Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 447
Filter
2.
Int Immunopharmacol ; 113(Pt A): 109375, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36461592

ABSTRACT

BACKGROUND: Recent studies have uncovered that hyperuricemia (HUA) leads to cognitive deficits, which are accompanied by neuronal damage and neuroinflammation. Here, we aim to explore the role of methyltransferase-like 3 (METTL3) in HUA-mediated neuronal apoptosis and microglial inflammation. METHODS: A HUA mouse model was constructed. The spatial memory ability of the mice was assessed by the Morris water maze experiment (MWM), and neuronal apoptosis was analyzed by the TdT-mediated dUTP nick end labeling (TUNEL) assay. Besides, enzyme-linked immunosorbent assay (ELISA) was utilized to measure the contents of inflammatory factors (IL-1ß, IL-6, and TNF-α) and oxidative stress markers (MDA, SOD, and CAT) in the serum of mice. In vitro, the mouse hippocampal neuron (HT22) and microglia (BV2) were treated with uric acid (UA). Flow cytometry was applied to analyze HT22 and BV2 cell apoptosis, and ELISA was conducted to observe neuroinflammation and oxidative stress. In addition, the expression of MyD88, p-NF-κB, NF-κB, NLRP3, ASC and Caspase1 was determined by Western blot. RESULTS: METTL3 and miR-124-3p were down-regulated, while the MyD88-NF-κB pathway was activated in the HUA mouse model. UA treatment induced neuronal apoptosis in HT22 and stimulated microglial activation in BV2. Overexpressing METTL3 alleviated HT22 neuronal apoptosis and resisted the release of inflammatory cytokines and oxidative stress mediators in BV2 cells. METTL3 repressed MyD88-NF-κB and NLRP3-ASC-Caspase1 inflammasome. In addition, METTL3 overexpression enhanced miR-124-3p expression, while METTL3 knockdown aggravated HT22 cell apoptosis and BV2 cell overactivation. CONCLUSION: METTL3 improves neuronal apoptosis and microglial activation in the HUA model by choking the MyD88/NF-κB pathway and up-regulating miR-124-3p.


Subject(s)
Cognitive Dysfunction , Hyperuricemia , Inflammasomes , Methyltransferases , Animals , Mice , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/immunology , Caspase 1/genetics , Caspase 1/immunology , Cells, Cultured , Cognitive Dysfunction/etiology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/immunology , Disease Models, Animal , Hyperuricemia/complications , Hyperuricemia/genetics , Hyperuricemia/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Methyltransferases/genetics , Methyltransferases/immunology , MicroRNAs/genetics , MicroRNAs/immunology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Nervous System/drug effects , Nervous System/immunology , Nervous System/physiopathology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/immunology , NF-kappa B , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Uric Acid/administration & dosage , Uric Acid/adverse effects , Uric Acid/pharmacology
3.
J Neuroinflammation ; 19(1): 289, 2022 Dec 03.
Article in English | MEDLINE | ID: mdl-36463233

ABSTRACT

BACKGROUND: Neuroinflammation is one of the most important processes in secondary injury after traumatic brain injury (TBI). Triggering receptor expressed on myeloid cells 2 (TREM2) has been proven to exert neuroprotective effects in neurodegenerative diseases and stroke by modulating neuroinflammation, and promoting phagocytosis and cell survival. However, the role of TREM2 in TBI has not yet been elucidated. In this study, we are the first to use COG1410, an agonist of TREM2, to assess the effects of TREM2 activation in a murine TBI model. METHODS: Adult male wild-type (WT) C57BL/6 mice and adult male TREM2 KO mice were subjected to different treatments. TBI was established by the controlled cortical impact (CCI) method. COG1410 was delivered 1 h after CCI via tail vein injection. Western blot analysis, immunofluorescence, laser speckle contrast imaging (LSCI), neurological behaviour tests, brain electrophysiological monitoring, Evans blue assays, magnetic resonance imaging (MRI), and brain water content measurement were performed in this study. RESULTS: The expression of endogenous TREM2 peaked at 3 d after CCI, and it was mainly expressed on microglia and neurons. We found that COG1410 improved neurological functions within 3 d, as well as neurological functions and brain electrophysiological activity at 2 weeks after CCI. COG1410 exerted neuroprotective effects by inhibiting neutrophil infiltration and microglial activation, and suppressing neuroinflammation after CCI. In addition, COG1410 treatment alleviated blood brain barrier (BBB) disruption and brain oedema; furthermore, COG1410 promoted cerebral blood flow (CBF) recovery at traumatic injury sites after CCI. In addition, COG1410 suppressed neural apoptosis at 3 d after CCI. TREM2 activation upregulated p-Akt, p-CREB, BDNF, and Bcl-2 and suppressed TNF-α, IL-1ß, Bax, and cleaved caspase-3 at 3 d after CCI. Moreover, TREM2 knockout abolished the effects of COG1410 on vascular phenotypes and microglial states. Finally, the neuroprotective effects of COG1410 were suppressed by TREM2 depletion. CONCLUSIONS: Altogether, we are the first to demonstrate that TREM2 activation by COG1410 alleviated neural damage through activation of Akt/CREB/BDNF signalling axis in microglia after CCI. Finally, COG1410 treatment improved neurological behaviour and brain electrophysiological activity after CCI.


