Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.114
Filter
1.
Front Immunol ; 15: 1458967, 2024.
Article in English | MEDLINE | ID: mdl-39351233

ABSTRACT

Varicella-zoster virus (VZV) encephalitis and meningitis are potential central nervous system (CNS) complications following primary VZV infection or reactivation. With Type-I interferon (IFN) signalling being an important first line cellular defence mechanism against VZV infection by the peripheral tissues, we here investigated the triggering of innate immune responses in a human neural-like environment. For this, we established and characterised 5-month matured hiPSC-derived neurospheroids (NSPHs) containing neurons and astrocytes. Subsequently, NSPHs were infected with reporter strains of VZV (VZVeGFP-ORF23) or Sendai virus (SeVeGFP), with the latter serving as an immune-activating positive control. Live cell and immunocytochemical analyses demonstrated VZVeGFP-ORF23 infection throughout the NSPHs, while SeVeGFP infection was limited to the outer NSPH border. Next, NanoString digital transcriptomics revealed that SeVeGFP-infected NSPHs activated a clear Type-I IFN response, while this was not the case in VZVeGFP-ORF23-infected NSPHs. Moreover, the latter displayed a strong suppression of genes related to IFN signalling and antigen presentation, as further demonstrated by suppression of IL-6 and CXCL10 production, failure to upregulate Type-I IFN activated anti-viral proteins (Mx1, IFIT2 and ISG15), as well as reduced expression of CD74, a key-protein in the MHC class II antigen presentation pathway. Finally, even though VZVeGFP-ORF23-infection seems to be immunologically ignored in NSPHs, its presence does result in the formation of stress granules upon long-term infection, as well as disruption of cellular integrity within the infected NSPHs. Concluding, in this study we demonstrate that 5-month matured hiPSC-derived NSPHs display functional innate immune reactivity towards SeV infection, and have the capacity to recapitulate the strong immune evasive behaviour towards VZV.


Subject(s)
Herpesvirus 3, Human , Induced Pluripotent Stem Cells , Humans , Herpesvirus 3, Human/immunology , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/virology , Immunity, Innate , Neurons/immunology , Neurons/virology , Varicella Zoster Virus Infection/immunology , Varicella Zoster Virus Infection/virology , Cells, Cultured , Interferon Type I/metabolism , Interferon Type I/immunology , Immune Evasion , Cytokines/metabolism , Cytokines/immunology , Astrocytes/immunology , Astrocytes/virology , Astrocytes/metabolism , Signal Transduction/immunology
2.
J Neuroinflammation ; 21(1): 252, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39375720

ABSTRACT

BACKGROUND: Neuroinflammation reportedly plays a critical role in the pathogenesis of sepsis-associated encephalopathy (SAE). We previously reported that circulating plasma extracellular vesicles (EVs) from septic mice are proinflammatory. In the current study, we tested the role of sepsis plasma EVs in neuroinflammation. METHODS: To track EVs in cells and tissues, HEK293T cell-derived EVs were labeled with the fluorescent dye PKH26. Cecal ligation and puncture (CLP) was conducted to model polymicrobial sepsis in mice. Plasma EVs were isolated by ultracentrifugation and their role in promoting neuronal inflammation was tested following intracerebroventricular (ICV) injection. miRNA inhibitors (anti-miR-146a, -122, -34a, and -145a) were applied to determine the effects of EV cargo miRNAs in the brain. A cytokine array was performed to profile microglia-released protein mediators. TLR7- or MyD88-knockout (KO) mice were utilized to determine the underlying mechanism of EVs-mediated neuroinflammation. RESULTS: We observed the uptake of fluorescent PKH26-EVs inside the cell bodies of both microglia and neurons. Sepsis plasma EVs led to a dose-dependent cytokine release in cultured microglia, which was partially attenuated by miRNA inhibitors against the target miRNAs and in TLR7-KO cells. When administered via the ICV, sepsis plasma EVs resulted in a marked increase in the accumulation of innate immune cells, including monocyte and neutrophil and cytokine gene expression, in the brain. Although sepsis plasma EVs had no direct effect on cytokine production or neuronal injury in vitro, the conditioned media (CM) of microglia treated with sepsis plasma EVs induced neuronal cell death as evidenced by increased caspase-3 cleavage and Annexin-V staining. Cytokine arrays and bioinformatics analysis of the microglial CM revealed multiple cytokines/chemokines and other factors functionally linked to leukocyte chemotaxis and migration, TLR signaling, and neuronal death. Moreover, sepsis plasma EV-induced brain inflammation in vivo was significantly dependent on MyD88. CONCLUSIONS: Circulating plasma EVs in septic mice cause a microglial proinflammatory response in vitro and a brain innate immune response in vivo, some of which are in part mediated by TLR7 in vitro and MyD88 signaling in vivo. These findings highlight the importance of circulating EVs in brain inflammation during sepsis.


