Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
1.
J Dermatol ; 51(5): 621-631, 2024 May.
Article in English | MEDLINE | ID: mdl-38605467

ABSTRACT

Alopecia areata refers to an autoimmune illness indicated by persistent inflammation. The key requirement for alopecia areata occurrence is the disruption of immune-privileged regions within the hair follicles. Recent research has indicated that neuropeptides play a role in the damage to hair follicles by triggering neurogenic inflammation, stimulating mast cells ambient the follicles, and promoting apoptotic processes in keratinocytes. However, the exact pathogenesis of alopecia areata requires further investigation. Recently, there has been an increasing focus on understanding the mechanisms of immune diseases resulting from the interplay between the nervous and the immune system. Neurogenic inflammation due to neuroimmune disorders of the skin system may disrupt the inflammatory microenvironment of the hair follicle, which plays a crucial part in the progression of alopecia areata.


Subject(s)
Alopecia Areata , Hair Follicle , Neurogenic Inflammation , Alopecia Areata/immunology , Alopecia Areata/etiology , Alopecia Areata/pathology , Humans , Hair Follicle/immunology , Hair Follicle/pathology , Neurogenic Inflammation/immunology , Neurogenic Inflammation/etiology , Neuropeptides/metabolism , Neuropeptides/immunology , Mast Cells/immunology , Keratinocytes/immunology , Keratinocytes/pathology , Apoptosis/immunology , Animals
2.
Int J Mol Sci ; 22(23)2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34884907

ABSTRACT

Endometriosis (EM) is an estrogen-dependent disease characterized by the presence of epithelial, stromal, and smooth muscle cells outside the uterine cavity. It is a chronic and debilitating condition affecting ~10% of women. EM is characterized by infertility and pain, such as dysmenorrhea, chronic pelvic pain, dyspareunia, dysuria, and dyschezia. Although EM was first described in 1860, its aetiology and pathogenesis remain uncertain. Recent evidence demonstrates that the peripheral nervous system plays an important role in the pathophysiology of this disease. Sensory nerves, which surround and innervate endometriotic lesions, not only drive the chronic and debilitating pain associated with EM but also contribute to a growth phenotype by secreting neurotrophic factors and interacting with surrounding immune cells. Here we review the role that peripheral nerves play in driving and maintaining endometriotic lesions. A better understanding of the role of this system, as well as its interactions with immune cells, will unearth novel disease-relevant pathways and targets, providing new therapeutics and better-tailored treatment options.


Subject(s)
Endometriosis/immunology , Nerve Growth Factors/metabolism , Neurogenic Inflammation/etiology , Endometriosis/complications , Female , Gene Expression Regulation , Humans , Neurogenic Inflammation/immunology , Pelvic Pain/etiology , Pelvic Pain/immunology , Sensory Receptor Cells/immunology
3.
Immunity ; 54(3): 484-498.e8, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33581044

ABSTRACT

Pathologic roles of innate immunity in neurologic disorders are well described, but their beneficial aspects are less understood. Dectin-1, a C-type lectin receptor (CLR), is largely known to induce inflammation. Here, we report that Dectin-1 limited experimental autoimmune encephalomyelitis (EAE), while its downstream signaling molecule, Card9, promoted the disease. Myeloid cells mediated the pro-resolution function of Dectin-1 in EAE with enhanced gene expression of the neuroprotective molecule, Oncostatin M (Osm), through a Card9-independent pathway, mediated by the transcription factor NFAT. Furthermore, we find that the Osm receptor (OsmR) functioned specifically in astrocytes to reduce EAE severity. Notably, Dectin-1 did not respond to heat-killed Mycobacteria, an adjuvant to induce EAE. Instead, endogenous Dectin-1 ligands, including galectin-9, in the central nervous system (CNS) were involved to limit EAE. Our study reveals a mechanism of beneficial myeloid cell-astrocyte crosstalk regulated by a Dectin-1 pathway and identifies potential therapeutic targets for autoimmune neuroinflammation.