Subject(s)
Brain Injuries, Traumatic , Animals , Male , Mice , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/immunology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/immunology , Membrane Glycoproteins/agonists , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Receptors, Immunologic/agonists , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Disease Models, Animal , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/immunology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/immunology , Nervous System/drug effects , Nervous System/immunology
4.
Neuron ; 110(21): 3415-3417, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36327891

ABSTRACT

In an interview with Neuron, Gloria Choi gives us an overview of her work to characterize how neural circuits and behaviors are affected after immune challenges in various contexts. We also learn about her unique perspective in marrying the two disciplines of neuroscience and immunology.


Subject(s)
Allergy and Immunology , Nervous System , Neurosciences , Humans , Nervous System/immunology
5.
Cells ; 10(9)2021 09 15.
Article in English | MEDLINE | ID: mdl-34572077

ABSTRACT

Ischemic stroke is still among the leading causes of mortality and morbidity worldwide. Despite intensive advancements in medical sciences, the clinical options to treat ischemic stroke are limited to thrombectomy and thrombolysis using tissue plasminogen activator within a narrow time window after stroke. Current state of the art knowledge reveals the critical role of local and systemic inflammation after stroke that can be triggered by interactions taking place at the brain and immune system interface. Here, we discuss different cellular and molecular mechanisms through which brain-immune interactions can take place. Moreover, we discuss the evidence how the brain influence immune system through the release of brain derived antigens, damage-associated molecular patterns (DAMPs), cytokines, chemokines, upregulated adhesion molecules, through infiltration, activation and polarization of immune cells in the CNS. Furthermore, the emerging concept of stemness-induced cellular immunity in the context of neurodevelopment and brain disease, focusing on ischemic implications, is discussed. Finally, we discuss current evidence on brain-immune system interaction through the autonomic nervous system after ischemic stroke. All of these mechanisms represent potential pharmacological targets and promising future research directions for clinically relevant discoveries.


Subject(s)
Brain Ischemia/immunology , Brain/immunology , Neuroimmunomodulation , Stroke/immunology , Alarmins/metabolism , Blood-Brain Barrier/immunology , Brain Injuries/immunology , Brain Ischemia/therapy , Chemokines/metabolism , Cytokines/metabolism , Drug Delivery Systems , Humans , Inflammation/immunology , Nervous System/immunology , Stem Cells/immunology , Stroke/therapy
7.
J Neurochem ; 158(1): 8-13, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33993505

ABSTRACT

The rapidly expanding field of immunometabolism has highlighted an intricate association between the metabolic pathways that program cellular pro-inflammatory versus anti-inflammatory activity. This Special Issue on Neuroimmune Metabolism showcases a growing body of work characterizing the metabolic profiles of the major CNS-resident and peripheral immune cell players in neuroinflammation, neurodegeneration, and brain injury. The review articles address the roles of glycolytic, oxidative, and lipid metabolism that are associated with beneficial or detrimental properties in various neurological conditions, address unanswered questions in the field, and discuss promising avenues for future therapeutics. Cover Image for this issue: https://doi.org/10.1111/jnc.15069.


