Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Sci Rep ; 11(1): 7841, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33837283

ABSTRACT

Type 1 narcolepsy is strongly (98%) associated with human leukocyte antigen (HLA) class II DQA1*01:02/DQB1*06:02 (DQ0602) and highly associated with T cell receptor (TCR) alpha locus polymorphism as well as other immune regulatory loci. Increased incidence of narcolepsy was detected following the 2009 H1N1 pandemic and linked to Pandemrix vaccination, strongly supporting that narcolepsy is an autoimmune disorder. Although recent results suggest CD4+ T cell reactivity to neuropeptide hypocretin/orexin and cross-reactive flu peptide is involved, identification of other autoantigens has remained elusive. Here we study whether autoimmunity directed against Regulatory Factor X4 (RFX4), a protein co-localized with hypocretin, is involved in some cases of narcolepsy. Studying human serum, we found that autoantibodies against RFX4 were rare. Using RFX4 peptides bound to DQ0602 tetramers, antigen RFX4-86, -95, and -60 specific human CD4+ T cells were detected in 4/10 patients and 2 unaffected siblings, but not in others. Following culture with each cognate peptide, enriched autoreactive TCRαß clones were isolated by single-cell sorting and TCR sequenced. Homologous clones bearing TRBV4-2 and recognizing RFX4-86 in patients and one twin control of patient were identified. These results suggest the involvement of RFX4 CD4+ T cell autoreactivity in some cases of narcolepsy, but also in healthy donors.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity , CD4-Positive T-Lymphocytes/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/adverse effects , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Narcolepsy/immunology , Regulatory Factor X Transcription Factors/immunology , Vaccination/methods , Adolescent , Adult , Autoantibodies/blood , Autoantibodies/immunology , Autoantigens/blood , Autoantigens/immunology , Autoimmune Diseases/blood , Case-Control Studies , Child , Cross Reactions , Female , HEK293 Cells , Humans , Influenza, Human/virology , Male , Narcolepsy/blood , Orexins/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Regulatory Factor X Transcription Factors/genetics , Transfection , Young Adult
2.
Rev Neurol ; 71(12): 460-466, 2020 Dec 16.
Article in Spanish | MEDLINE | ID: mdl-33319349

ABSTRACT

INTRODUCTION: The orexinergic system is one of the chemical mediators that modulate the gut-brain axis, given the involvement of hypothalamic orexin A (OXA) in gastrointestinal motility and secretion, and the presence of OXA in enteroendocrine cells of the intestinal mucosa and in primary afferent neurons of the mesenteric plexus, permitting its participation in gut-brain signaling. AIM: The source of OXA and the signal(s) triggering its peripheral release are not fully understood, and it is not known whether it acts on orexigenic receptors in peripheral tissues to meet physiological or pathological demands. The aim of this review is to address these questions in the light of new data indicating that OXA may have functions in the gut-brain axis that go beyond its participation in energy homeostasis. DEVELOPMENT: OXA in the enteric system protects against systemic and central inflammation, and hypothalamic OXA orchestrates numerous peripheral effects to suppress the systemic inflammatory response. For this reason, OXA may act as an immunomodulator in chronic inflammations or autoimmune diseases. OXA is also involved in the stress response, regulating physiological responses to emotional or stressful stimuli. CONCLUSIONS: OXA exerts anti-inflammatory and gastroprotective effects on the intestinal mucosa; however, it may increase the response to external and/or internal stress in individuals with chronic inflammation, exacerbating the gastrointestinal inflammation. Hence, pharmacologic interventions in the orexinergic system have been proposed to treat diseases in which intestinal hypersensitivity is combined with appetite loss, sleep disturbance, stress, and anxiety.