Subject(s)
Brain , Extracellular Vesicles , Immunity, Innate , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs , Neurons , Sepsis , Signal Transduction , Animals , Extracellular Vesicles/metabolism , Mice , MicroRNAs/metabolism , Sepsis/immunology , Sepsis/metabolism , Sepsis/pathology , Humans , Signal Transduction/physiology , Neurons/metabolism , Neurons/immunology , Brain/metabolism , Brain/immunology , Brain/pathology , HEK293 Cells , Male , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Microglia/metabolism , Microglia/immunology , Inflammation/metabolism , Inflammation/immunology , Inflammation/pathology , Membrane Glycoproteins , Toll-Like Receptor 7
3.
Front Immunol ; 15: 1445867, 2024.
Article in English | MEDLINE | ID: mdl-39253085

ABSTRACT

Mast cells serve as crucial effector cells within the innate immune system and are predominantly localized in the skin, airways, gastrointestinal tract, urinary and reproductive tracts, as well as in the brain. Under physiological conditions, brain-resident mast cells secrete a diverse array of neuro-regulatory mediators to actively participate in neuroprotection. Meanwhile, as the primary source of molecules causing brain inflammation, mast cells also function as the "first responders" in brain injury. They interact with neuroglial cells and neurons to facilitate the release of numerous inflammatory mediators, proteases, and reactive oxygen species. This process initiates and amplifies immune-inflammatory responses in the brain, thereby contributing to the regulation of neuroinflammation and blood-brain barrier permeability. This article provides a comprehensive overview of the potential mechanisms through which mast cells in the brain may modulate neuroprotection and their pathological implications in various neurological disorders. It is our contention that the inhibition of mast cell activation in brain disorders could represent a novel avenue for therapeutic breakthroughs.


Subject(s)
Mast Cells , Humans , Mast Cells/immunology , Mast Cells/metabolism , Animals , Brain/immunology , Brain/pathology , Brain/metabolism , Brain Diseases/immunology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Neurons/immunology , Neurons/metabolism , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology
4.
Microbiol Immunol ; 68(10): 348-358, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39239735

ABSTRACT

Botulism is a deadly neuroparalytic condition caused by the botulinum neurotoxin (BoNT) produced by Clostridium botulinum and related species. Toxin-neutralizing antibodies are the most effective treatments for BoNT intoxication. We generated human monoclonal antibodies neutralizing type B botulinum neurotoxin (BoNT/B), designated M2 and M4. The combination of these antibodies exhibited a strong neutralizing effect against BoNT/B toxicity. In this study, we analyzed the mechanisms of action of these antibodies in vitro. M4 binds to the C-terminus of the heavy chain (the receptor-binding domain) and inhibits BoNT/B binding to neuronal PC12 cells. Although M2 recognized the light (L) chain (the metalloprotease domain), it did not inhibit substrate (VAMP2) cleavage in the cleavage assay. M2 increased the surface localization of BoNT/B in PC12 cells at a later time point, suggesting that M2 inhibits the translocation of the L chain from synaptic vesicles to the cytosol. These results indicate that M2 and M4 inhibit the different processes of BoNT/B individually and that multistep inhibition is important for the synergistic effect of the combination of monoclonal antibodies. Our findings may facilitate the development of effective therapeutic antibodies against BoNTs.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , PC12 Cells , Animals , Rats , Antibodies, Monoclonal/immunology , Humans , Antibodies, Neutralizing/immunology , Botulinum Toxins, Type A/immunology , Botulism/immunology , Botulinum Toxins/immunology , Botulinum Toxins/antagonists & inhibitors , Neurons/immunology , Neurons/drug effects , Clostridium botulinum/immunology , Vesicle-Associated Membrane Protein 2/immunology , Vesicle-Associated Membrane Protein 2/metabolism , Protein Binding , Synaptic Vesicles/metabolism , Synaptic Vesicles/immunology
5.
Trends Neurosci ; 47(10): 753-765, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39242246

ABSTRACT

A major aim of neuroscience is to identify and model the functional properties of neural cells whose dysfunction underlie neuropsychiatric illness. In this article, we propose that human-derived monoclonal autoantibodies (HD-mAbs) are well positioned to selectively target and manipulate neural subpopulations as defined by their protein expression; that is, cellular proteotypes. Recent technical advances allow for efficient cloning of autoantibodies from neuropsychiatric patients. These HD-mAbs can be introduced into animal models to gain biological and pathobiological insights about neural proteotypes of interest. Protein engineering can be used to modify, enhance, silence, or confer new functional properties to native HD-mAbs, thereby enhancing their versatility. Finally, we discuss the challenges and limitations confronting HD-mAbs as experimental research tools for neuroscience.