Subject(s)
Astrocytes/immunology , Brain/pathology , CARD Signaling Adaptor Proteins/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Lectins, C-Type/metabolism , Multiple Sclerosis/immunology , Myeloid Cells/immunology , Neurogenic Inflammation/immunology , Receptors, Mitogen/metabolism , Animals , Cell Communication , Cells, Cultured , Disease Models, Animal , Galectins/metabolism , Gene Expression Regulation , Lectins, C-Type/genetics , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/immunology , Oncostatin M/genetics , Oncostatin M/metabolism , Oncostatin M Receptor beta Subunit/metabolism , Peptide Fragments/immunology , Receptors, Mitogen/genetics , Signal Transduction
4.
Immunity ; 54(2): 308-323.e6, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33421362

ABSTRACT

Th17 cells are known to exert pathogenic and non-pathogenic functions. Although the cytokine transforming growth factor ß1 (TGF-ß1) is instrumental for Th17 cell differentiation, it is dispensable for generation of pathogenic Th17 cells. Here, we examined the T cell-intrinsic role of Activin-A, a TGF-ß superfamily member closely related to TGF-ß1, in pathogenic Th17 cell differentiation. Activin-A expression was increased in individuals with relapsing-remitting multiple sclerosis and in mice with experimental autoimmune encephalomyelitis. Stimulation with interleukin-6 and Activin-A induced a molecular program that mirrored that of pathogenic Th17 cells and was inhibited by blocking Activin-A signaling. Genetic disruption of Activin-A and its receptor ALK4 in T cells impaired pathogenic Th17 cell differentiation in vitro and in vivo. Mechanistically, extracellular-signal-regulated kinase (ERK) phosphorylation, which was essential for pathogenic Th17 cell differentiation, was suppressed by TGF-ß1-ALK5 but not Activin-A-ALK4 signaling. Thus, Activin-A drives pathogenic Th17 cell differentiation, implicating the Activin-A-ALK4-ERK axis as a therapeutic target for Th17 cell-related diseases.


Subject(s)
Activins/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Neurogenic Inflammation/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Activins/genetics , Animals , Cell Differentiation , Cells, Cultured , Humans , Mice , Mice, Knockout , Molecular Targeted Therapy , Signal Transduction
5.
Front Immunol ; 11: 571844, 2020.
Article in English | MEDLINE | ID: mdl-33193354

ABSTRACT

Multiple sclerosis (MS) is an inflammatory and demyelinating disease of the central nervous system (CNS). The persistent inflammation is being mainly attributed to local oxidative stress and inflammasome activation implicated in the ensuing demyelination and axonal damage. Since new control measures remain necessary, we evaluated the preventive and therapeutic potential of a beta-selenium-lactic acid derivative (LAD-ßSe), which is a source of organic selenium under development, to control experimental autoimmune encephalomyelitis (EAE) that is an animal model for MS. Two EAE murine models: C57BL/6 and SJL/J immunized with myelin oligodendrocyte glycoprotein and proteolipid protein, respectively, and a model of neurodegeneration induced by LPS in male C57BL/6 mice were used. The preventive potential of LAD-ßSe was initially tested in C57BL/6 mice, the chronic MS model, by three different protocols that were started 14 days before or 1 or 7 days after EAE induction and were extended until the acute disease phase. These three procedures were denominated preventive therapy -14 days, 1 day, and 7 days, respectively. LAD-ßSe administration significantly controlled clinical EAE development without triggering overt hepatic and renal dysfunction. In addition of a tolerogenic profile in dendritic cells from the mesenteric lymph nodes, LAD-ßSe also downregulated cell amount, activation status of macrophages and microglia, NLRP3 (NOD-like receptors) inflammasome activation and other pro-inflammatory parameters in the CNS. The high Se levels found in the CNS suggested that the product crossed the blood-brain barrier having a possible local effect. The hypothesis that LAD-ßSe was acting locally was then confirmed by using the LPS-induced neurodegeneration model that also displayed Se accumulation and downmodulation of pro-inflammatory parameters in the CNS. Remarkably, therapy with LAD-ßSe soon after the first remitting episode in SJL/J mice, also significantly downmodulated local inflammation and clinical disease severity. This study indicates that LAD-ßSe, and possibly other derivatives containing Se, are able to reach the CNS and have the potential to be used as preventive and therapeutic measures in distinct clinical forms of MS.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Central Nervous System/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Inflammasomes/metabolism , Microglia/pathology , Multiple Sclerosis/drug therapy , Neurogenic Inflammation/drug therapy , Selenium/therapeutic use , Animals , Central Nervous System/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Lactic Acid/chemistry , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neurogenic Inflammation/immunology , Selenium/chemistry
6.
Front Immunol ; 11: 565521, 2020.
Article in English | MEDLINE | ID: mdl-33013930