TITLE: Orexina A como mediadora en el diálogo intestino-cerebro.Introducción. Entre los mediadores químicos que modulan el eje intestino-cerebro debe incluirse el sistema orexinérgico, ya que la orexina A (OXA) hipotalámica interviene en la motilidad y en la secreción gastrointestinal. También está presente en las células enteroendocrinas de la mucosa intestinal y en las neuronas aferentes primarias del plexo mientérico, y puede intervenir en la señalización intestino-cerebro. Objetivo. No se conoce con exactitud la fuente ni la señal que originan la liberación de OXA periférica, ni tampoco si actúa en los receptores orexinérgicos de los tejidos periféricos ante demandas fisiológicas o patológicas. Esta revisión intenta analizar estas cuestiones a la luz de nuevos datos que indican que la OXA en el eje intestino-cerebro puede tener funciones más allá de su participación en la homeostasis energética. Desarrollo. La OXA en el sistema entérico protege de la inflamación sistémica y central, y en el hipotálamo orquesta numerosos efectos periféricos para suprimir la respuesta inflamatoria sistémica. Por ello, podría actuar como sustancia inmunomoduladora en inflamaciones crónicas o en enfermedades autoinmunitarias. La OXA también se relaciona con la respuesta de estrés, regulando las respuestas fisiológicas a estímulos emocionales o estresantes. Conclusiones. Aunque la OXA tiene efectos antiinflamatorios y gastroprotectores de la mucosa intestinal, en procesos de inflamación crónica podría incrementar la respuesta a estímulos estresantes, tanto externos como internos, y exacerbar la inflamación gastrointestinal. Por ello, se han propuesto intervenciones farmacológicas sobre el sistema orexinérgico como tratamiento para enfermedades en las que la hipersensibilidad intestinal coexiste con pérdida de apetito, alteraciones del sueño, estrés y ansiedad.


Subject(s)
Gastrointestinal Tract/immunology , Gastrointestinal Tract/physiology , Orexins/immunology , Orexins/physiology , Signal Transduction/physiology , Animals , Humans , Hypothalamus/metabolism , Hypothalamus/physiology , Mice , Neuroimmunomodulation/physiology , Neurons/physiology , Neuropeptides/immunology , Neuropeptides/metabolism , Neuropeptides/physiology , Neurotransmitter Agents/immunology , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/physiology , Orexin Receptors/physiology , Orexins/metabolism , Psychological Distress
3.
Int J Mol Sci ; 20(21)2019 Nov 05.
Article in English | MEDLINE | ID: mdl-31694154

ABSTRACT

Recent data highlight the important roles of the gut microbiome, gut permeability, and alterations in mitochondria functioning in the pathophysiology of multiple sclerosis (MS). This article reviews such data, indicating two important aspects of alterations in the gut in the modulation of mitochondria: (1) Gut permeability increases toll-like receptor (TLR) activators, viz circulating lipopolysaccharide (LPS), and exosomal high-mobility group box (HMGB)1. LPS and HMGB1 increase inducible nitric oxide synthase and superoxide, leading to peroxynitrite-driven acidic sphingomyelinase and ceramide. Ceramide is a major driver of MS pathophysiology via its impacts on glia mitochondria functioning; (2) Gut dysbiosis lowers production of the short-chain fatty acid, butyrate. Butyrate is a significant positive regulator of mitochondrial function, as well as suppressing the levels and effects of ceramide. Ceramide acts to suppress the circadian optimizers of mitochondria functioning, viz daytime orexin and night-time melatonin. Orexin, melatonin, and butyrate increase mitochondria oxidative phosphorylation partly via the disinhibition of the pyruvate dehydrogenase complex, leading to an increase in acetyl-coenzyme A (CoA). Acetyl-CoA is a necessary co-substrate for activation of the mitochondria melatonergic pathway, allowing melatonin to optimize mitochondrial function. Data would indicate that gut-driven alterations in ceramide and mitochondrial function, particularly in glia and immune cells, underpin MS pathophysiology. Aryl hydrocarbon receptor (AhR) activators, such as stress-induced kynurenine and air pollutants, may interact with the mitochondrial melatonergic pathway via AhR-induced cytochrome P450 (CYP)1b1, which backward converts melatonin to N-acetylserotonin (NAS). The loss of mitochnodria melatonin coupled with increased NAS has implications for altered mitochondrial function in many cell types that are relevant to MS pathophysiology. NAS is increased in secondary progressive MS, indicating a role for changes in the mitochondria melatonergic pathway in the progression of MS symptomatology. This provides a framework for the integration of diverse bodies of data on MS pathophysiology, with a number of readily applicable treatment interventions, including the utilization of sodium butyrate.