Subject(s)
Antibodies, Monoclonal , Autoantibodies , Brain , Humans , Autoantibodies/immunology , Animals , Brain/metabolism , Brain/immunology , Antibodies, Monoclonal/immunology , Neurons/metabolism , Neurons/immunology
6.
J Neurophysiol ; 132(3): 929-942, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39163023

ABSTRACT

Alzheimer's disease (AD) was described more than a century ago. However, there are still no effective approaches to its treatment, which may suggest that the search for the cure is not being conducted in the most productive direction. AD begins as selective impairments of declarative memory with no deficits in other cognitive functions. Therefore, understanding of the AD pathogenesis has to include the understanding of this selectivity. Currently, the main efforts aimed at prevention and treatment of AD are based on the dominating hypothesis for the AD pathogenesis: the amyloid hypothesis. But this hypothesis does not explain selective memory impairments since ß-amyloid accumulates extracellularly and should be toxic to all types of cerebral neurons, not only to "memory engram neurons." To explain selective memory impairment, I propose the autoimmune hypothesis of AD, based on the analysis of risk factors for AD and molecular mechanisms of memory formation. Memory formation is associated with epigenetic modifications of chromatin in memory engram neurons and, therefore, might be accompanied by the expression of memory-specific proteins recognized by the adaptive immune system as "non-self" antigens. Normally, the brain is protected by the blood-brain barrier (BBB). All risk factors for AD provoke BBB disruptions, possibly leading to an autoimmune reaction against memory engram neurons. This reaction would make them selectively sensitive to tauopathy. If this hypothesis is confirmed, the strategies for AD prevention and treatment would be radically changed.


Subject(s)
Alzheimer Disease , Alzheimer Disease/immunology , Humans , Animals , Autoimmunity , Blood-Brain Barrier , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/immunology , Neurons/metabolism , Neurons/immunology
7.
Lupus ; 33(11): 1227-1234, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39167163

ABSTRACT

INTRODUCTION: Anti-neuronal antibodies target antigens produced by tumour cells and cells of nervous system. These antibodies are formed as a result of autoimmune response elicited by the underlying malignancy, when proteins restricted to immune privileged neurons are presented by the tumour. Previous studies have shown presence of anti-neuronal antibodies in systemic lupus erythematosus and neuropsychiatric lupus (NPSLE) but information on individual antibodies and their pathogenic role is lacking. AIMS/OBJECTIVE: To assess the frequency of anti-neuronal antibodies in our neuropsychiatric lupus cohort and to assess any significant association with specific neurological syndrome and to see if the antibodies were more likely to occur in active rather than inactive neuropsychiatric lupus. METHODOLOGY: This cross-sectional study was conducted in our center from 2019 to 2022. Neuropsychiatric manifestations were defined according to 1999 American College of Rheumatology (ACR) nomenclature and case definitions for neuropsychiatric lupus. Samples were taken from active or inactive NPSLE patients with their informed consent. Testing was done on an anti-neuronal antigen panel which consisted of [Amphiphysin, CV2, GAD 65, PNMA2 (Ma-2/Ta), Ri, Yo, Hu, recoverin, SOX1, titin, Zic, Tr)] by semi-quantitative Line immune assay. Association between the categorical variables and antibody positivity group was established using chi-square/Fisher's exact test as appropriate. RESULTS: 65 patients were recruited, of which 23 (35%) patients had active NPSLE at the time of sample collection. Anti-neuronal antibodies were positive in 13/65 (20%) patients with anti-Gad 65 antibodies having the highest frequency (6.2%) followed by anti CV 2 (3.1%), anti Sox1 (3.1%), anti Amphiphysin (3.1%) anti recoverin (1.5%), anti Yo (1.5%) and anti Zic (1.5%). The panel of anti-neuronal antibodies did not show any specific association with NPSLE features.However, an interesting finding was that, patients with active disease had higher odds of having anti-neuronal antibodies with an OR = 10 (95% CI:2.38 -42) (p < 0.001) than inactive disease. CONCLUSION: Anti-neuronal antibodies were more likely to be positive in active neuropsychiatric lupus patients, and these antibodies which are commonly used to diagnose paraneoplastic syndromes may have a potential role in the diagnosis of NPSLE.


