Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
BMJ Neurol Open ; 3(2): e000192, 2021.
Article in English | MEDLINE | ID: mdl-34786556

ABSTRACT

BACKGROUND AND OBJECTIVE: The concentration of neurofilament light (NfL) protein in cerebrospinal fluid (CSF) and blood is widely considered as a quantitative measure of neuro-axonal injury. Immune reactivity to NfL released into extracellular fluids induces specific autoantibody response. We investigated the levels and avidity of antibodies to NfL in patients with multiple sclerosis (MS) treated with disease-modifying therapies (DMTs) and their correlation with disease worsening and NfL protein concentration. METHODS: We conducted a prospective longitudinal study in 246 patients with MS (125 DMT-treated and 121 untreated at baseline). Serum levels of NfL antibodies, antibody avidity and immune complexes were determined by ELISA. NfL protein was measured using the Simoa platform. Clinical variables were tested for their association with the measured parameters in multivariate generalised estimating equation models. RESULTS: Multivariate analysis showed that levels of NfL antibodies were higher in progressive MS compared with clinically isolated syndrome (CIS)/relapsing remitting multiple sclerosis (RRMS) (p=0.010). Anti-NfL levels drop with increasing disability score (Expanded Disability Status Scale (EDSS)) (p=0.002), although conversely, were significantly elevated in CIS/RRMS after a recent EDSS increase (p=0.012). Patients receiving DMTs showed decreased levels of anti-NfL (p=0.008), high-avidity antibodies (p=0.017) and immune-complexes compared with untreated CIS/RRMS. Patients with MS switching to natalizumab showed lower levels of anti-NfL but higher immune complexes compared with healthy controls (p=0.0071). A weak association was observed between the levels of NfL protein and NfL antibodies. CONCLUSIONS: These results support the potential usefulness of quantifying antibody response to NfL as potential markers of progression and treatment response in MS and need to be considered when interpreting peripheral blood NfL levels.

2.
Clin Exp Immunol ; 206(3): 301-313, 2021 12.
Article in English | MEDLINE | ID: mdl-34510431

ABSTRACT

Neuropathology studies of amyotrophic lateral sclerosis (ALS) and animal models of ALS reveal a strong association between aberrant protein accumulation and motor neurone damage, as well as activated microglia and astrocytes. While the role of neuroinflammation in the pathology of ALS is unclear, imaging studies of the central nervous system (CNS) support the idea that innate immune activation occurs early in disease in both humans and rodent models of ALS. In addition, emerging studies also reveal changes in monocytes, macrophages and lymphocytes in peripheral blood as well as at the neuromuscular junction. To more clearly understand the association of neuroinflammation (innate and adaptive) with disease progression, the use of biomarkers and imaging modalities allow monitoring of immune parameters in the disease process. Such approaches are important for patient stratification, selection and inclusion in clinical trials, as well as to provide readouts of response to therapy. Here, we discuss the different imaging modalities, e.g. magnetic resonance imaging, magnetic resonance spectroscopy and positron emission tomography as well as other approaches, including biomarkers of inflammation in ALS, that aid the understanding of the underlying immune mechanisms associated with motor neurone degeneration in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/diagnostic imaging , Amyotrophic Lateral Sclerosis/pathology , Brain/diagnostic imaging , Magnetic Resonance Imaging/methods , Magnetic Resonance Spectroscopy/methods , Positron-Emission Tomography/methods , Amyotrophic Lateral Sclerosis/immunology , Animals , Brain/pathology , Disease Models, Animal , Disease Progression , Humans , Inflammation/pathology , Neuroinflammatory Diseases/diagnostic imaging , Neuroinflammatory Diseases/pathology
3.
Brain Commun ; 3(3): fcab148, 2021.
Article in English | MEDLINE | ID: mdl-34396108