ABSTRACT

Neurological disorders caused by neuroviral infections are an obvious pathogenic manifestation. However, non-neurotropic viruses or peripheral viral infections pose a considerable challenge as their neuropathological manifestations do not emerge because of primary infection. Their secondary or bystander pathologies develop much later, like a syndrome, during and after the recovery of patients from the primary disease. Massive inflammation caused by peripheral viral infections can trigger multiple neurological anomalies. These neurological damages may range from a general cognitive and motor dysfunction up to a wide spectrum of CNS anomalies, such as Acute Necrotizing Hemorrhagic Encephalopathy, Guillain-Barré syndrome, Encephalitis, Meningitis, anxiety, and other audio-visual disabilities. Peripheral viruses like Measles virus, Enteroviruses, Influenza viruses (HIN1 series), SARS-CoV-1, MERS-CoV, and, recently, SARS-CoV-2 are reported to cause various neurological manifestations in patients and are proven to be neuropathogenic even in cellular and animal model systems. This review presents a comprehensive picture of CNS susceptibilities toward these peripheral viral infections and explains some common underlying themes of their neuropathology in the human brain.


Subject(s)
Betacoronavirus/immunology , Coronavirus Infections/complications , Coronavirus Infections/immunology , Middle East Respiratory Syndrome Coronavirus/immunology , Neurogenic Inflammation/complications , Neurogenic Inflammation/immunology , Pneumonia, Viral/complications , Pneumonia, Viral/immunology , Severe Acute Respiratory Syndrome/complications , Severe acute respiratory syndrome-related coronavirus/immunology , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/virology , COVID-19 , Coronavirus Infections/virology , Cytokines/blood , Disease Models, Animal , Humans , Microglia/immunology , Microglia/virology , Neurogenic Inflammation/virology , Pandemics , Pneumonia, Viral/virology , SARS-CoV-2 , Severe Acute Respiratory Syndrome/immunology , Severe Acute Respiratory Syndrome/virology
7.
Front Immunol ; 11: 2013, 2020.
Article in English | MEDLINE | ID: mdl-33117329

ABSTRACT

Acute-on-chronic liver failure (ACLF) is a severe life-threatening condition with high risk of multiorgan failure, sepsis, and mortality. ACLF activates a multifaceted interplay of both innate and adaptive immune response in the host which governs the overall outcome. Innate immune cells recognize the conserved elements of microbial and viral origin, both to extort instant defense by transforming into diverse modules of effector responses and to generate long-lasting immunity but can also trigger a massive intrahepatic immune inflammatory response. Acute insult results in the activation of innate immune cells which provokes cytokine and chemokine cascade and subsequently initiates aggressive systemic inflammatory response syndrome, hepatic damage, and high mortality in ACLF. Dysregulated innate immune response not only plays a critical role in disease progression but also potentially correlates with clinical disease severity indices including Child-Turcotte-Pugh, a model for end-stage liver disease, and sequential organ failure assessment score. A better understanding of the pathophysiological basis of the disease and precise immune mechanisms associated with liver injury offers a novel approach for the development of new and efficient therapies to treat this severely ill entity. Immunotherapies could be helpful in targeting immune-mediated organ damage which may constrain progression toward liver failure and eventually reduce the requirement for liver transplantation. Here, in this review we discuss the defects of different innate immune cells in ACLF which updates the current knowledge of innate immune response and provide potential targets for new therapeutic interventions.


Subject(s)
Acute-On-Chronic Liver Failure/immunology , Immunotherapy/methods , Neurogenic Inflammation/immunology , Animals , Cytokines/metabolism , Humans , Immunity, Innate , Neuroimmunomodulation
8.
J Immunol ; 205(7): 1787-1798, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32848034