Subject(s)
Autoantibodies , Lupus Vasculitis, Central Nervous System , Humans , Cross-Sectional Studies , Autoantibodies/immunology , Autoantibodies/blood , Female , Male , Lupus Vasculitis, Central Nervous System/immunology , Adult , Middle Aged , Paraneoplastic Syndromes, Nervous System/immunology , Neurons/immunology , Young Adult , Aged
8.
J Neuroinflammation ; 21(1): 189, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095837

ABSTRACT

BACKGROUND: Infection with the protozoan parasite Toxoplasma gondii leads to the formation of lifelong cysts in neurons that can have devastating consequences in the immunocompromised. In the immunocompetent individual, anti-parasitic effector mechanisms and a balanced immune response characterized by pro- and anti-inflammatory cytokine production establishes an asymptomatic infection that rarely leads to neurological symptoms. Several mechanisms are known to play a role in this successful immune response in the brain including T cell production of IFNγ and IL-10 and the involvement of CNS resident cells. This limitation of clinical neuropathology during chronic infection suggests a balance between immune response and neuroprotective mechanisms that collectively prevent clinical manifestations of disease. However, how these two vital mechanisms of protection interact during chronic Toxoplasma infection remains poorly understood. MAIN TEXT: This study demonstrates a previously undescribed connection between innate neutrophils found chronically in the brain, termed "chronic brain neutrophils" (CBNeuts), and neuroprotective mechanisms during Toxoplasma infection. Lack of CBNeuts during chronic infection, accomplished via systemic neutrophil depletion, led to enhanced infection and deleterious effects on neuronal regeneration and repair mechanisms in the brain. Phenotypic and transcriptomic analysis of CBNeuts identified them as distinct from peripheral neutrophils and revealed two main subsets of CBNeuts that display heterogeneity towards both classical effector and neuroprotective functions in an age-dependent manner. Further phenotypic profiling defined expression of the neuroprotective molecules NRG-1 andErbB4 by these cells, and the importance of this signaling pathway during chronic infection was demonstrated via NRG-1 treatment studies. CONCLUSIONS: In conclusion, this work identifies CBNeuts as a heterogenous population geared towards both classical immune responses and neuroprotection during chronic Toxoplasma infection and provides the foundation for future mechanistic studies of these cells.


Subject(s)
Neutrophils , Toxoplasmosis , Animals , Neutrophils/immunology , Neutrophils/metabolism , Mice , Toxoplasmosis/immunology , Mice, Inbred C57BL , Neurons/metabolism , Neurons/immunology , Toxoplasma/immunology , Female , Neuroprotection/physiology , Male , Brain/immunology , Brain/pathology , Brain/parasitology
9.
Microb Pathog ; 195: 106896, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39208957

ABSTRACT

BACKGROUND: Bovine herpesvirus 1 (BoHV-1) is a major pathogen that affects the global bovine population, primarily inducing respiratory and reproductive disorders. Its ability to establish latent infections in neuronal cells and to reactivate under certain conditions poses a continual threat to uninfected hosts. In this study, we aimed to analyze the replication characteristics of BoHV-1 in neuronal cells, as well as the effects of viral replication on host cell immunity and physiology. METHODS: Using the Neuro-2a neuronal-origin cell line as a model, we explored the dynamics of BoHV-1 replication and analyzed differential gene expression profiles post-BoHV-1 infection using high-throughput RNA sequencing. RESULTS: BoHV-1 demonstrated restricted replication in Neuro-2a cells. BoHV-1 induced apoptotic pathways and enhanced the transcription of interferon-stimulated genes and interferon regulatory factors while suppressing the complement cascade in Neuro-2a cells. CONCLUSIONS: Different from BoHV-1 infection in other non-highly differentiated somatic cells result in viral dominance, BoHV-1 regulated the innate immune response in neuronal cells formed a "virus-nerve cell" relative equilibrium state, which may account for the restricted replication of BoHV-1 in neuronal cells, leading to a latent infection. These findings provide a foundation for further research into the mechanism underlying BoHV-1-induced latent infection in nerve cells.