ABSTRACT

Plasma proteome composition reflects the inflammatory and metabolic state of the organism and can be predictive of system-level and organ-specific pathologies. Circulating protein aggregates are enriched with neurofilament heavy chain-axonal proteins involved in brain aggregate formation and recently identified as biomarkers of the fatal neuromuscular disorder amyotrophic lateral sclerosis. Using unbiased proteomic methods, we have fully characterized the content in neuronal proteins of circulating protein aggregates from amyotrophic lateral sclerosis patients and healthy controls, with reference to brain protein aggregate composition. We also investigated circulating protein aggregate protein aggregation propensity, stability to proteolytic digestion and toxicity for neuronal and endothelial cell lines. Circulating protein aggregates separated by ultracentrifugation are visible as electron-dense macromolecular particles appearing as either large globular or as small filamentous formations. Analysis by mass spectrometry revealed that circulating protein aggregates obtained from patients are enriched with proteins involved in the proteasome system, possibly reflecting the underlying basis of dysregulated proteostasis seen in the disease, while those from healthy controls show enrichment of proteins involved in metabolism. Compared to the whole human proteome, proteins within circulating protein aggregates and brain aggregates show distinct chemical features of aggregation propensity, which appear dependent on the tissue or fluid of origin and not on the health status. Neurofilaments' two high-mass isoforms (460 and 268 kDa) showed a strong differential expression in amyotrophic lateral sclerosis compared to healthy control circulating protein aggregates, while aggregated neurofilament heavy chain was also partially resistant to enterokinase proteolysis in patients, demonstrated by immunoreactive bands at 171 and 31 kDa fragments not seen in digested healthy controls samples. Unbiased proteomics revealed that a total of 4973 proteins were commonly detected in circulating protein aggregates and brain, including 24 expressed from genes associated with amyotrophic lateral sclerosis. Interestingly, 285 circulating protein aggregate proteins (5.7%) were regulated (P < 0.05) and are present in biochemical pathways linked to disease pathogenesis and protein aggregation. Biologically, circulating protein aggregates from both patients and healthy controls had a more pronounced effect on the viability of hCMEC/D3 endothelial and PC12 neuronal cells compared to immunoglobulins extracted from the same plasma samples. Furthermore, circulating protein aggregates from patients exerted a more toxic effect than healthy control circulating protein aggregates on both cell lines at lower concentrations (P: 0.03, in both cases). This study demonstrates that circulating protein aggregates are significantly enriched with brain proteins which are representative of amyotrophic lateral sclerosis pathology and a potential source of biomarkers and therapeutic targets for this incurable disorder.

4.
Ann Clin Transl Neurol ; 8(9): 1831-1844, 2021 09.
Article in English | MEDLINE | ID: mdl-34318620

ABSTRACT

OBJECTIVE: To appraise the utility as biomarkers of blood antibodies and immune complexes to neurofilaments and dipeptide repeat proteins, the products of translation of the most common genetic mutation in amyotrophic lateral sclerosis (ALS). METHODS: Antibodies and immune complexes against neurofilament light, medium, heavy chains as well as poly-(GP)-(GR) dipeptide repeats were measured in blood samples from the ALS Biomarkers (n = 107) and the phenotype-genotype biomarker (n = 129) studies and in 140 healthy controls. Target analyte levels were studied longitudinally in 37 ALS cases. Participants were stratified according to the rate of disease progression estimated before and after baseline and C9orf72 genetic status. Survival and longitudinal analyses were undertaken with reference to matched neurofilament protein expression. RESULTS: Compared to healthy controls, total neurofilament proteins and antibodies, neurofilament light immune complexes (p < 0.0001), and neurofilament heavy antibodies (p = 0.0061) were significantly elevated in ALS, patients with faster progressing disease (p < 0.0001) and in ALS cases with a C9orf72 mutation (p < 0.0003). Blood neurofilament light protein discriminated better ALS from healthy controls (AUC: 0.92; p < 0.0001) and faster from slower progressing ALS (AUC: 0.86; p < 0.0001) compared to heavy-chain antibodies and light-chain immune complexes (AUC: 0.79; p < 0.0001 and AUC: 0.74; p < 0.0001). Lower neurofilament heavy antibodies were associated with longer survival (Log-rank Chi-square: 7.39; p = 0.0065). Increasing levels of antibodies and immune complexes between time points were observed in faster progressing ALS. CONCLUSIONS: We report a distinctive humoral response characterized by raising antibodies against neurofilaments and dipeptide repeats in faster progressing and C9orf72 genetic mutation carriers ALS patients. We confirm the significance of plasma neurofilament proteins in the clinical stratification of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Dipeptides , Disease Progression , Neurofilament Proteins , Adult , Aged , Amyotrophic Lateral Sclerosis/blood , Amyotrophic Lateral Sclerosis/diagnosis , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/physiopathology , Biomarkers , Cohort Studies , Dipeptides/blood , Dipeptides/immunology , Female , Humans , Longitudinal Studies , Male , Middle Aged , Neurofilament Proteins/blood , Neurofilament Proteins/immunology
5.
Immunology ; 152(4): 580-588, 2017 12.
Article in English | MEDLINE | ID: mdl-28718500