Subject(s)
Central Nervous System Diseases/immunology , Central Nervous System Diseases/metabolism , Immune System/metabolism , Nervous System/immunology , Nervous System/metabolism , Animals , Humans
8.
Metab Brain Dis ; 36(6): 1369-1379, 2021 08.
Article in English | MEDLINE | ID: mdl-33864573

ABSTRACT

Males are more likely to develop autism as a neurodevelopmental disorder than females, but the mechanisms underlying male susceptibility are not fully understood. In this paper, we used a well-characterized propionic acid (PPA) rodent model of autism to study sex differences in stress hormones, antioxidants' status, and the neuroimmune response that may contribute to the preponderance of autism in males. Sprague Dawley rats of both sexes were divided into a saline-treated group as controls and PPA-treated groups, receiving 250 mg/kg of PPA per day for three days. Animals' social behavior was examined using the three-chamber social test. Hormones (ACTH, corticosterone, melatonin, and oxytocin), oxidative stress biomarkers (glutathione, glutathione-S-transferase, and ascorbic acid), and cytokines (IL-6, IL-1α, IL-10, and IFNγ) were measured in the brain tissue of all the animals. The results showed a sex dimorphic social response to PPA treatment, where males were more susceptible to the PPA treatment and exhibited a significant reduction in social behavior with no effects observed in females. Also, sex differences were observed in the levels of hormones, antioxidants, and cytokines. Female rats showed significantly higher corticosterone and lower oxytocin, antioxidants, and cytokine levels than males. The PPA treatment later modulated these baseline differences. Our study indicates that the behavioral manifestation of autism in PPA-treated males and not females could be linked to neural biochemical differences between the sexes at baseline, which might play a protective role in females. Our results can contribute to early intervention strategies and treatments used to control autism, an increasingly prevalent disorder.


Subject(s)
Antioxidants/metabolism , Autistic Disorder/immunology , Autistic Disorder/metabolism , Hormones/blood , Nervous System/immunology , Social Interaction , Animals , Autistic Disorder/chemically induced , Brain Chemistry , Corticosterone/metabolism , Cytokines/metabolism , Female , Male , Oxytocin/metabolism , Propionates , Rats , Rats, Sprague-Dawley , Sex Characteristics
9.
Elife ; 102021 04 26.
Article in English | MEDLINE | ID: mdl-33900197

ABSTRACT

Detecting danger is key to the survival and success of all species. Animal nervous and immune systems cooperate to optimize danger detection. Preceding studies have highlighted the benefits of bringing neurons into the defense game, including regulation of immune responses, wound healing, pathogen control, and survival. Here, we summarize the body of knowledge in neuroimmune communication and assert that neuronal participation in the immune response is deeply beneficial in each step of combating infection, from inception to resolution. Despite the documented tight association between the immune and nervous systems in mammals or invertebrate model organisms, interdependence of these two systems is largely unexplored across metazoans. This review brings a phylogenetic perspective of the nervous and immune systems in the context of danger detection and advocates for the use of non-model organisms to diversify the field of neuroimmunology. We identify key taxa that are ripe for investigation due to the emergence of key evolutionary innovations in their immune and nervous systems. This novel perspective will help define the primordial principles that govern neuroimmune communication across taxa.


Subject(s)
Immune System/physiology , Nervous System/immunology , Neuroimmunomodulation , Animals , Biological Evolution , Phylogeny
10.
mBio ; 12(2)2021 03 30.
Article in English | MEDLINE | ID: mdl-33785621