Subject(s)
Gene Expression Profiling , Herpesvirus 1, Bovine , Immunity, Innate , Neurons , Virus Replication , Herpesvirus 1, Bovine/immunology , Herpesvirus 1, Bovine/genetics , Herpesvirus 1, Bovine/physiology , Animals , Cattle , Neurons/virology , Neurons/immunology , Cell Line , Mice , Herpesviridae Infections/virology , Herpesviridae Infections/immunology , Herpesviridae Infections/veterinary , Apoptosis , Transcriptome , Virus Latency , Host-Pathogen Interactions/immunology , Cattle Diseases/virology , Cattle Diseases/immunology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , High-Throughput Nucleotide Sequencing
10.
Front Immunol ; 15: 1419712, 2024.
Article in English | MEDLINE | ID: mdl-39114659

ABSTRACT

Background and objectives: Up to 46% of patients with presumed autoimmune limbic encephalitis are seronegative for all currently known central nervous system (CNS) antigens. We developed a cell-based assay (CBA) to screen for novel neural antibodies in serum and cerebrospinal fluid (CSF) using neurons and astrocytes derived from human-induced pluripotent stem cells (hiPSCs). Methods: Human iPSC-derived astrocytes or neurons were incubated with serum/CSF from 99 patients [42 with inflammatory neurological diseases (IND) and 57 with non-IND (NIND)]. The IND group included 11 patients with previously established neural antibodies, six with seronegative neuromyelitis optica spectrum disorder (NMOSD), 12 with suspected autoimmune encephalitis/paraneoplastic syndrome (AIE/PNS), and 13 with other IND (OIND). IgG binding to fixed CNS cells was detected using fluorescently-labeled antibodies and analyzed through automated fluorescence measures. IgG neuronal/astrocyte reactivity was further analyzed by flow cytometry. Peripheral blood mononuclear cells (PBMCs) were used as CNS-irrelevant control target cells. Reactivity profile was defined as positive using a Robust regression and Outlier removal test with a false discovery rate at 10% following each individual readout. Results: Using our CBA, we detected antibodies recognizing hiPSC-derived neural cells in 19/99 subjects. Antibodies bound specifically to astrocytes in nine cases, to neurons in eight cases, and to both cell types in two cases, as confirmed by microscopy single-cell analyses. Highlighting the significance of our comprehensive 96-well CBA assay, neural-specific antibody binding was more frequent in IND (15 of 42) than in NIND patients (4 of 57) (Fisher's exact test, p = 0.0005). Two of four AQP4+ NMO and four of seven definite AIE/PNS with intracellular-reactive antibodies [1 GFAP astrocytopathy, 2 Hu+, 1 Ri+ AIE/PNS)], as identified in diagnostic laboratories, were also positive with our CBA. Most interestingly, we showed antibody-reactivity in two of six seronegative NMOSD, six of 12 probable AIE/PNS, and one of 13 OIND. Flow cytometry using hiPSC-derived CNS cells or PBMC-detected antibody binding in 13 versus zero patients, respectively, establishing the specificity of the detected antibodies for neural tissue. Conclusion: Our unique hiPSC-based CBA allows for the testing of novel neuron-/astrocyte-reactive antibodies in patients with suspected immune-mediated neurological syndromes, and negative testing in established routine laboratories, opening new perspectives in establishing a diagnosis of such complex diseases.


Subject(s)
Astrocytes , Autoantibodies , Immunoglobulin G , Induced Pluripotent Stem Cells , Neurons , Humans , Astrocytes/immunology , Astrocytes/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/blood , Neurons/immunology , Neurons/metabolism , Induced Pluripotent Stem Cells/immunology , Male , Female , Middle Aged , Autoantibodies/immunology , Autoantibodies/blood , Adult , Aged , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/diagnosis , Young Adult , Nervous System Diseases/immunology , Nervous System Diseases/diagnosis
11.
Transl Psychiatry ; 14(1): 289, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39009558

ABSTRACT

Prenatal exposure to infections is a risk factor for neurodevelopmental disorders in offspring, and alterations in mitochondrial function are discussed as a potential underlying factor. Here, using a mouse model of viral-like maternal immune activation (MIA) based on poly(I:C) (POL) treatment at gestational day (GD) 12, we show that adult offspring exhibit behavioral deficits, such as reduced levels of social interaction. In addition, we found increased nicotinamidadenindinucleotid (NADH)- and succinate-linked mitochondrial respiration and maximal electron transfer capacity in the prefrontal cortex (PFC) and in the amygdala (AMY) of males and females. The increase in respiratory capacity resulted from an increase in mitochondrial mass in neurons (as measured by complex IV activity and transcript expression), presumably to compensate for a reduction in mitochondrion-specific respiration. Moreover, in the PFC of control (CON) male offspring a higher excess capacity compared to females was observed, which was significantly reduced in the POL-exposed male offspring, and, along with a higher leak respiration, resulted in a lower mitochondrial coupling efficiency. Transcript expression of the uncoupling proteins (UCP4 and UCP5) showed a reduction in the PFC of POL male mice, suggesting mitochondrial dysfunction. In addition, in the PFC of CON females, a higher expression of the antioxidant enzyme superoxide dismutase (SOD1) was observed, suggesting a higher antioxidant capacity as compared to males. Finally, transcripts analysis of genes involved in mitochondrial biogenesis and dynamics showed reduced expression of fission/fusion transcripts in PFC of POL offspring of both sexes. In conclusion, we show that MIA causes alterations in neuronal mitochondrial function and mass in the PFC and AMY of adult offspring with some effects differing between males and females.