ABSTRACT

Antibodies to neuronal antigens are associated with many neurological diseases including paraneoplastic neurological disorders, epilepsy, amyotrophic lateral sclerosis and multiple sclerosis. Immunization with neuronal antigens such as neurofilament light (NF-L), a neuronal intermediate filament in axons, has been shown to induce neurological disease and spasticity in mice. Also, although antibodies to NF-L are widely used as surrogate biomarkers of axonal injury in amyotrophic lateral sclerosis and multiple sclerosis, it remains to be elucidated if antibodies to NF-L contribute to neurodegeneration and neurological disease. To address this, we examined the pathogenic role of antibodies directed to NF-L in vitro using spinal cord co-cultures and in vivo in experimental autoimmune encephalomyelitis (EAE) and optic neuritis animal models of multiple sclerosis. Here we show that peripheral injections of antibodies to NF-L augmented clinical signs of neurological disease in acute EAE, increased retinal ganglion cell loss in experimental optic neuritis and induced neurological signs following intracerebral injection into control mice. The pathogenicity of antibodies to NF-L was also observed in spinal cord co-cultures where axonal loss was induced. Taken together, our results reveal that as well as acting as reliable biomarkers of neuronal damage, antibodies to NF-L exacerbate neurological disease, suggesting that antibodies to NF-L generated during disease may also be pathogenic and play a role in the progression of neurodegeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Autoantibodies/immunology , Axons/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Intermediate Filaments/immunology , Optic Neuritis/immunology , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Male , Mice , Mice, Transgenic , Optic Neuritis/pathology , Retinal Ganglion Cells/immunology , Retinal Ganglion Cells/pathology , Spine/immunology , Spine/pathology
6.
Mult Scler ; 23(11): 1442-1452, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28537780

ABSTRACT

Neurodegeneration plays a key role in multiple sclerosis (MS) contributing to long-term disability in patients. The prognosis is, however, unpredictable coloured by complex disease mechanisms which can only be clearly appreciated using biomarkers specific to pathobiology of the underlying process. Here, we describe six promising neurodegenerative biomarkers in MS (neurofilament proteins, neurofilament antibodies, tau, N-acetylaspartate, chitinase and chitinase-like proteins and osteopontin), critically evaluating the evidence using a modified Bradford Hill criteria.


Subject(s)
Biomarkers , Multiple Sclerosis/diagnosis , Humans
7.
J Neuroimmune Pharmacol ; 11(4): 749-762, 2016 12.
Article in English | MEDLINE | ID: mdl-27518777

ABSTRACT

Parasite proteins containing repeats are essential invasion ligands, important for their ability to evade the host immune system and to induce immunosuppression. Here, the intrinsic suppressive potential of repetitive structures within parasite proteins was exploited to induce immunomodulation in order to establish self-tolerance in an animal model of autoimmune neurological disease. We tested the tolerogenic potential of fusion proteins containing repeat sequences of parasites linked to self-antigens. The fusion constructs consist of a recombinant protein containing repeat sequences derived from the S-antigen protein (SAg) of Plasmodium falciparum linked to a CD4 T cell epitope of myelin. They were tested for their efficacy to control the development of experimental autoimmune encephalomyelitis (EAE), In addition, we used the DO11.10 transgenic mouse model to study the immune mechanisms involved in tolerance induced by SAg fusion proteins. We found that repeated sequences of P. falciparum SAg protein linked to self-epitopes markedly protected mice from EAE. These fusion constructs were powerful tolerizing agents not only in a preventive setting but also in the treatment of ongoing disease. The tolerogenic effect was shown to be antigen-specific and strongly dependent on the physical linkage of the T cell epitope to the parasite structure and on the action of anti-inflammatory cytokines like IL-10 and TGF-ß. Other mechanisms include down-regulation of TNF-α accompanied by increased numbers of FoxP3+ cells. This study describes the use of repetitive structures from parasites linked to defined T cell epitopes as an effective method to induce antigen-specific tolerance with potential applicability for the treatment and prevention of autoimmune diseases.


Subject(s)
Autoantigens/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Plasmodium falciparum/immunology , Repetitive Sequences, Nucleic Acid/immunology , Amino Acid Sequence , Animals , Autoantigens/administration & dosage , Autoantigens/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Immunization/methods , Immunomodulation/drug effects , Immunomodulation/genetics , Immunomodulation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Parasites/genetics , Parasites/immunology , Plasmodium falciparum/genetics , Repetitive Sequences, Nucleic Acid/genetics
8.
Brain Pathol ; 26(2): 248-57, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26780491