Subject(s)
Ceramides/immunology , Gastrointestinal Microbiome , Melatonin/immunology , Multiple Sclerosis/immunology , Orexins/immunology , Platelet Activation , Animals , Butyrates/immunology , Chronobiology Disorders/immunology , Chronobiology Disorders/microbiology , Chronobiology Disorders/pathology , Chronobiology Disorders/physiopathology , Humans , Immunity, Cellular , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Inflammation/physiopathology , Mitochondria/immunology , Mitochondria/pathology , Multiple Sclerosis/microbiology , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Neuroglia/immunology , Neuroglia/pathology
4.
Ann Clin Transl Neurol ; 6(12): 2566-2572, 2019 12.
Article in English | MEDLINE | ID: mdl-31730293

ABSTRACT

Narcolepsy type 1 (NT1) is caused by severe loss of the orexin neurons, and is highly associated with HLA DQB1*06:02. Using intracellular cytokine staining, we observed a higher frequency of IFN-γ- and TNF-α-producing CD4+ and CD8+ T-cells in response to orexins in 27 children with NT1 compared to 15 healthy control children. Conversely, no such difference was observed between 14 NT1 and 16 HC adults. In addition, priming with flu peptides amplified the T-cell response to orexins in children with NT1. Our data suggests that NT1 may be caused by an autoimmune T-cell response to orexins, possibly triggered by flu antigens.


Subject(s)
Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Narcolepsy/immunology , Orexins/immunology , Tumor Necrosis Factor-alpha/immunology , Adolescent , Adult , Child , Female , Humans , Male , Middle Aged , Narcolepsy/blood , Narcolepsy/etiology , Young Adult
5.
Nat Commun ; 10(1): 5247, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31748512

ABSTRACT

Individuals with narcolepsy suffer from abnormal sleep patterns due to loss of neurons that uniquely supply hypocretin (HCRT). Previous studies found associations of narcolepsy with the human leukocyte antigen (HLA)-DQ6 allele and T-cell receptor α (TRA) J24 gene segment and also suggested that in vitro-stimulated T cells can target HCRT. Here, we present evidence of in vivo expansion of DQ6-HCRT tetramer+/TRAJ24+/CD4+ T cells in DQ6+ individuals with and without narcolepsy. We identify related TRAJ24+ TCRαß clonotypes encoded by identical α/ß gene regions from two patients and two controls. TRAJ24-G allele+ clonotypes only expand in the two patients, whereas a TRAJ24-C allele+ clonotype expands in a control. A representative tetramer+/G-allele+ TCR shows signaling reactivity to the epitope HCRT87-97. Clonally expanded G-allele+ T cells exhibit an unconventional effector phenotype. Our analysis of in vivo expansion of HCRT-reactive TRAJ24+ cells opens an avenue for further investigation of the autoimmune contribution to narcolepsy development.


Subject(s)
Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Narcolepsy/immunology , Orexins/immunology , Animals , Autoimmunity/genetics , Case-Control Studies , Cell Proliferation , Crystallography, X-Ray , Drosophila , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , Humans , Immunoglobulin Joining Region/genetics , Narcolepsy/genetics , Peripheral Tolerance , Phenotype , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology
6.
Med Hypotheses ; 131: 109294, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31443760

ABSTRACT

Narcolepsy type 1 is a lifelong sleep disorder characterized by the loss of hypocretin-producing neurons in the brain. Environmental agents, including influenza, neurotoxic metals, and combustion smoke, have been implicated in the pathogenesis, especially in carriers of the human leukocyte antigen class II DQB1*06:02 allele. Sensitive experimental approaches have recently revealed hypocretin-autoreactive CD4+ and CD8+ T cells in the blood of narcoleptic patients. However, such potentially harmful cells are also detectable, to a lesser degree, in control DQB1*06:02 carriers, suggesting that the integrity of the blood-brain barrier (BBB) provides a neuroprotective effect. Here, we present the hypothesis that external toxic agents induce neuroinflammation in the olfactory bulb and concomitant overproduction of proinflammatory cytokines (e.g., tumor necrosis factor-α and interferon-γ); this, in turn, compromises the BBB, allowing autoimmune cells to access and kill hypocretinergic neurons. Such sequential pathological alterations could occur insidiously, passing unnoticed and consequently being underestimated. The elevated number of autoreactive T cells in narcoleptics relative to controls might reflect externally induced immunomodulation rather than a direct disease trigger.