Subject(s)
Mitochondria , Prefrontal Cortex , Prenatal Exposure Delayed Effects , Animals , Female , Prenatal Exposure Delayed Effects/immunology , Pregnancy , Mitochondria/metabolism , Mice , Male , Prefrontal Cortex/metabolism , Prefrontal Cortex/immunology , Poly I-C/pharmacology , Disease Models, Animal , Brain/immunology , Brain/metabolism , Amygdala/metabolism , Amygdala/immunology , Behavior, Animal , Mice, Inbred C57BL , Neurons/metabolism , Neurons/immunology
12.
Transl Psychiatry ; 14(1): 306, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39054336

ABSTRACT

Studies in adults have linked stress-related activation of the immune system to the manifestation of psychiatric conditions. Using a translational design, this study aimed to examine the impact of social stress on immune activity in adolescents and on neuronal activity in a preclinical mouse model. Participants were 31 adolescents (ages 12-19), including 25 with mood and anxiety symptoms. Whole-blood samples were collected before and after the Trier Social Stress Test (TSST), a stress-inducing public speaking task, then cultured for 6 hours in the presence and absence of the inflammatory endotoxin lipopolysaccharide (LPS). Effects of TSST and LPS on 41 immune biomarkers were examined using repeated-measures analysis of variance. Separately, juvenile (8-week-old) male mice were non-stressed or exposed to reminder social defeat then intraperitoneally injected with saline or LPS (n = 6/group). Brains were perfused and collected for immunohistochemistry and confocal microscopy at 0, 1, 6, and 24 hours post-injection. The activity was determined by the density of cFos-positive neurons in the paraventricular hypothalamus, paraventricular thalamus, and basolateral amygdala, regions known to show sustained activation to immunological challenge. Analyses in the adolescent study indicated a strong effect of LPS but no effects of TSST or TSST×LPS interaction on immune biomarkers. Similarly, reminder social defeat did not induce sustained neuronal activity changes comparable to LPS immunological challenge in juvenile mice. Our convergent findings across species suggest that the acute immune response to stress documented in adults is not present in youth. Thus, aging and chronicity effects may play an important role in the inflammatory response to acute psychosocial stress.


Subject(s)
Lipopolysaccharides , Stress, Psychological , Animals , Stress, Psychological/immunology , Stress, Psychological/physiopathology , Male , Humans , Adolescent , Mice , Lipopolysaccharides/pharmacology , Child , Female , Young Adult , Neurons/immunology , Social Defeat , Brain/immunology , Disease Models, Animal , Mice, Inbred C57BL , Amygdala/immunology , Amygdala/physiopathology
13.
Front Immunol ; 15: 1388998, 2024.
Article in English | MEDLINE | ID: mdl-38863705

ABSTRACT

Background: Neuronal nicotinic acetylcholine receptors (nAChRs) are abundant in the central nervous system (CNS), playing critical roles in brain function. Antigenicity of nAChRs has been well demonstrated with antibodies to ganglionic AChR subtypes (i.e., subunit α3 of α3ß4-nAChR) and muscle AChR autoantibodies, thus making nAChRs candidate autoantigens in autoimmune CNS disorders. Antibodies to several membrane receptors, like NMDAR, have been identified in autoimmune encephalitis syndromes (AES), but many AES patients have yet to be unidentified for autoantibodies. This study aimed to develop of a cell-based assay (CBA) that selectively detects potentially pathogenic antibodies to subunits of the major nAChR subtypes (α4ß2- and α7-nAChRs) and its use for the identification of such antibodies in "orphan" AES cases. Methods: The study involved screening of sera derived from 1752 patients from Greece, Turkey and Italy, who requested testing for AES-associated antibodies, and from 1203 "control" patients with other neuropsychiatric diseases, from the same countries or from Germany. A sensitive live-CBA with α4ß2-or α7-nAChR-transfected cells was developed to detect antibodies against extracellular domains of nAChR major subunits. Flow cytometry (FACS) was performed to confirm the CBA findings and indirect immunohistochemistry (IHC) to investigate serum autoantibodies' binding to rat brain tissue. Results: Three patients were found to be positive for serum antibodies against nAChR α4 subunit by CBA and the presence of the specific antibodies was quantitatively confirmed by FACS. We detected specific binding of patient-derived serum anti-nAChR α4 subunit antibodies to rat cerebellum and hippocampus tissue. No serum antibodies bound to the α7-nAChR-transfected or control-transfected cells, and no control serum antibodies bound to the transfected cells. All patients positive for serum anti-nAChRs α4 subunit antibodies were negative for other AES-associated antibodies. All three of the anti-nAChR α4 subunit serum antibody-positive patients fall into the AES spectrum, with one having Rasmussen encephalitis, another autoimmune meningoencephalomyelitis and another being diagnosed with possible autoimmune encephalitis. Conclusion: This study lends credence to the hypothesis that the major nAChR subunits are autoimmune targets in some cases of AES and establishes a sensitive live-CBA for the identification of such patients.