ABSTRACT

Neurological dysfunction and motor neuron degeneration in amyotrophic lateral sclerosis (ALS) is strongly associated with neuroinflammation reflected by activated microglia and astrocytes in the CNS. In ALS endogenous triggers in the CNS such as aggregated protein and misfolded proteins activate a pathogenic response by innate immune cells. However, there is also strong evidence for a neuroprotective immune response in ALS. Emerging evidence also reveals changes in the peripheral adaptive immune responses as well as alterations in the blood brain barrier that may aid traffic of lymphocytes and antibodies into the CNS. Understanding the triggers of neuroinflammation is key to controlling neuronal loss. Here, we review the current knowledge regarding the roles of non-neuronal cells as well as the innate and adaptive immune responses in ALS. Existing ALS animal models, in particular genetic rodent models, are very useful to study the underlying pathogenic mechanisms of motor neuron degeneration. We also discuss the approaches used to target the pathogenic immune responses and boost the neuroprotective immune pathways as novel immunotherapies for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Blood-Brain Barrier/metabolism , Immune System/physiopathology , Lymphocytes/physiology , Neuroglia/physiology , Amyotrophic Lateral Sclerosis/drug therapy , Animals , Blood-Brain Barrier/drug effects , Humans , Immune System/drug effects , Lymphocytes/drug effects , Neuroglia/drug effects
9.
J Biol Chem ; 291(9): 4356-73, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26679998

ABSTRACT

The mitochondrial permeability transition pore is a recognized drug target for neurodegenerative conditions such as multiple sclerosis and for ischemia-reperfusion injury in the brain and heart. The peptidylprolyl isomerase, cyclophilin D (CypD, PPIF), is a positive regulator of the pore, and genetic down-regulation or knock-out improves outcomes in disease models. Current inhibitors of peptidylprolyl isomerases show no selectivity between the tightly conserved cyclophilin paralogs and exhibit significant off-target effects, immunosuppression, and toxicity. We therefore designed and synthesized a new mitochondrially targeted CypD inhibitor, JW47, using a quinolinium cation tethered to cyclosporine. X-ray analysis was used to validate the design concept, and biological evaluation revealed selective cellular inhibition of CypD and the permeability transition pore with reduced cellular toxicity compared with cyclosporine. In an experimental autoimmune encephalomyelitis disease model of neurodegeneration in multiple sclerosis, JW47 demonstrated significant protection of axons and improved motor assessments with minimal immunosuppression. These findings suggest that selective CypD inhibition may represent a viable therapeutic strategy for MS and identify quinolinium as a mitochondrial targeting group for in vivo use.


Subject(s)
Cerebral Cortex/drug effects , Cyclophilins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Multiple Sclerosis/prevention & control , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Quinolinium Compounds/therapeutic use , Amino Acid Substitution , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Peptidyl-Prolyl Isomerase F , Cyclophilins/genetics , Cyclophilins/metabolism , Cyclosporins/adverse effects , Cyclosporins/chemical synthesis , Cyclosporins/pharmacology , Cyclosporins/therapeutic use , Hep G2 Cells , Humans , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred Strains , Mice, Knockout , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Mutation , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacology , Peptides, Cyclic/adverse effects , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Quinolinium Compounds/adverse effects , Quinolinium Compounds/chemical synthesis , Quinolinium Compounds/pharmacology , Random Allocation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/pathology
10.
Int Immunol ; 27(3): 117-29, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25344935

ABSTRACT

The immune system is inextricably linked with many neurodegenerative diseases including amyotrophic lateral sclerosis (ALS), a devastating neuromuscular disorder affecting motor cell function with an average survival of 3 years from symptoms onset. In ALS, there is a dynamic interplay between the resident innate immune cells, that is, microglia and astrocytes, which may become progressively harmful to motor neurons. Although innate and adaptive immune responses are associated with progressive neurodegeneration, in the early stages of ALS immune activation pathways are primarily considered to be beneficial promoting neuronal repair of the damaged tissues, though a harmful effect of T cells at this stage of disease has also been observed. In addition, although auto-antibodies against neuronal antigens are present in ALS, it is unclear whether these arise as a primary or secondary event to neuronal damage, and whether the auto-antibodies are indeed pathogenic. Understanding how the immune system contributes to the fate of motor cells in ALS may shed light on the triggers of disease as well as on the mechanisms contributing to the propagation of the pathology. Immune markers may also act as biomarkers while pathways involved in immune action may be targets of new therapeutic strategies. Here, we review the modalities by which the immune system senses the core pathological process in motor neuron disorders, focusing on tissue-specific immune responses in the neuromuscular junction and in the neuroaxis observed in affected individuals and in animal models of ALS. We elaborate on existing data on the immunological fingerprint of ALS that could be used to identify clues on the disease origin and patterns of progression.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Astrocytes/physiology , Microglia/physiology , Motor Neurons/metabolism , Neurogenic Inflammation/immunology , Amyotrophic Lateral Sclerosis/therapy , Animals , Disease Models, Animal , Disease Progression , Humans , Immunity, Innate , Molecular Targeted Therapy , Motor Neurons/pathology , Neurogenic Inflammation/therapy , Neuromuscular Junction/immunology
11.
Mult Scler ; 20(10): 1355-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24515731