Subject(s)
Blood-Brain Barrier/physiology , HLA-DQ beta-Chains/immunology , Models, Immunological , Narcolepsy/immunology , Olfactory Bulb/physiopathology , Animals , Autoantigens/immunology , Autoantigens/metabolism , Cytokines/physiology , Environmental Pollutants/pharmacokinetics , Environmental Pollutants/toxicity , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Mice , Mice, Transgenic , Microglia/metabolism , Molecular Mimicry , Narcolepsy/etiology , Narcolepsy/physiopathology , Neurons/metabolism , Neurons/pathology , Olfactory Bulb/drug effects , Orexins/immunology , Orexins/metabolism , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/immunology
7.
J Neuroimmunol ; 332: 167-175, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31048269

ABSTRACT

Following the 2009 H1N1 influenza pandemic, an increased risk of narcolepsy type 1 was observed. Homology between an H1N1 hemagglutinin and two hypocretin sequences has been reported. T cell reactivity to these peptides was assessed in 81 narcolepsy type 1 patients and 19 HLA-DQ6-matched healthy controls. HLA-DQ6-restricted H1N1 hemagglutinin-specific T cell responses were detected in 28.4% of patients and 15.8% of controls. Despite structural homology between HLA-DQ6-hypocretin and -H1N1 peptide complexes, T cell cross-reactivity was not detected. These results indicate that it is unlikely that cross-reactivity between H1N1 hemagglutinin and hypocretin peptides presented by HLA-DQ6 is involved in the development of narcolepsy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HLA-DQ Antigens/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Narcolepsy/immunology , Orexins/immunology , T-Lymphocyte Subsets/immunology , Adolescent , Adult , Cerebrospinal Fluid Proteins/analysis , Child , Crystallography, X-Ray , Female , HLA-DQ Antigens/chemistry , HLA-DQ alpha-Chains/analysis , HLA-DQ beta-Chains/analysis , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Humans , Influenza A Virus, H1N1 Subtype , Male , Middle Aged , Models, Molecular , Molecular Mimicry , Narcolepsy/etiology , Orexins/cerebrospinal fluid , Orexins/chemistry , Pandemics , Peptide Fragments/chemistry , Peptide Fragments/immunology , Protein Conformation , Young Adult
8.
J Neurol ; 266(7): 1809-1815, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31004212

ABSTRACT

The last two decades have seen an explosion in our understanding of the clinical nature of narcolepsy and its pathogenesis, fuelling new approaches to potentially effective treatments. It is now recognised that the full narcoleptic syndrome has significant adverse effects on sleep regulation across the full 24-h period and is often associated with clinical features outside the sleep-wake domain. The discovery that most narcoleptic subjects specifically lack a hypothalamic neuropeptide (hypocretin, also called orexin) was a truly original and landmark observation in 1999, greatly furthering our understanding both of the syndrome itself and sleep biology in general. An autoimmune pathophysiology has long been suggested by the tight association with specific histocompatibility antigens and very recently partly confirmed by detailed analysis of T-cell immunological function in affected subjects. Drug treatments remain symptomatic but may soon become more focussed by restoring central hypocretin signalling with replacement therapy. Potentially disease-modifying, immunological approaches have yet to be studied systematically, although the interval between disease onset and development of the full clinical syndrome may be longer than previously appreciated, affording a realistic window of opportunity for limiting neuronal damage in this disabling condition.


Subject(s)
Brain/immunology , Narcolepsy/immunology , Nerve Net/immunology , Orexins/immunology , Brain/metabolism , Emotions/physiology , Humans , Narcolepsy/diagnosis , Narcolepsy/metabolism , Nerve Net/metabolism , Orexins/metabolism
10.
Nat Commun ; 10(1): 837, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30783092

ABSTRACT

Narcolepsy Type 1 (NT1) is a neurological sleep disorder, characterized by the loss of hypocretin/orexin signaling in the brain. Genetic, epidemiological and experimental data support the hypothesis that NT1 is a T-cell-mediated autoimmune disease targeting the hypocretin producing neurons. While autoreactive CD4+ T cells have been detected in patients, CD8+ T cells have only been examined to a minor extent. Here we detect CD8+ T cells specific toward narcolepsy-relevant peptides presented primarily by NT1-associated HLA types in the blood of 20 patients with NT1 as well as in 52 healthy controls, using peptide-MHC-I multimers labeled with DNA barcodes. In healthy controls carrying the disease-predisposing HLA-DQB1*06:02 allele, the frequency of autoreactive CD8+ T cells was lower as compared with both NT1 patients and HLA-DQB1*06:02-negative healthy individuals. These findings suggest that a certain level of CD8+ T-cell reactivity combined with HLA-DQB1*06:02 expression is important for NT1 development.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HLA-DQ beta-Chains/genetics , Narcolepsy/immunology , Orexins/immunology , Peptides/immunology , Adolescent , Adult , Case-Control Studies , Child , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Narcolepsy/genetics , Neurons/metabolism , Orexins/metabolism
12.
Proc Natl Acad Sci U S A ; 115(52): E12323-E12332, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30541895

ABSTRACT

Type 1 narcolepsy (T1N) is caused by hypocretin/orexin (HCRT) neuronal loss. Association with the HLA DQB1*06:02/DQA1*01:02 (98% vs. 25%) heterodimer (DQ0602), T cell receptors (TCR) and other immune loci suggest autoimmunity but autoantigens are unknown. Onset is seasonal and associated with influenza A, notably pandemic 2009 H1N1 (pH1N1) infection and vaccination (Pandemrix). Peptides derived from HCRT and influenza A, including pH1N1, were screened for DQ0602 binding and presence of cognate DQ0602 tetramer-peptide-specific CD4+ T cells tested in 35 T1N cases and 22 DQ0602 controls. Higher reactivity to influenza pHA273-287 (pH1N1 specific), PR8 (H1N1 pre-2009 and H2N2)-specific NP17-31 and C-amidated but not native version of HCRT54-66 and HCRT86-97 (HCRTNH2) were observed in T1N. Single-cell TCR sequencing revealed sharing of CDR3ß TRBV4-2-CASSQETQGRNYGYTF in HCRTNH2 and pHA273-287-tetramers, suggesting molecular mimicry. This public CDR3ß uses TRBV4-2, a segment modulated by T1N-associated SNP rs1008599, suggesting causality. TCR-α/ß CDR3 motifs of HCRT54-66-NH2 and HCRT86-97-NH2 tetramers were extensively shared: notably public CDR3α, TRAV2-CAVETDSWGKLQF-TRAJ24, that uses TRAJ24, a chain modulated by T1N-associated SNPs rs1154155 and rs1483979. TCR-α/ß CDR3 sequences found in pHA273-287, NP17-31, and HCRTNH2 tetramer-positive CD4+ cells were also retrieved in single INF-γ-secreting CD4+ sorted cells stimulated with Pandemrix, independently confirming these results. Our results provide evidence for autoimmunity and molecular mimicry with flu antigens modulated by genetic components in the pathophysiology of T1N.


Subject(s)
Narcolepsy/immunology , Orexins/immunology , Orexins/metabolism , Adolescent , Adult , Autoantigens/metabolism , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Child , Epitopes/immunology , Female , HLA-DQ beta-Chains , Hemagglutinins , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A virus/immunology , Influenza A virus/pathogenicity , Influenza, Human/immunology , Intracellular Signaling Peptides and Proteins/genetics , Male , Middle Aged , Molecular Mimicry/immunology , Orexins/genetics , Peptides/genetics , Receptors, Antigen, T-Cell/genetics , Vaccination
13.
Nature ; 562(7725): 63-68, 2018 10.
Article in English | MEDLINE | ID: mdl-30232458

ABSTRACT

Narcolepsy is a chronic sleep disorder caused by the loss of neurons that produce hypocretin. The close association with HLA-DQB1*06:02, evidence for immune dysregulation and increased incidence upon influenza vaccination together suggest that this disorder has an autoimmune origin. However, there is little evidence of autoreactive lymphocytes in patients with narcolepsy. Here we used sensitive cellular screens and detected hypocretin-specific CD4+ T cells in all 19 patients that we tested; T cells specific for tribbles homologue 2-another self-antigen of hypocretin neurons-were found in 8 out of 13 patients. Autoreactive CD4+ T cells were polyclonal, targeted multiple epitopes, were restricted primarily by HLA-DR and did not cross-react with influenza antigens. Hypocretin-specific CD8+ T cells were also detected in the blood and cerebrospinal fluid of several patients with narcolepsy. Autoreactive clonotypes were serially detected in the blood of the same-and even of different-patients, but not in healthy control individuals. These findings solidify the autoimmune aetiology of narcolepsy and provide a basis for rapid diagnosis and treatment of this disease.


Subject(s)
Autoantigens/immunology , Autoantigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Narcolepsy/immunology , Neurons/immunology , Neurons/metabolism , Orexins/immunology , Orexins/metabolism , Antigens, Viral , Autoimmune Diseases/diagnosis , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Calcium-Calmodulin-Dependent Protein Kinases/immunology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Case-Control Studies , Cell Separation , Cross Reactions , Humans , Immunologic Memory , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Narcolepsy/blood , Narcolepsy/cerebrospinal fluid , Narcolepsy/diagnosis , Orthomyxoviridae/immunology
14.
J Neuroimmunol ; 309: 7-11, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28601291

ABSTRACT

Narcolepsy type 1, a neurological sleep disorder strongly associated with Human Leukocyte Antigen (HLA-)DQB1*06:02, is caused by the loss of hypothalamic neurons producing the wake-promoting neuropeptide hypocretin (hcrt, also known as orexin). This loss is believed to be caused by an autoimmune reaction. To test whether hcrt itself could be a possible target in the autoimmune attack, CD4+ T-cell reactivity towards six different 15-mer peptides from prepro-hypocretin with high predicted affinity to the DQA1*01:02/DQB1*06:02 MHC class II dimer was tested using EliSpot in a cohort of 22 narcolepsy patients with low CSF hcrt levels, and 23 DQB1*06:02 positive healthy controls. Our ELISpot assay had a detection limit of 1:10,000 cells. We present data showing that autoreactive CD4+ T-cells targeting epitopes from the hcrt precursor in the context of MHC-DQA1*01:02/DQB1*06:02 are either not present or present in a frequency is <1:10,000 among peripheral CD4+ T-cells from narcolepsy type 1 patients.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Enzyme-Linked Immunospot Assay/methods , Epitopes/metabolism , HLA-DQ alpha-Chains/blood , HLA-DQ beta-Chains/blood , Narcolepsy/blood , Orexins/blood , Adolescent , Adult , Amino Acid Sequence , CD4-Positive T-Lymphocytes/immunology , Child , Cohort Studies , Epitopes/immunology , Female , HLA-DQ alpha-Chains/immunology , HLA-DQ beta-Chains/immunology , Humans , Male , Middle Aged , Narcolepsy/diagnosis , Narcolepsy/immunology , Orexins/immunology , Young Adult
15.
Ross Fiziol Zh Im I M Sechenova ; 103(3): 217-29, 2017 Mar.
Article in Russian | MEDLINE | ID: mdl-30199203

ABSTRACT

The review represents a modern concept about cells-molecular basis of mechanisms of neuro-immune interactions, the data on the effects of destabilizing factors (electric pain stimulation, rotation, cold and psychoemotional stress) on the functioning of neurons and immune cells. It must be underlined, that under the stress conditions take place the alterations of ligand-receptors interactions on the membrane of lymphocyte. In particular the reaction of these cells to regulating signal - application of Interleikin-1 grow up after mild stress, but it falls down after an influence of severe stress factors. Special attention is paid to the role of the orexinergic system in mechanism of realization of CNS reactions to application of antigens. In the present work the possible methods of correction of imbalance in functional interactions between nervous and immune systems, caused by different destabilizing factors, are reviewed.


Subject(s)
Central Nervous System/immunology , Immune System/metabolism , Neuroimmunomodulation/genetics , Signal Transduction/immunology , Stress, Psychological/immunology , Animals , Cell Differentiation/drug effects , Central Nervous System/metabolism , Central Nervous System/physiopathology , Corticosterone/blood , Corticosterone/immunology , DNA/pharmacology , Gene Expression Regulation , Humans , Immune System/physiopathology , Immunologic Factors/pharmacology , Interleukin-1alpha/blood , Interleukin-1alpha/immunology , Interleukin-1beta/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Neuroimmunomodulation/drug effects , Neurons/immunology , Neurons/metabolism , Orexin Receptors/genetics , Orexin Receptors/immunology , Orexins/genetics , Orexins/immunology , Signal Transduction/genetics , Stress, Psychological/genetics , Stress, Psychological/physiopathology , Stress, Psychological/prevention & control , Thymocytes/cytology , Thymocytes/drug effects , Thymocytes/immunology
17.
Sleep ; 39(3): 581-7, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26518595

ABSTRACT

STUDY OBJECTIVES: Narcolepsy with cataplexy is tightly associated with the HLA class II allele DQB1*06:02. Evidence indicates a complex contribution of HLA class II genes to narcolepsy susceptibility with a recent independent association with HLA-DPB1. The cause of narcolepsy is supposed be an autoimmune attack against hypocretin-producing neurons. Despite the strong association with HLA class II, there is no evidence for CD4+ T-cell-mediated mechanism in narcolepsy. Since neurons express class I and not class II molecules, the final effector immune cells involved might include class I-restricted CD8+ T-cells. METHODS: HLA class I (A, B, and C) and II (DQB1) genotypes were analyzed in 944 European narcolepsy with cataplexy patients and in 4,043 control subjects matched by country of origin. All patients and controls were DQB1*06:02 positive and class I associations were conditioned on DQB1 alleles. RESULTS: HLA-A*11:01 (OR = 1.49 [1.18-1.87] P = 7.0*10(-4)), C*04:01 (OR = 1.34 [1.10-1.63] P = 3.23*10(-3)), and B*35:01 (OR = 1.46 [1.13-1.89] P = 3.64*10(-3)) were associated with susceptibility to narcolepsy. Analysis of polymorphic class I amino-acids revealed even stronger associations with key antigen-binding residues HLA-A-Tyr(9) (OR = 1.32 [1.15-1.52] P = 6.95*10(-5)) and HLA-C-Ser(11) (OR = 1.34 [1.15-1.57] P = 2.43*10(-4)). CONCLUSIONS: Our findings provide a genetic basis for increased susceptibility to infectious factors or an immune cytotoxic mechanism in narcolepsy, potentially targeting hypocretin neurons.


Subject(s)
Alleles , CD8-Positive T-Lymphocytes/immunology , Cataplexy/genetics , Cataplexy/immunology , Histocompatibility Antigens Class I/genetics , Neurons/immunology , Neurons/pathology , Case-Control Studies , Cataplexy/pathology , Epitopes/immunology , Europe/ethnology , Genetic Predisposition to Disease , HLA-DQ beta-Chains/genetics , Haplotypes/genetics , Haplotypes/immunology , Humans , Linkage Disequilibrium , Neurons/metabolism , Odds Ratio , Orexins/immunology , Orexins/metabolism , T-Lymphocytes, Cytotoxic/immunology , White People/genetics
18.
Neuro Endocrinol Lett ; 36(5): 458-64, 2015.
Article in English | MEDLINE | ID: mdl-26707047

ABSTRACT

OBJECTIVES: Many of peptides synthesized in gastrointestinal tract (GI) and adipose tissues, regulate growth and food intake. The GI microflora is an antigenic source. Based on the molecular mimicry hypothesis, intestinal microbe-derived antigens may trigger the production of autoantibodies cross-reacting with some neuropeptides. DESIGN: The aim of the study was to assess whether in idiopathic short stature (ISS) children with Candida albicans (C.albicans) colonisation and/or Helicobacter pylori (H.pylori) infection the autoantibodies (in positive levels) against selected neuropeptides [anti-NP Abs(+)]: ghrelin, leptin, orexin A, αMSH are more prevalent than in Controls. SETTING: The study group comprised 64 children with ISS and 36 children with normal height (Controls). In each child, IgG antibodies against H.pylori, ghrelin, leptin, orexin A and αMSH were assessed in serum, while presence of C.albicans - in stool samples. RESULTS: The higher prevalence of anti-NP Abs(+) in ISS children with C.albicans and/or H.pylori than in normal height children with the colonization in question (34.4% vs 21.1%, p<0.01) was found. The prevalence of anti-NP Abs(+) in groups of children without C.albicans and H.pylori were low, anti-NP Abs(+) were detected in 9.4% of ISS children only, while in Controls they were not found. CONCLUSIONS: In short children with C.albicans and/or H.pylori the incidence of autoantibodies against selected neuropeptides is high. It probably is connected with molecular mimicry between antigens of these microbiota and the mentioned peptides. It is tempting to speculate that presence of cross-reacting autoantibodies against regulatory neuropeptides may results in worse growth velocity. However, further studies are necessary to elucidate this issue.


Subject(s)
Autoantibodies/immunology , Candidiasis/immunology , Growth Disorders/immunology , Helicobacter Infections/immunology , Molecular Mimicry/immunology , Neuropeptides/immunology , Adolescent , Candida albicans , Carrier State/immunology , Child , Child, Preschool , Cross Reactions , Female , Ghrelin/immunology , Helicobacter pylori , Humans , Leptin/immunology , Male , Orexins/immunology , alpha-MSH/immunology
19.
Alcohol Clin Exp Res ; 39(1): 21-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25623402

ABSTRACT

BACKGROUND: Orexin (OX) neurons originating in the lateral hypothalamus (LH) are ideally positioned to modulate reward processing as they form connections with several key brain regions known to be involved in the reward pathway. Consistent with these findings, a growing number of studies have implicated the OX system in modulating the rewarding properties of several drugs of abuse, including ethanol (EtOH). However, the role of the OX system in excessive binge-like EtOH intake remains relatively unexplored. Here, we assessed changes in OX immunoreactivity (IR) in the hypothalamus following repeated cycles of binge-like EtOH drinking and assessed the participation of the OX-1 receptor (OX1R) in binge-like EtOH consumption. METHODS: The drinking-in-the-dark (DID) paradigm was used to model binge-like EtOH drinking in male C57BL/6J mice. In the first experiment, mice experienced 1 or 3 cycles of binge-like EtOH or sucrose drinking with DID procedures to assess changes in OX IR in distinct subregions of the hypothalamus. Subsequent experiments examined binge-like EtOH and saccharin drinking following peripheral injections of 0.0, 5.0, or 10.0 mg/kg SB-334867 (SB), a selective OX1R antagonist. Finally, mice were given peripheral injections of SB and open-field locomotor activity was measured. RESULTS: Relative to water drinking controls, binge-like consumption of EtOH and sucrose resulted in a marked reduction in OX IR in the LH. Inhibition of the OX1R via SB blunted EtOH and saccharin drinking, but did not alter open-field locomotor activity. CONCLUSIONS: Our observed reduction in OX IR in the LH indicates that the OX system in engaged during binge-like consumption of EtOH and sucrose. The observation that inhibition of the OX1R signaling blunted binge-like EtOH, and saccharin drinking suggests that reward-related OX circuits originating in the LH participate in the consumption of salient reinforcers regardless of calories.


Subject(s)
Binge Drinking/drug therapy , Binge Drinking/metabolism , Hypothalamic Area, Lateral/drug effects , Orexin Receptors/metabolism , Orexins/metabolism , Reinforcement, Psychology , Saccharin/pharmacology , Sucrose/pharmacology , Animals , Benzoxazoles/pharmacology , Benzoxazoles/therapeutic use , Dose-Response Relationship, Drug , Hypothalamic Area, Lateral/metabolism , Male , Mice , Motor Activity/drug effects , Naphthyridines , Orexins/analysis , Orexins/immunology , Sucrose/antagonists & inhibitors , Urea/analogs & derivatives , Urea/pharmacology , Urea/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...