Subject(s)
Autoantibodies , Central Nervous System Diseases , Neurons , Receptors, Nicotinic , Adolescent , Adult , Aged , Animals , Female , Humans , Male , Middle Aged , Rats , Young Adult , Autoantibodies/immunology , Autoantibodies/blood , Central Nervous System Diseases/immunology , Encephalitis/immunology , Neurons/immunology , Receptors, Nicotinic/immunology
14.
Int J Mol Sci ; 25(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38891900

ABSTRACT

Toll-like receptors (TLRs) are among the main components of the innate immune system. They can detect conserved structures in microorganisms and molecules associated with stress and cellular damage. TLRs are expressed in resident immune cells and both neurons and glial cells of the nervous system. Increasing evidence is emerging on the participation of TLRs not only in the immune response but also in processes of the nervous system, such as neurogenesis and cognition. Below, we present a review of the literature that evaluates the expression and role of TLRs in processes such as neurodevelopment, behavior, cognition, infection, neuroinflammation, and neurodegeneration.


Subject(s)
Nervous System , Neurogenesis , Toll-Like Receptors , Humans , Toll-Like Receptors/metabolism , Animals , Nervous System/metabolism , Nervous System/immunology , Immunity, Innate , Neurons/metabolism , Neurons/immunology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/immunology , Signal Transduction
15.
Annu Rev Immunol ; 42(1): 489-519, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38941607

ABSTRACT

Recent advances have contributed to a mechanistic understanding of neuroimmune interactions in the intestine and revealed an essential role of this cross talk for gut homeostasis and modulation of inflammatory and infectious intestinal diseases. In this review, we describe the innervation of the intestine by intrinsic and extrinsic neurons and then focus on the bidirectional communication between neurons and immune cells. First, we highlight the contribution of neuronal subtypes to the development of colitis and discuss the different immune and epithelial cell types that are regulated by neurons via the release of neuropeptides and neurotransmitters. Next, we review the role of intestinal inflammation in the development of visceral hypersensitivity and summarize how inflammatory mediators induce peripheral and central sensitization of gut-innervating sensory neurons. Finally, we outline the importance of immune cells and gut microbiota for the survival and function of different neuronal populations at homeostasis and during bacterial and helminth infection.


Subject(s)
Neuroimmunomodulation , Humans , Animals , Intestines/immunology , Homeostasis , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Neurons/metabolism , Neurons/immunology , Neuropeptides/metabolism , Enteric Nervous System/immunology , Enteric Nervous System/metabolism
16.
J Clin Immunol ; 44(6): 140, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38829425

ABSTRACT

Autoimmune polyendocrine syndrome type 1 (APS-1) is a rare monogenic disease caused by mutations in the autoimmune regulator gene. Although the disease-associated autoantibodies mostly target endocrine organs, autoantibodies from patients with APS-1 bind also to rat brain structures. The patients often have GAD65-antibodies, that can cause autoimmune encephalitis. However, neurological manifestations of APS-1 have not been systematically explored. We conducted a retrospective chart review on 44 Finnish patients with APS-1 (median age 38 years, 61% females) and collected all their neurological diagnoses. To assess the prevalence of serum antineuronal antibodies in APS-1, serum samples of 24 patients (median age 36 years, 63% females) were analyzed using a fixed cell-based assay. Of the 44 APS-1 patients, 10 (23%) had also received a diagnosis of a neurological disease. Of these neurological comorbidities, migraine (n = 7; 16%), central nervous system infections (n = 3; 7%), and epilepsy (n = 2; 5%) were the most prevalent. Other diagnoses recorded for single patients were axonal sensorimotor polyneuropathy, essential tremor, idiopathic intracranial hypertension, ischemic stroke, and trigeminal neuralgia. Serum antineuronal antibodies were detected in 42% of patients tested (10/24, 50% females, median age 42 years), GAD65 antibodies being the most common finding. Antibodies against glycine and aquaporin 4 were found in low titers. In four patients, relatively high titers of GAD65 antibodies without coexisting type 1 diabetes were found, but none presented with GAD65-encephalitis. Our study suggests an association between APS-1 and neurological disorders, the mechanisms of which are to be further investigated.