ABSTRACT

BACKGROUND: Increased levels of antibodies to neurofilament light protein (NF-L) in biological fluids have been found to reflect neuroinflammatory responses and neurodegeneration in multiple sclerosis (MS). OBJECTIVE: To evaluate whether levels of serum antibodies against NF-L correlate with clinical variants and treatment response in MS. METHODS: The autoantibody reactivity to NF-L protein was tested in serum samples from patients with relapsing-remitting MS (RRMS) (n=22) and secondary progressive MS (SPMS) (n=26). Two other cohorts of RRMS patients under treatment with natalizumab were analysed cross-sectionally (n=16) and longitudinally (n=24). The follow-up samples were taken at 6, 12, 18 and 24 months after treatment, and the NF-L antibody levels were compared against baseline levels. RESULTS: NF-L antibodies were higher in MS clinical groups than healthy controls and in RRMS compared to SPMS patients (p<0.001). NF-L antibody levels were lower in natalizumab treated than in untreated patients (p<0.001). In the longitudinal series, NF-L antibody levels decreased over time and a significant difference was found following 24 months of treatment compared with baseline measurements (p=0.001). CONCLUSIONS: Drug efficacy in MS treatment indicates the potential use of monitoring the content of antibodies against the NF-L chain as a predictive biomarker of treatment response in MS.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Autoantibodies/blood , Immunosuppressive Agents/therapeutic use , Multiple Sclerosis, Chronic Progressive/drug therapy , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Neurofilament Proteins/immunology , Adult , Biomarkers/blood , Case-Control Studies , Cross-Sectional Studies , Disability Evaluation , Drug Monitoring/methods , Enzyme-Linked Immunosorbent Assay , Europe , Female , Humans , Longitudinal Studies , Male , Middle Aged , Multiple Sclerosis, Chronic Progressive/blood , Multiple Sclerosis, Chronic Progressive/diagnosis , Multiple Sclerosis, Chronic Progressive/immunology , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Multiple Sclerosis, Relapsing-Remitting/immunology , Natalizumab , Predictive Value of Tests , Time Factors , Treatment Outcome
12.
J Neurol Neurosurg Psychiatry ; 85(3): 274-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24078718

ABSTRACT

BACKGROUND: Neurofilament (NF) proteins detection in biological fluids as a by-product of axonal loss is technically challenging and to date relies mostly on cerebrospinal fluid (CSF) measurements. Plasma antibodies against NF proteins and particularly to their soluble light chain (NF-L) could be a more practical surrogate marker for disease staging in amyotrophic lateral sclerosis (ALS), an invariably fatal and clinically heterogeneous neuromuscular disorder. METHODOLOGY: We have used a recombinant neurofilament light chain (NF-L) protein for the ELISA detection of antibodies against NF proteins in plasma samples from a well-characterised cohort of ALS individuals (n:73). The use of an established functional rating scale and of a recently proposed staging of disease progression allowed stratification of the ALS cohort based on disease stage, site of onset, survival and speed of disease progression. RESULTS: Antibody levels to NF proteins in plasma were significantly higher in ALS individuals compared to healthy controls (p<0.001). Higher NF plasma immunoreactivity was seen in advanced ALS cases (stage IVA-B) compared to earlier phases of the disease (p<0.05). There was no difference in anti-NF plasma antibodies between ALS individuals treated with riluzole and untreated patients; although riluzole-treated ALS cases with an earlier age of onset and with a shorter diagnostic delay displayed higher anti-NFL antibody levels compared to untreated ALS patients with similar features. CONCLUSIONS: Immunoreactivity to plasma NF-L and homologous NF proteins is informative of the stage of disease progression in ALS. The determination of NF antibody levels in plasma could be added to the growing panel of disease-monitoring biomarkers in ALS targeting cytoskeletal antigens.


Subject(s)
Amyotrophic Lateral Sclerosis/diagnosis , Neurofilament Proteins/immunology , Aged , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/immunology , Antibodies/blood , Antibodies/immunology , Biomarkers/blood , Case-Control Studies , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Neurofilament Proteins/blood , Neuroprotective Agents/therapeutic use , Recombinant Proteins , Riluzole/therapeutic use
13.
Mult Scler Relat Disord ; 3(2): 163-75, 2014 Mar.
Article in English | MEDLINE | ID: mdl-25878004

ABSTRACT

Multiple Sclerosis (MS) is a demyelinating disease characterized by chronic inflammation of the central nervous system (CNS) gray and white matter. Although the cause of MS is unknown, it is widely appreciated that innate and adaptive immune processes contribute to its pathogenesis. These include microglia/macrophage activation, pro-inflammatory T-cell (Th1) responses and humoral responses. Additionally, there is evidence indicating that MS has a neurodegenerative component since neuronal and axonal loss occurs even in the absence of overt inflammation. These aspects also form the rationale for clinical management of the disease. However, the currently available therapies to control the disease are only partially effective at best indicating that more effective therapeutic solutions are urgently needed. It is appreciated that in the immune-driven and neurodegenerative processes MS-specific deregulation of gene expressions and resulting protein dysfunction are thought to play a central role. These deviations in gene expression patterns contribute to the inflammatory response in the CNS, and to neuronal or axonal loss. Epigenetic mechanisms control transcription of most, if not all genes, in nucleated cells including cells of the CNS and in haematopoietic cells. MS-specific alterations in epigenetic regulation of gene expression may therefore lie at the heart of the deregulation of gene expression in MS. As such, epigenetic mechanisms most likely play an important role in disease pathogenesis. In this review we discuss a role for MS-specific deregulation of epigenetic features that control gene expression in the CNS and in the periphery. Furthermore, we discuss the application of small molecule inhibitors that target the epigenetic machinery to ameliorate disease in experimental animal models, indicating that such approaches may be applicable to MS patients.

14.
J Neuroinflammation ; 10: 118, 2013 Sep 22.
Article in English | MEDLINE | ID: mdl-24053384

ABSTRACT

BACKGROUND: Autoimmunity to neuronal proteins occurs in several neurological syndromes, where cellular and humoral responses are directed to surface as well as intracellular antigens. Similar to myelin autoimmunity, pathogenic immune response to neuroaxonal components such as neurofilaments may contribute to neurodegeneration in multiple sclerosis. METHODS: We studied the immune response to the axonal protein neurofilament light (NF-L) in the experimental autoimmune encephalomyelitis animal model of multiple sclerosis. To examine the association between T cells and axonal damage, pathology studies were performed on NF-L immunized mice. The interaction of T cells and axons was analyzed by confocal microscopy of central nervous system tissues and T-cell and antibody responses to immunodominant epitopes identified in ABH (H2-Ag7) and SJL/J (H2-As) mice. These epitopes, algorithm-predicted peptides and encephalitogenic motifs within NF-L were screened for encephalitogenicity. RESULTS: Confocal microscopy revealed both CD4+ and CD8+ T cells alongside damaged axons in the lesions of NF-L immunized mice. CD4+ T cells dominated the areas of axonal injury in the dorsal column of spastic mice in which the expression of granzyme B and perforin was detected. Identified NF-L epitopes induced mild neurological signs similar to the observed with the NF-L protein, yet distinct from those characteristic of neurological disease induced with myelin oligodendrocyte glycoprotein. CONCLUSIONS: Our data suggest that CD4+ T cells are associated with spasticity, axonal damage and neurodegeneration in NF-L immunized mice. In addition, defined T-cell epitopes in the NF-L protein might be involved in the pathogenesis of the disease.


Subject(s)
Autoantigens/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Neurofilament Proteins/immunology , Spinal Cord/immunology , Spinal Cord/pathology , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme-Linked Immunosorbent Assay , Epitopes, T-Lymphocyte/immunology , Female , Immunohistochemistry , Male , Mice , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes
15.
PLoS One ; 8(5): e64888, 2013.
Article in English | MEDLINE | ID: mdl-23738007

ABSTRACT

BACKGROUND: Autoimmune diseases result from a breakdown in self-tolerance to autoantigens. Self-tolerance is induced and sustained by central and peripheral mechanisms intended to deviate harmful immune responses and to maintain homeostasis, where regulatory T cells play a crucial role. The use of self-antigens in the study and treatment of a range of autoimmune diseases has been widely described; however, the mechanisms underlying the induced protection by these means are unclear. This study shows that protection of experimental autoimmune disease induced by T cell self-epitopes in a multimerized form (oligomers) is mediated by the induction of active suppression. PRINCIPAL FINDINGS: The experimental autoimmune encephalomyelitis (EAE) animal model for multiple sclerosis was used to study the mechanisms of protection induced by the treatment of oligomerized T cell epitope of myelin proteolipid protein (PLP139-151). Disease protection attained by the administration of oligomers was shown to be antigen specific and effective in both prevention and treatment of ongoing EAE. Oligomer mediated tolerance was actively transferred by cells from treated mice into adoptive hosts. The induction of active suppression was correlated with the recruitment of cells in the periphery associated with increased production of IL-10 and reduction of the pro-inflammatory cytokine TNF-α. The role of suppressive cytokines was demonstrated by the reversion of oligomer-induced protection after in vivo blocking of either IL-10 or TGF-ß cytokines. CONCLUSIONS: This study strongly supports an immunosuppressive role of repeat auto-antigens to control the development of EAE with potential applications in vaccination and antigen specific treatment of autoimmune diseases.


Subject(s)
Autoantigens/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Epitopes, T-Lymphocyte/immunology , Immunosuppression Therapy/methods , Peptide Fragments/immunology , Adoptive Transfer , Animals , Cell Proliferation/drug effects , Cytokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/therapy , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/pharmacology , Female , Mice , Myelin Proteolipid Protein/immunology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Protein Multimerization , Protein Structure, Quaternary , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
16.
CNS Neurol Disord Drug Targets ; 11(5): 570-88, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22583443

ABSTRACT

Multiple sclerosis (MS) is widely considered to be the result of an aggressive autoreactive T cell attack on myelin. How these autoimmune responses arise in MS is unclear, but they could result from virus infections. Thus, viral and autoimmune diseases in animals have been used to investigate the possible pathogenic mechanisms operating in MS. The autoimmune model, experimental autoimmune encephalomyelitis, is the most widely-used animal model and has greatly influenced therapeutic approaches targeting autoimmune responses. To investigate demyelination and remyelination in the absence of the adaptive immune response, toxin-induced demyelination models are used. These include using cuprizone, ethidium bromide and lysolecithin to induce myelin damage, which rapidly lead to remyelination when the toxins are withdrawn. The virus models include natural and experimental infections such as canine distemper, visna infection of sheep, and infection of non-human primates. The most commonly used viral models in rodents are Semliki Forest virus and Theiler's murine encephalomyelitis virus. The viral and experimental autoimmune encephalomyelitis models have been instrumental in the understanding of how viruses trigger inflammation, demyelination and neurodegeneration in the central nervous system. However, due to complexity of the animal models, pathological mechanisms are also examined in central nervous system cell culture systems including co-cultures, aggregate cultures and brain slice cultures. Here we critically review in vitro and in vivo models used to investigate MS. Since knowledge gained from these models forms the basis for the development of new therapeutic approaches for MS, we address the applicability of the models. Finally, we provide guidance for using and reporting animal studies with the aim of improving translational studies to the clinic.


Subject(s)
Autoimmunity , Disease Models, Animal , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Nerve Degeneration/etiology , Animals , Axons/immunology , Axons/pathology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Cell Line , Cells, Cultured , Central Nervous System/immunology , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Humans , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , Myelin Sheath/immunology , Myelin Sheath/pathology
17.
Mult Scler Relat Disord ; 1(1): 15-28, 2012 Jan.
Article in English | MEDLINE | ID: mdl-25876447

ABSTRACT

Although the primary cause of multiple sclerosis (MS) is unknown, the widely accepted view is that aberrant (auto)immune responses possibly arising following infection(s) are responsible for the destructive inflammatory demyelination and neurodegeneration in the central nervous system (CNS). This notion, and the limited access of human brain tissue early in the course of MS, has led to the development of autoimmune, viral and toxin-induced demyelination animal models as well as the development of human CNS cell and organotypic brain slice cultures in an attempt to understand events in MS. The autoimmune models, collectively known as experimental autoimmune encephalomyelitis (EAE), and viral models have shaped ideas of how environmental factors may trigger inflammation, demyelination and neurodegeneration in the CNS. Understandably, these models have also heavily influenced the development of therapies targeting the inflammatory aspect of MS. Demyelination and remyelination in the absence of overt inflammation are better studied in toxin-induced demyelination models using cuprizone and lysolecithin. The paradigm shift of MS as an autoimmune disease of myelin to a neurodegenerative disease has required more appropriate models reflecting the axonal and neuronal damage. Thus, secondary progressive EAE and spastic models have been crucial to develop neuroprotective approaches. In this review the current in vivo and in vitro experimental models to examine pathological mechanisms involved in inflammation, demyelination and neuronal degeneration, as well as remyelination and repair in MS are discussed. Since this knowledge is the basis for the development of new therapeutic approaches for MS, we particularly address whether the currently available models truly reflect the human disease, and discuss perspectives to further optimise and develop more suitable experimental models to study MS.

18.
Glia ; 60(3): 422-31, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22161990

ABSTRACT

Neuroaxonal degeneration is a pathological hallmark of multiple sclerosis (MS) contributing to irreversible neurological disability. Pathological mechanisms leading to axonal damage include autoimmunity to neuronal antigens. In actively demyelinating lesions, myelin is phagocytosed by microglia and blood-borne macrophages, whereas the fate of degenerating or damaged axons is unclear. Phagocytosis is essential for clearing neuronal debris to allow repair and regeneration. However, phagocytosis may lead to antigen presentation and autoimmunity, as has been described for neuroaxonal antigens. Despite this notion, it is unknown whether phagocytosis of neuronal antigens occurs in MS. Here, we show using novel, well-characterized antibodies to axonal antigens, that axonal damage is associated with HLA-DR expressing microglia/macrophages engulfing axonal bulbs, indicative of axonal damage. Neuronal proteins were frequently observed inside HLA-DR(+) cells in areas of axonal damage. In vitro, phagocytosis of neurofilament light (NF-L), present in white and gray matter, was observed in human microglia. The number of NF-L or myelin basic protein (MBP) positive cells was quantified using the mouse macrophage cell line J774.2. Intracellular colocalization of NF-L with the lysosomal membrane protein LAMP1 was observed using confocal microscopy confirming that NF-L is taken up and degraded by the cell. In vivo, NF-L and MBP was observed in cerebrospinal fluid cells from patients with MS, suggesting neuronal debris is drained by this route after axonal damage. In summary, neuroaxonal debris is engulfed, phagocytosed, and degraded by HLA-DR(+) cells. Although uptake is essential for clearing neuronal debris, phagocytic cells could also play a role in augmenting autoimmunity to neuronal antigens.


Subject(s)
Microglia/physiology , Multiple Sclerosis/pathology , Neurons/pathology , Phagocytosis/physiology , Adult , Aged , Aged, 80 and over , Animals , Cathepsin D/pharmacology , Cathepsins/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Female , HLA-DR Antigens/metabolism , Humans , Male , Mice , Microglia/drug effects , Microscopy, Confocal , Middle Aged , Multiple Sclerosis/cerebrospinal fluid , Myelin Basic Protein/cerebrospinal fluid , Myelin Basic Protein/metabolism , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Neurofilament Proteins/cerebrospinal fluid , Neurofilament Proteins/drug effects , Neurofilament Proteins/metabolism , Neurons/drug effects , Phagocytosis/drug effects , Time Factors
19.
Immunology ; 129(2): 154-69, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20561356

ABSTRACT

Neurodegeneration, the slow and progressive dysfunction and loss of neurons and axons in the central nervous system, is the primary pathological feature of acute and chronic neurodegenerative conditions such as Alzheimer's disease and Parkinson's disease, neurotropic viral infections, stroke, paraneoplastic disorders, traumatic brain injury and multiple sclerosis. Despite different triggering events, a common feature is chronic immune activation, in particular of microglia, the resident macrophages of the central nervous system. Apart from the pathogenic role of immune responses, emerging evidence indicates that immune responses are also critical for neuroregeneration. Here, we review the impact of innate and adaptive immune responses on the central nervous system in autoimmune, viral and other neurodegenerative disorders, and discuss their contribution to either damage or repair. We also discuss potential therapies aimed at the immune responses within the central nervous system. A better understanding of the interaction between the immune and nervous systems will be crucial to either target pathogenic responses, or augment the beneficial effects of immune responses as a strategy to intervene in chronic neurodegenerative diseases.


Subject(s)
Immunotherapy , Inflammation , Microglia/immunology , Neurodegenerative Diseases/immunology , Virus Diseases/immunology , Adaptive Immunity , Animals , Humans , Immunity, Innate , Models, Animal , Neurodegenerative Diseases/complications , Neurodegenerative Diseases/therapy , Virus Diseases/complications , Virus Diseases/therapy
20.
Blood ; 105(7): 2877-86, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15613550

ABSTRACT

Regulatory CD25(+)CD4+ T cells (Treg cells) are a central element of peripheral tolerance. Little is known, however, about phenotypic and functional characteristics of these cells with regard to memory. In this study we show that the chemokine receptor CCR6 is expressed on a distinct subset of mouse Treg cells. Similar to their CD25- counterparts, CCR6+ Treg cells exhibit markers of activation, memory, and expansion that are indicative for an effector-memory function. They are memory-like cells, generated in vivo from CCR6(-)CD25+ T cells after the encounter of antigen. As conventional CD25- effector-memory T cells, they have a high turnover rate and, in contrast to CCR6- Treg cells, they respond rapidly to restimulation in vitro with up-regulation of interleukin 10. CCR6+ Treg cells are enriched in the peripheral blood and accumulate in the central nervous system after induction of experimental autoimmune encephalomyelitis (EAE). This subset therefore seems to represent a population of regulatory effector-memory T cells (T(REM)), destined to control potentially destructive immune responses directly in inflamed tissues. Importantly, these cells are also present in humans. Here the expression of CCR6 fully cosegregates with CD45RO, an established marker of human memory T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory/physiology , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Division/immunology , Chemokine CCL20 , Chemokines, CC/pharmacology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression/immunology , Interleukin-10/metabolism , Leukocyte Common Antigens/metabolism , Macrophage Inflammatory Proteins/pharmacology , Mice , Phenotype , Receptors, CCR6 , Receptors, Interleukin-2/metabolism , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...