ABSTRACT

Although Caenorhabditis elegans has been used as a model host for studying host-pathogen interactions for more than 20 years, the mechanisms by which it identifies pathogens are not well understood. This is largely due to its lack of most known pattern recognition receptors (PRRs) that recognize pathogen-derived molecules. Recent behavioral research in C. elegans indicates that its nervous system plays a major role in microbe sensing. With the increasing integration of neurobiology in immunological research, future studies may find that neuronal detection of pathogens is an integral part of C. elegans-pathogen interactions. Similar to that of mammals, the C. elegans nervous system regulates its immune system to maintain immunological homeostasis. Studies in the nematode have revealed unprecedented details regarding the molecules, cells, and signaling pathways involved in neural regulation of immunity. Notably, some of the studies indicate that some neuroimmune regulatory circuits need not be "activated" by pathogen infection because they are tonically active and that there could be a predetermined set point for internal immunity, around which the nervous system adjusts immune responses to internal or external environmental changes. Here, we review recent progress on the roles of the C. elegans nervous system in pathogen detection and immune regulation. Because of its advantageous characteristics, we expect that the C. elegans model will be critical for deciphering complex neuroimmune signaling mechanisms that integrate and process multiple sensory cues.


Subject(s)
Caenorhabditis elegans/immunology , Nervous System/immunology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/immunology , Host-Pathogen Interactions , Immunity, Innate , Signal Transduction
11.
Cells ; 10(2)2021 02 20.
Article in English | MEDLINE | ID: mdl-33672617

ABSTRACT

Surgery remains an essential therapeutic approach for most solid malignancies. Although for more than a century accumulating clinical and experimental data have indicated that surgical procedures themselves may promote the appearance and progression of recurrent and metastatic lesions, only in recent years has renewed interest been taken in the mechanism by which metastasizing of cancer occurs following operative procedures. It is well proven now that surgery constitutes a risk factor for the promotion of pre-existing, possibly dormant micrometastases and the acceleration of new metastases through several mechanisms, including the release of neuroendocrine and stress hormones and wound healing pathway-associated immunosuppression, neovascularization, and tissue remodeling. These postoperative consequences synergistically facilitate the establishment of new metastases and the development of pre-existing micrometastases. While only in recent years the role of the peripheral nervous system has been recognized as another contributor to cancer development and metastasis, little is known about the contribution of tumor-associated neuronal and neuroglial elements in the metastatic disease related to surgical trauma and wound healing. Specifically, although numerous clinical and experimental data suggest that biopsy- and surgery-induced wound healing can promote survival and metastatic spread of residual and dormant malignant cells, the involvement of the tumor-associated neuroglial cells in the formation of metastases following tissue injury has not been well understood. Understanding the clinical significance and underlying mechanisms of neuroimmune regulation of surgery-associated metastasis will not only advance the field of neuro-immuno-oncology and contribute to basic science and translational oncology research but will also produce a strong foundation for developing novel mechanism-based therapeutic approaches that may protect patients against the oncologically adverse effects of primary tumor biopsy and excision.


Subject(s)
Neoplasm Metastasis/pathology , Nervous System/immunology , Surgical Procedures, Operative/adverse effects , Animals , Disease Progression , Humans , Neoplasms/pathology , Neoplasms/surgery , Peripheral Nervous System/pathology
12.
Mol Neurobiol ; 58(2): 703-718, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33006752

ABSTRACT

Central innate immunity assists time-dependent neurodevelopment by recruiting and interacting with peripheral immune cells. Microglia are the major player of central innate immunity integrating peripheral signals arising from the circumventricular regions lacking the blood-brain barrier (BBB), via neural afferent pathways such as the vagal nerve and also by choroid plexus into the brain ventricles. Defective and/or unrestrained activation of central and peripheral immunity during embryonic development might set an aberrant connectome establishment and brain function, leading to major psychiatric disorders in postnatal stages. Molecular candidates leading to central and peripheral innate immune overactivation identified metabolic substrates and lipid species as major contributors of immunological priming, supporting the role of a metabolic flexibility node during trained immunity. Mechanistically, trained immunity is established by an epigenetic program including DNA methylation and histone acetylation, as the major molecular epigenetic signatures to set immune phenotypes. By definition, immunological training sets reprogramming of innate immune cells, enhancing or repressing immune responses towards a second challenge which potentially might contribute to neurodevelopment disorders. Notably, the innate immune training might be set during pregnancy by maternal immune activation stimuli. In this review, we integrate the most valuable scientific evidence supporting the role of metabolic cues assisting metabolic flexibility, leading to innate immune training during development and its effects on aberrant neurological phenotypes in the offspring. We also add reports supporting the role of methylation and histone acetylation signatures as a major epigenetic mechanism regulating immune training.


Subject(s)
Immunity, Innate , Metabolism , Nervous System/growth & development , Nervous System/immunology , Animals , Epigenesis, Genetic , Humans , Immunity, Innate/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lipids/chemistry , Metabolism/genetics
13.
Gut Microbes ; 12(1): 1813532, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32900255

ABSTRACT

A growing number of studies support that the bidirectional interactions between the gut microbiota, the immune system and the CNS are relevant for the pathophysiology of MS. Several studies have reported alterations in the gut microbiome of MS patients. In addition, a variety of studies in animal models of MS have suggested that specific members of the gut commensal microbiota can exacerbate or ameliorate neuroinflammation. Probiotics represent oral nontoxic immunomodulatory agents that would exert benefits when using in combination with current MS therapy. Here we investigate the effect of Vivomixx on the gut microbiome and central and peripheral immune responses in a murine model of primary progressive MS. Vivomixx administration was associated with increased abundance of many taxa such as Bacteroidetes, Actinobacteria, Tenericutes and TM7. This was accompanied by a clear improvement of the motor disability of Theiler's virus infected mice; in the CNS Vivomixx reduced microgliosis, astrogliosis and leukocyte infiltration. Notably, the presence of Breg cells (CD19+CD5+CD1dhigh) in the CNS was enhanced by Vivomixx, and while spinal cord gene expression of IL-1ß and IL-6 was diminished, the probiotic promoted IL-10 gene expression. One of the most significant findings was the increased plasma levels of butyrate and acetate levels in TMEV-mice that received Vivomixx. Peripheral immunological changes were subtle but interestingly, the probiotic restricted IL-17 production by Th17-polarized CD4+ T-cells purified from the mesenteric lymph nodes of Theiler's virus infected mice. Our data reinforce the beneficial effects of oral probiotics that would be coadjuvant treatments to current MS therapies.


Subject(s)
Gastrointestinal Microbiome , Multiple Sclerosis/drug therapy , Multiple Sclerosis/microbiology , Nervous System/drug effects , Probiotics/administration & dosage , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Nervous System/immunology , Neuroimmunomodulation/drug effects
14.
Annu Rev Cell Dev Biol ; 36: 441-468, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32722920

ABSTRACT

Fetal neurodevelopment in utero is profoundly shaped by both systemic maternal immunity and local processes at the maternal-fetal interface. Immune pathways are a critical participant in the normal physiology of pregnancy and perturbations of maternal immunity due to infections during this period have been increasingly linked to a diverse array of poor neurological outcomes, including diseases that manifest much later in postnatal life. While experimental models of maternal immune activation (MIA) have provided groundbreaking characterizations of the maternal pathways underlying pathogenesis, less commonly examined are the immune factors that serve pathogen-independent developmental functions in the embryo and fetus. In this review, we explore what is known about the in vivo role of immune factors in fetal neurodevelopment during normal pregnancy and provide an overview of how MIA perturbs the proper orchestration of this sequence of events. Finally, we discuss how the dysregulation of immune factors may contribute to the manifestation of a variety of neurological disorders.


Subject(s)
Fetus/embryology , Fetus/immunology , Immunologic Factors/metabolism , Nervous System/embryology , Nervous System/immunology , Animals , Cytokines/metabolism , Female , Humans , Maternal-Fetal Exchange/immunology , Models, Biological , Pregnancy
15.
J Leukoc Biol ; 108(1): 189-198, 2020 07.
Article in English | MEDLINE | ID: mdl-32645257

ABSTRACT

Acupuncture is a centuried and unfading treatment of traditional Chinese medicine, which has been proved to exert curative effects on various disorders. Numerous works have been put in to uncover the effective mechanisms of acupuncture. And the interdependent interaction between acupuncture and acupoint microenvironment is a crucial topic. As a benign minimally invasive stimulation, the insertion and manipulation of needle at acupoint could cause deformation of local connective tissue and secretion of various molecules, such as high mobility group box 1 and ATP. The molecules are secreted into extracellular space and bind to the corresponding receptors thus active NF-κB, MAPK, ERK pathways on mast cells, fibroblasts, keratinocytes, and monocytes/macrophages, among others. This is supposed to trigger following transcription and translation of immune factors and neural active substance, as well as promote the free ion movement (such as Ca2+ influx) and the expansion of blood vessels to recruit more immune cells to acupoint. Finally, acupuncture could enhance network connectivity of local microenvironment at acupoints. The earlier mentioned substances further act on a variety of receptors in local nerve endings, transmitting electrical and biochemical signals to the CNS, and giving full play to the acupuncture action. In conclusion, we portrayed a neuro-immune microenvironment network of acupoints that medicates the acupuncture action, and would lay a foundation for the systematic study of the complex network relationship of acupoints in the future.


Subject(s)
Acupuncture Points , Acupuncture Therapy , Cellular Microenvironment/immunology , Nervous System/immunology , Humans
16.
J Neurovirol ; 26(4): 482-495, 2020 08.
Article in English | MEDLINE | ID: mdl-32495195

ABSTRACT

Varicella zoster virus (VZV) is a pathogenic human herpes virus which causes varicella as a primary infection, following which it becomes latent in peripheral autonomic, sensory, and cranial nerve ganglionic neurons from where it may reactivate after decades to cause herpes zoster. VZV reactivation may also cause a wide spectrum of neurological syndromes, in particular, acute encephalitis and vasculopathy. While there is potentially a large number of coding viral mutations that might predispose certain individuals to VZV infections, in practice, a variety of host factors are the main determinants of VZV infection, both disseminated and specifically affecting the nervous system. Host factors include increasing age with diminished cell-mediated immunity to VZV, several primary immunodeficiency syndromes, secondary immunodeficiency syndromes, and drug-induced immunosuppression. In some cases, the molecular immunological basis underlying the increased risk of VZV infections has been defined, in particular, the role of POL III mutations, but in other cases, the mechanisms have yet to be determined. The role of immunization in immunosuppressed individuals as well as its possible efficacy in preventing both generalized and CNS-specific infections will require further investigation to clarify in such patients.


Subject(s)
Acquired Immunodeficiency Syndrome/virology , Encephalitis, Varicella Zoster/virology , Herpesvirus 3, Human/pathogenicity , Host-Pathogen Interactions/immunology , Immunocompromised Host , Nervous System/virology , Primary Immunodeficiency Diseases/virology , Acquired Immunodeficiency Syndrome/complications , Acquired Immunodeficiency Syndrome/genetics , Acquired Immunodeficiency Syndrome/immunology , DNA Polymerase III/genetics , DNA Polymerase III/immunology , Encephalitis, Varicella Zoster/complications , Encephalitis, Varicella Zoster/genetics , Encephalitis, Varicella Zoster/immunology , Gene Expression , Herpesvirus 3, Human/immunology , Host-Pathogen Interactions/genetics , Humans , Immunity, Cellular , Immunosuppressive Agents/adverse effects , Lymphocytes/immunology , Lymphocytes/pathology , Lymphocytes/virology , Mutation , Nervous System/immunology , Nervous System/pathology , Primary Immunodeficiency Diseases/complications , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Virus Latency/immunology
17.
Front Immunol ; 11: 947, 2020.
Article in English | MEDLINE | ID: mdl-32582147

ABSTRACT

A critical role for IL-17, a cytokine produced by T helper 17 (Th17) cells, has been indicated in the pathogenesis of chronic inflammatory and autoimmune diseases. A positive effect of blockade of IL-17 secreted by autoreactive T cells has been shown in various inflammatory diseases. Several cytokines, whose production is affected by environmental factors, control Th17 differentiation and its maintenance in tissues during chronic inflammation. The roles of IL-17 in the pathogenesis of chronic neuroinflammatory conditions, multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE), Alzheimer's disease, and ischemic brain injury are reviewed here. The role of environmental stimuli in Th17 differentiation is also summarized, highlighting the role of viral infection in the regulation of pathogenic T helper cells in EAE.


Subject(s)
Inflammation/metabolism , Interleukin-17/metabolism , Nervous System Diseases/metabolism , Nervous System/metabolism , Th17 Cells/metabolism , Animals , Cell Differentiation , Chronic Disease , Humans , Inflammation/immunology , Inflammation/physiopathology , Nervous System/immunology , Nervous System/physiopathology , Nervous System Diseases/immunology , Nervous System Diseases/physiopathology , Phenotype , Receptors, Interleukin-17/metabolism , Signal Transduction , Th17 Cells/immunology
18.
Trends Immunol ; 41(5): 359-362, 2020 05.
Article in English | MEDLINE | ID: mdl-32249062

ABSTRACT

There is a major gap in our understanding of how the intestinal immune and nervous systems are integrated to regulate protective adaptations to enteric infections while maintaining tissue homeostasis. Three recent complementary reports published in Cell (2020) provide new mechanistic insights into how this enteric neuro-immune crosstalk may occur.


Subject(s)
Homeostasis , Intestinal Diseases , Intestines , Nervous System , Animals , Homeostasis/immunology , Humans , Intestinal Diseases/immunology , Intestinal Diseases/microbiology , Intestines/immunology , Nervous System/immunology
19.
Immunity ; 52(3): 464-474, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32187517

ABSTRACT

The ability of the nervous system to sense environmental stimuli and to relay these signals to immune cells via neurotransmitters and neuropeptides is indispensable for effective immunity and tissue homeostasis. Depending on the tissue microenvironment and distinct drivers of a certain immune response, the same neuronal populations and neuro-mediators can exert opposing effects, promoting or inhibiting tissue immunity. Here, we review the current understanding of the mechanisms that underlie the complex interactions between the immune and the nervous systems in different tissues and contexts. We outline current gaps in knowledge and argue for the importance of considering infectious and inflammatory disease within a conceptual framework that integrates neuro-immune circuits both local and systemic, so as to better understand effective immunity to develop improved approaches to treat inflammation and disease.


Subject(s)
Immune System/immunology , Nervous System/immunology , Neuroimmunomodulation/immunology , Neurons/immunology , Animals , Humans , Immune System/cytology , Immune System/metabolism , Immunity, Innate/immunology , Nervous System/cytology , Nervous System/metabolism , Neurogenic Inflammation/immunology , Neurogenic Inflammation/metabolism , Neurons/metabolism , Neuropeptides/immunology , Neuropeptides/metabolism , Signal Transduction/immunology
20.
Curr Protein Pept Sci ; 21(5): 517-526, 2020.
Article in English | MEDLINE | ID: mdl-31613726

ABSTRACT

Lyme disease (LD) is an infectious disease caused by the spirochetes of genus borrelia, which are transmitted by the ticks of the genus ixodes. LD is transmitted by the spirochete B. burgdorferi sensu lato. Once in contact with the host through a tick bite, the pathogen comes into contact with the host defense, and must escape this machinery to establish LD, thus using a large number of mechanisms involving the vector of the pathogen, the pathogen itself and also the host. The initial diagnosis of the disease can be made based on the clinical symptoms of LD and the disease can be treated and cured with antibiotics if the diagnosis is made early in the beginning of the disease. Contrariwise, if LD is left untreated, the pathogen disseminates throughout the tissues and organs of the body, where it establishes different types of disease manifestations. In the nervous system, the inflammation caused by B. burgdorferi is known as Lyme neuroborreliosis (LNB). LNB is one of the principal manifestations of LD. In this review, we systematically describe the different molecular interactions among B. burgdorferi, the vector (tick) and the mammalian host.


Subject(s)
Arachnid Vectors/microbiology , Bacterial Proteins/genetics , Borrelia burgdorferi/pathogenicity , Host-Pathogen Interactions/genetics , Ixodes/microbiology , Lyme Disease/genetics , Membrane Proteins/genetics , Receptors, Cell Surface/genetics , Animals , Arachnid Vectors/immunology , Bacterial Proteins/immunology , Borrelia burgdorferi/immunology , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation , Host-Pathogen Interactions/immunology , Humans , Ixodes/immunology , Lyme Disease/immunology , Lyme Disease/microbiology , Lyme Disease/pathology , Membrane Proteins/immunology , Nervous System/immunology , Nervous System/microbiology , Nervous System/pathology , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptors, Cell Surface/immunology , Saliva/microbiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...