Subject(s)
Autoantibodies , Polyendocrinopathies, Autoimmune , Humans , Polyendocrinopathies, Autoimmune/immunology , Polyendocrinopathies, Autoimmune/epidemiology , Polyendocrinopathies, Autoimmune/blood , Female , Male , Adult , Autoantibodies/blood , Autoantibodies/immunology , Middle Aged , Finland/epidemiology , Prevalence , Retrospective Studies , Cohort Studies , Young Adult , Nervous System Diseases/immunology , Nervous System Diseases/epidemiology , Nervous System Diseases/etiology , Neurons/immunology , Adolescent , Glutamate Decarboxylase/immunology , Aged
18.
J Autoimmun ; 147: 103256, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38788538

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder associated with the loss of dopaminergic neurons and neuroinflammation. Recent studies have identified a role of T cells in the pathogenesis of PD. Additionally, these studies suggested that α-synuclein (α-Syn) is related to abnormal T-cell responses and may act as an epitope and trigger autoimmune T-cell responses. However, it is unclear whether the α-Syn-mediated autoimmune response occurs and whether it is related to neuronal cell death and glial cell activation. In this study, we investigated the autoimmune T-cell response induced by α-Syn peptides and evaluated the neurotoxic effect of the α-Syn peptide-mediated autoimmune response. The immunization of mice with α-Syn peptides resulted in enhanced autoimmune responses, such as the peptide recall response, polarization toward Th1/Th17 cells, and regulatory T cell imbalance. Furthermore, the α-Syn autoimmune response led to the death of primary neurons cocultured with splenocytes. Treatment with conditioned media from α-Syn peptide-immunized splenocytes induced microglia and toxic A1-type astrocyte activation. Taken together, our results provide evidence of the potential role of the α-Syn-initiated autoimmune response and its contribution to neuronal cell death and glial cell activation.


Subject(s)
Autoimmunity , Cell Death , Neurons , alpha-Synuclein , Animals , alpha-Synuclein/immunology , alpha-Synuclein/metabolism , Mice , Cell Death/drug effects , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Neuroglia/immunology , Neuroglia/metabolism , Neuroglia/drug effects , Parkinson Disease/immunology , Parkinson Disease/pathology , Parkinson Disease/metabolism , Mice, Inbred C57BL , Humans , Lymphocyte Activation/immunology , Lymphocyte Activation/drug effects , Peptides/immunology , Cells, Cultured , Female , T-Lymphocytes, Regulatory/immunology
19.
Cancer Cell ; 42(6): 936-938, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38821062

ABSTRACT

Cellular mechanisms mediating immunotherapy resistances are incompletely understood. In this issue, Li et al. reveal how breast cancer hijacks neuronal mechanisms of neuroprotection to shield itself from the immune system. Secretion of N-acetylaspartate impairs immune synapse formation in both neuroinflammation and breast cancer models, paving the way for novel therapeutic approaches.


Subject(s)
Breast Neoplasms , Neurons , Humans , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Female , Neurons/metabolism , Neurons/immunology , Immune System/immunology , Animals
20.
Front Immunol ; 15: 1353747, 2024.
Article in English | MEDLINE | ID: mdl-38751431

ABSTRACT

Pathogen avoidance behaviour has been observed across animal taxa as a vital host-microbe interaction mechanism. The nematode Caenorhabditis elegans has evolved multiple diverse mechanisms for pathogen avoidance under natural selection pressure. We summarise the current knowledge of the stimuli that trigger pathogen avoidance, including alterations in aerotaxis, intestinal bloating, and metabolites. We then survey the neural circuits involved in pathogen avoidance, transgenerational epigenetic inheritance of pathogen avoidance, signalling crosstalk between pathogen avoidance and innate immunity, and C. elegans avoidance of non-Pseudomonas bacteria. In this review, we highlight the latest advances in understanding host-microbe interactions and the gut-brain axis.


Subject(s)
Caenorhabditis elegans , Host-Pathogen Interactions , Immunity, Innate , Animals , Caenorhabditis elegans/immunology , Caenorhabditis elegans/microbiology , Host-Pathogen Interactions/immunology , Epigenesis, Genetic , Signal Transduction , Neurons/immunology , Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL