immunology , Motor Neuron Disease/immunology , T-Lymphocytes, Regulatory/immunology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Humans , Inflammation/pathology , Motor Neuron Disease/genetics , Motor Neuron Disease/pathology , Motor Neurons/immunology , Motor Neurons/metabolism , Motor Neurons/pathology , Quality of Life , T-Lymphocytes, Regulatory/pathology
12.
Cell Rep ; 29(13): 4496-4508.e4, 2019 Dec 24.
Article in English | MEDLINE | ID: mdl-31875556

ABSTRACT

Mutations in the FUS gene cause familial amyotrophic lateral sclerosis (ALS-FUS). In ALS-FUS, FUS-positive inclusions are detected in the cytoplasm of neurons and glia, a condition known as FUS proteinopathy. Mutant FUS incorporates into stress granules (SGs) and can spontaneously form cytoplasmic RNA granules in cultured cells. However, it is unclear what can trigger the persistence of mutant FUS assemblies and lead to inclusion formation. Using CRISPR/Cas9 cell lines and patient fibroblasts, we find that the viral mimic dsRNA poly(I:C) or a SG-inducing virus causes the sustained presence of mutant FUS assemblies. These assemblies sequester the autophagy receptor optineurin and nucleocytoplasmic transport factors. Furthermore, an integral component of the antiviral immune response, type I interferon, promotes FUS protein accumulation by increasing FUS mRNA stability. Finally, mutant FUS-expressing cells are hypersensitive to dsRNA toxicity. Our data suggest that the antiviral immune response is a plausible second hit for FUS proteinopathy.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Host-Pathogen Interactions/immunology , Motor Neurons/immunology , RNA-Binding Protein FUS/immunology , Respiratory Syncytial Viruses/immunology , Spinal Cord/immunology , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/immunology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/virology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Cell Line , Cytoplasmic Granules/genetics , Cytoplasmic Granules/immunology , Cytoplasmic Granules/virology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/immunology , Gene Expression Regulation , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Inclusion Bodies/genetics , Inclusion Bodies/immunology , Inclusion Bodies/virology , Interferon Type I/genetics , Interferon Type I/immunology , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/immunology , Motor Neurons/metabolism , Motor Neurons/virology , Neuroglia/immunology , Neuroglia/metabolism , Neuroglia/virology , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/immunology , Poly I-C/pharmacology , Primary Cell Culture , Protein Aggregates/genetics , Protein Aggregates/immunology , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA-Binding Protein FUS/genetics , Respiratory Syncytial Viruses/pathogenicity , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/virology
14.
JCI Insight ; 52019 07 16.
Article in English | MEDLINE | ID: mdl-31310593

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease with diverse etiologies. Therefore, the identification of common disease mechanisms and therapeutics targeting these mechanisms could dramatically improve clinical outcomes. To this end, we developed induced motor neuron (iMN) models from C9ORF72 and sporadic ALS (sALS) patients to identify targets that are effective against these types of cases, which together comprise ~90% of patients. We find that iMNs from C9ORF72 and several sporadic ALS patients share two common defects - impaired autophagosome formation and the aberrant accumulation of glutamate receptors. Moreover, we show that an anticoagulation-deficient form of activated protein C, 3K3A-APC, rescues these defects in both C9ORF72 and sporadic ALS iMNs. As a result, 3K3A-APC treatment lowers C9ORF72 dipeptide repeat protein (DPR) levels, restores nuclear TDP-43 localization, and rescues the survival of both C9ORF72 and sporadic ALS iMNs. Importantly, 3K3A-APC also lowers glutamate receptor levels and rescues proteostasis in vivo in C9ORF72 gain- and loss-of-function mouse models. Thus, motor neurons from C9ORF72 and at least a subset of sporadic ALS patients share common, early defects in autophagosome formation and glutamate receptor homeostasis and a single therapeutic approach may be efficacious against these disease processes.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Autophagosomes/drug effects , Motor Neurons/drug effects , Protein C/administration & dosage , Adult , Aged , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Animals , Autophagosomes/immunology , Autophagy/genetics , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , CHO Cells , Cells, Cultured , Cricetulus , Disease Models, Animal , Female , Gain of Function Mutation , Humans , Induced Pluripotent Stem Cells , Loss of Function Mutation , Lymphocytes , Male , Mice , Middle Aged , Motor Neurons/immunology , Motor Neurons/pathology , Primary Cell Culture , Protein C/genetics , Proteostasis/drug effects , Proteostasis/immunology , Receptor, PAR-1/agonists , Receptor, PAR-1/metabolism , Receptors, Glutamate/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics
15.
J Clin Invest ; 129(6): 2222-2236, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30869655

ABSTRACT

Neurofascin-155 (Nfasc155) is an essential glial cell adhesion molecule expressed in paranodal septate-like junctions of peripheral and central myelinated axons. The genetic deletion of Nfasc155 results in the loss of septate-like junctions and in conduction slowing. In humans, IgG4 antibodies against Nfasc155 are implicated in the pathogenesis of chronic inflammatory demyelinating polyneuropathy (CIDP). These antibodies are associated with an aggressive onset, a refractoriness to intravenous immunoglobulin, and tremor of possible cerebellar origin. Here, we examined the pathogenic effects of patient-derived anti-Nfasc155 IgG4. These antibodies did not inhibit the ability of Nfasc155 to complex with its axonal partners contactin-1/CASPR1 or induce target internalization. Passive transfer experiments revealed that IgG4 antibodies target Nfasc155 on Schwann cell surface, and diminished Nfasc155 protein levels and prevented paranodal complex formation in neonatal animals. In adult animals, chronic intrathecal infusions of antibodies also induced the loss of Nfasc155 and of paranodal specialization and resulted in conduction alterations in motor nerves. These results indicate that anti-Nfasc155 IgG4 perturb conduction in absence of demyelination, validating the existence of paranodopathy. These results also shed light on the mechanisms regulating protein insertion at paranodes.


Subject(s)
Axons/immunology , Cell Adhesion Molecules/antagonists & inhibitors , Immunoglobulin G/pharmacology , Nerve Growth Factors/antagonists & inhibitors , Polyneuropathies , Polyradiculoneuropathy , Animals , Axons/pathology , Cell Adhesion Molecules/immunology , Chronic Disease , Female , HEK293 Cells , Humans , Immunoglobulin G/immunology , Male , Motor Neurons/immunology , Motor Neurons/pathology , Nerve Growth Factors/immunology , Polyneuropathies/drug therapy , Polyneuropathies/immunology , Polyneuropathies/pathology , Polyradiculoneuropathy/drug therapy , Polyradiculoneuropathy/immunology , Polyradiculoneuropathy/pathology , Rats , Rats, Inbred Lew , Schwann Cells/immunology , Schwann Cells/pathology
16.
Proc Natl Acad Sci U S A ; 116(6): 2312-2317, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30674678

ABSTRACT

Adaptive immune response is part of the dynamic changes that accompany motoneuron loss in amyotrophic lateral sclerosis (ALS). CD4+ T cells that regulate a protective immunity during the neurodegenerative process have received the most attention. CD8+ T cells are also observed in the spinal cord of patients and ALS mice although their contribution to the disease still remains elusive. Here, we found that activated CD8+ T lymphocytes infiltrate the central nervous system (CNS) of a mouse model of ALS at the symptomatic stage. Selective ablation of CD8+ T cells in mice expressing the ALS-associated superoxide dismutase-1 (SOD1)G93A mutant decreased spinal motoneuron loss. Using motoneuron-CD8+ T cell coculture systems, we found that mutant SOD1-expressing CD8+ T lymphocytes selectively kill motoneurons. This cytotoxicity activity requires the recognition of the peptide-MHC-I complex (where MHC-I represents major histocompatibility complex class I). Measurement of interaction strength by atomic force microscopy-based single-cell force spectroscopy demonstrated a specific MHC-I-dependent interaction between motoneuron and SOD1G93A CD8+ T cells. Activated mutant SOD1 CD8+ T cells produce interferon-γ, which elicits the expression of the MHC-I complex in motoneurons and exerts their cytotoxic function through Fas and granzyme pathways. In addition, analysis of the clonal diversity of CD8+ T cells in the periphery and CNS of ALS mice identified an antigen-restricted repertoire of their T cell receptor in the CNS. Our results suggest that self-directed immune response takes place during the course of the disease, contributing to the selective elimination of a subset of motoneurons in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Gene Expression , Motor Neurons/metabolism , Mutation , Superoxide Dismutase-1/genetics , T-Lymphocytes, Cytotoxic/metabolism , Amyotrophic Lateral Sclerosis/diagnosis , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Cell Communication/immunology , Cell Death , Cell Survival/genetics , Disease Models, Animal , Granzymes/metabolism , Histocompatibility Antigens Class I/immunology , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Motor Neurons/immunology , Phenotype , Severity of Illness Index , Spinal Cord/cytology , T-Lymphocytes, Cytotoxic/immunology , fas Receptor/metabolism
17.
J Neurol ; 266(1): 27-36, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29845377

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is the most common neurodegenerative disease affecting motor neurons (MN). This fatal disease is characterized by progressive muscle wasting and lacks an effective treatment. ALS pathogenesis has not been elucidated yet. In a small proportion of ALS patients, the disease has a familial origin, related to mutations in specific genes, which directly result in MN degeneration. By contrast, the vast majority of cases are though to be sporadic, in which genes and environment interact leading to disease in genetically predisposed individuals. Lately, the role of the environment has gained relevance in this field and an extensive list of environmental conditions have been postulated to be involved in ALS. Among them, infectious agents, particularly viruses, have been suggested to play an important role in the pathogenesis of the disease. These agents could act by interacting with some crucial pathways in MN degeneration, such as gene processing, oxidative stress or neuroinflammation. In this article, we will review the main studies about the involvement of microorganisms in ALS, subsequently discussing their potential pathogenic effect and integrating them as another piece in the puzzle of ALS pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/microbiology , Bacterial Infections/immunology , Virus Diseases/immunology , Amyotrophic Lateral Sclerosis/virology , Animals , Humans , Motor Neurons/immunology , Motor Neurons/microbiology , Motor Neurons/parasitology , Motor Neurons/virology
18.
Neurobiol Dis ; 121: 131-137, 2019 01.
Article in English | MEDLINE | ID: mdl-30176351

ABSTRACT

Mutations in Cu/Zn superoxide dismutase (SOD1) are the cause of ~20% of cases of familial ALS (FALS), which comprise ~10% of the overall total number of cases of ALS. Mutant (mt) SOD1 is thought to cause FALS through a gain and not loss in function, perhaps as a result of the mutant protein's misfolding and aggregation. Previously we used a phage display library to raise single chain variable fragment antibodies (scFvs) against SOD1, which were found to decrease aggregation of mtSOD1 and toxicity in vitro. In the present study, we show that two scFvs directed against SOD1 ameliorate disease in G93A mtSOD1 transgenic mice and also decrease motor neuron loss, microgliosis, astrocytosis, as well as SOD1 burden and aggregation. The results suggest that the use of antibodies or antibody mimetics directed against SOD1 may be a useful therapeutic direction in mtSOD1-induced FALS. Since studies suggest that wild type SOD1 may be misfolded similar to that seen with mtSOD1, this therapeutic direction may be effective in sporadic as well as FALS.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Single-Chain Antibodies/administration & dosage , Superoxide Dismutase/immunology , Animals , Disease Models, Animal , Female , Gliosis/immunology , Male , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/immunology , Protein Aggregation, Pathological/immunology , Spinal Cord/immunology , Spinal Cord/pathology , Superoxide Dismutase/genetics
19.
JCI Insight ; 3(19)2018 10 04.
Article in English | MEDLINE | ID: mdl-30282815

ABSTRACT

Neuroinflammation is a recognized pathogenic mechanism underlying motor neuron degeneration in amyotrophic lateral sclerosis (ALS), but the inflammatory mechanisms influencing peripheral motor axon degeneration remain largely unknown. A recent report showed a pathogenic role for c-Kit-expressing mast cells mediating inflammation and neuromuscular junction denervation in muscles from SOD1G93A rats. Here, we have explored whether mast cells infiltrate skeletal muscles in autopsied muscles from ALS patients. We report that degranulating mast cells were abundant in the quadriceps muscles from ALS subjects but not in controls. Mast cells were associated with myofibers and motor endplates and, remarkably, interacted with neutrophils forming large extracellular traps. Mast cells and neutrophils were also abundant around motor axons in the extensor digitorum longus muscle, sciatic nerve, and ventral roots of symptomatic SOD1G93A rats, indicating that immune cell infiltration extends along the entire peripheral motor pathway. Postparalysis treatment of SOD1G93A rats with the tyrosine kinase inhibitor drug masitinib prevented mast cell and neutrophil infiltration, axonal pathology, secondary demyelination, and the loss of type 2B myofibers, compared with vehicle-treated rats. These findings provide further evidence for a yet unrecognized contribution of immune cells in peripheral motor pathway degeneration that can be therapeutically targeted by tyrosine kinase inhibitors.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Mast Cells/immunology , Motor Neurons/pathology , Neuromuscular Junction/pathology , Neutrophils/immunology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/drug effects , Axons/immunology , Axons/pathology , Benzamides , Cell Degranulation/drug effects , Cell Degranulation/immunology , Disease Models, Animal , Humans , Male , Mast Cells/drug effects , Motor Neurons/cytology , Motor Neurons/immunology , Muscle, Skeletal/cytology , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Neuromuscular Junction/drug effects , Neuromuscular Junction/immunology , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Piperidines , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines , Rats , Rats, Transgenic , Superoxide Dismutase/genetics , Superoxide Dismutase-1/genetics , Thiazoles/pharmacology , Thiazoles/therapeutic use , Treatment Outcome
20.
Physiol Rep ; 6(17): e13812, 2018 09.
Article in English | MEDLINE | ID: mdl-30178608

ABSTRACT

Inflammation is known to alter nervous system function, but its effect on muscle spindle afferent mechanosensation and sensory integration in the spinal cord has not been well studied. We tested the hypothesis that systemic inflammation induced by an intraperitoneal injection of the endotoxin lipopolysaccharide (LPS; 7.5 × 105 endotoxin units/kg 18 h before experiment) would alter muscle spindle afferent mechanosensation and spinal cord excitability to Group Ia input in male and female adult C57Bl/6 mice. LPS injection caused a systemic immune response, evidenced by decreased white blood cell, monocyte, and lymphocyte concentrations in the blood, increased blood granulocyte concentration, and body weight loss. The immune response in both sexes was qualitatively similar. We used an in vitro muscle-nerve preparation to assay muscle spindle afferent response to stretch and vibration. LPS injection did not significantly change the response to stretch or vibration, with the exception of small decreases in the ability to entrain to high-frequency vibration in male mice. Similarly, LPS injection did not alter spinal cord excitability to Group Ia muscle spindle afferent input as measured by the Hoffman's reflex test in anesthetized mice (100 mg/kg ketamine, 10 mg/kg xylazine). Specifically, there were no changes in M or H wave latencies nor in the percentage of motor neurons excited by electrical afferent stimulation (Hmax /Mmax ). Overall, we found no major alterations in muscle proprioceptor function or sensory integration following exposure to LPS at a dose and time course that causes changes in nociceptor function and central processing.


Subject(s)
Mechanotransduction, Cellular , Muscle Spindles/physiology , Neurons, Afferent/physiology , Spinal Cord/physiology , Animals , Female , H-Reflex , Inflammation , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , Motor Neurons/immunology , Motor Neurons/physiology , Muscle Spindles/immunology , Neurons, Afferent/immunology , Proprioception , Spinal Cord/immunology , Vibration
SELECTION OF CITATIONS
SEARCH DETAIL
...