ABSTRACT

Dengue virus (DENV) infection disrupts host innate immune signaling at various checkpoints. Cellular levels and stability of intermediate signaling molecules are a crucial hijacking point for a successful viral pathogenesis. Stability and turnover of all the cellular proteins including intermediate signaling molecules are principally regulated by proteasomal degradation pathway. In this study, we show that how DENV infection and particularly DENV-NS1 can modulate the host extracellular vesicle (EV) cargo to manipulate the deubiquitination machinery of the human microglial cell (CHME3). We have performed EV harvesting, size analysis by nanoparticle tracking analysis, identification of cargo microRNA via quantitative PCR, microRNA target validation by overexpression, and knockdown via mimics and anti-miRs, immunoblotting, dual luciferase reporter assay, in vivo ubiquitination assay, chase assay, and promoter activity assay to reach the conclusion. In this study, we show that DENV-infected monocytes and DENV-NS1-transfected cells release high amounts of EVs loaded with miR-148a. These EVs get internalized by human microglial cells, and miR-148a suppresses the ubiquitin-specific peptidase 33 (USP33) protein expression levels via binding to its 3' untranslated region. Reduced USP33 in turn decreases the stability of cellular ATF3 protein via deubiquitylation. ATF3 acts as a suppressor of major proinflammatory gene expression pathways of TNF-α, NF-κB, and IFN-ß. Our mechanistic model explains how DENV uses the EV pathway to transfer miR-148a for modulating USP33 and downstream ATF3 levels in human microglial cells and contributes in neuroinflammation within the CNS.


Subject(s)
Activating Transcription Factor 3/metabolism , Dengue Virus/physiology , Dengue/immunology , Extracellular Vesicles/metabolism , Microglia/physiology , Neurogenic Inflammation/immunology , Ubiquitin Thiolesterase/metabolism , Animals , Cell Line , Cells, Cultured , Culicidae , Cytokines/metabolism , Dengue/virology , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Inflammation Mediators/metabolism , MicroRNAs/genetics , Neurogenic Inflammation/virology , Signal Transduction , Ubiquitination/genetics , Virus Replication
9.
Front Immunol ; 11: 1024, 2020.
Article in English | MEDLINE | ID: mdl-32733433

ABSTRACT

The interaction between microglia and astrocytes significantly influences neuroinflammation. Microglia/astrocytes, part of the neurovascular unit (NVU), are activated by various brain insults. The local extracellular and intracellular signals determine their characteristics and switch of phenotypes. Microglia and astrocytes are activated into two polarization states: the pro-inflammatory phenotype (M1 and A1) and the anti-inflammatory phenotype (M2 and A2). During neuroinflammation, induced by stroke or lipopolysaccharides, microglia are more sensitive to pathogens, or damage; they are thus initially activated into the M1 phenotype and produce common inflammatory signals such as IL-1 and TNF-α to trigger reactive astrocytes into the A1 phenotype. These inflammatory signals can be amplified not only by the self-feedback loop of microglial activation but also by the unique anatomy structure of astrocytes. As the pathology further progresses, resulting in local environmental changes, M1-like microglia switch to the M2 phenotype, and M2 crosstalk with A2. While astrocytes communicate simultaneously with neurons and blood vessels to maintain the function of neurons and the blood-brain barrier (BBB), their subtle changes may be identified and responded by astrocytes, and possibly transferred to microglia. Although both microglia and astrocytes have different functional characteristics, they can achieve immune "optimization" through their mutual communication and cooperation in the NVU and build a cascaded immune network of amplification.


Subject(s)
Astrocytes/immunology , Brain/physiology , Microglia/immunology , Neurogenic Inflammation/immunology , Neurons/physiology , Neurovascular Coupling/physiology , Animals , Humans , Inflammation Mediators/metabolism
10.
Int Immunopharmacol ; 86: 106734, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32604067

ABSTRACT

Estrogen replacement therapy (ERT) has been proven to relieve menopausal-related mental disorders including depression in postmenopausal women. However, the unsafety of ERT hinders its clinical use. In this study, we would evaluate whether vitamin D (VD), a hormone with optimal safety profile, could relieve the depressive-like symptom in ovariectomized (OVX) rats. Furthermore, we would determine whether vitamin D and 17ß-estradiol (E2) exert neurological function through their immunomodulatory effect in OVX rats. Middle-aged female SD rats were randomly divided into four groups, namely, control (SHAM), OVX, OVX + VD, and OVX + E2. Vitamin D (calcitriol, 100 ng/kg) and 17ß-estradiol (30 µg/kg) had been daily gavaged in the OVX + VD and OVX + E2 group, respectively. After 10-week administration, vitamin D and 17ß-estradiol both showed anti-depressive-like activity in the OVX rats. Using the method of immunofluorescent staining and western blot, vitamin D and 17ß-estradiol were demonstrated to upregulate each other's receptors, including VDR, ERα, and ERß in the hippocampus of OVX rats. Additionally, the upregulation of VDR, calbindin-D28k, and calbindin-D9k suggested that the vitamin D signaling system was amplified by vitamin D and 17ß-estradiol. Vitamin D and 17ß-estradiol showed neuroprotective effects by decreasing OVX-induced apoptosis and neuronal damage, regulating the AMPK/NF-κB signaling pathway, and reducing the proinflammatory cytokines (IL-1ß, IL-6, and TNFα), as well as iNOS and COX-2 in the hippocampus of OVX rats. Collectively, the present study demonstrated that vitamin D and 17ß-estradiol could upregulate each other's receptors and regulate the AMPK/NF-κB pathway to relieve the OVX-induced depressive-like state. The results could stimulate translational research towards the vitamin D potential for prevention or treatment of menopause-related depression.


Subject(s)
Estradiol/metabolism , Neurogenic Inflammation/immunology , Vitamin D/metabolism , Adenylate Kinase/metabolism , Animals , Cells, Cultured , Depression , Female , Humans , NF-kappa B/metabolism , Neuroprotective Agents , Ovariectomy , Rats , Rats, Sprague-Dawley , Signal Transduction
11.
Int Immunopharmacol ; 87: 106814, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32707491

ABSTRACT

Systemic inflammation induces cognitive impairments via unclear mechanisms. Increasing evidence has suggested complement C3/C3a receptor signaling, a key component of innate immune pathogen defense, plays an important role in cognition and neurodegeneration, whereas its dysfunction is implicated in many neurological disorders. However, it remains unclear whether complement C3/C3a receptor signaling was involved in systemic inflammation-induced cognitive impairments. In the present study, we showed that hippocampal complement C3 levels in astrocytes and C3a receptor expressions in microglia were specifically up-regulated after lipopolysaccharide (LPS) injection. Interestingly, LPS selectively induced inhibitory but not excitatory synapse related protein loss. Notably, C3a receptor antagonist SB290157 trifluoroacetate attenuated LPS-induced hippocampal neuroinflammation and inhibitory synapse related protein loss, contributing to improved cognitive function. In conclusion, our study suggests that complement C3/C3a receptor signaling plays a key role in LPS-induced cognitive impairments, which may serve a therapeutic target for systemic inflammation related cognitive disorders.


Subject(s)
Astrocytes/immunology , Cognitive Dysfunction/immunology , Complement C3/metabolism , Hippocampus/pathology , Microglia/immunology , Neurogenic Inflammation/immunology , Receptors, Complement/metabolism , Animals , Arginine/administration & dosage , Arginine/analogs & derivatives , Benzhydryl Compounds/administration & dosage , Electrical Synapses , Humans , Male , Mice , Mice, Inbred C57BL , Signal Transduction
12.
Front Immunol ; 11: 1051, 2020.
Article in English | MEDLINE | ID: mdl-32547555

ABSTRACT

Corticosteroids are effective therapy for autoimmune diseases but serious adverse effects preclude their prolonged use. However, immune-suppressive biologics that inhibit lymphoid proliferation are now in use as corticosteroid sparing-agents but with variable success; thus, the need to develop alternative immune-suppressive approaches including cell-based therapies. Efficacy of ex-vivo-generated IL-35-producing regulatory B-cells (i35-Bregs) in suppressing/ameliorating encephalomyelitis or uveitis in mouse models of multiple sclerosis or uveitis, respectively, is therefore a promising therapeutic approach for CNS autoimmune diseases. However, i35-Breg therapy in human uveitis would require producing autologous Bregs from each patient to avoid immune-rejection. Because exosomes exhibit minimal toxicity and immunogenicity, we investigated whether i35-Bregs release exosomes that can be exploited therapeutically. Here, we demonstrate that i35-Bregs release exosomes that contain IL-35 (i35-Exosomes). In this proof-of-concept study, we induced experimental autoimmune uveitis (EAU), monitored EAU progression by fundoscopy, histology, optical coherence tomography and electroretinography, and investigated whether i35-Exosomes treatment would suppress uveitis. Mice treated with i35-Exosomes developed mild EAU with low EAU scores and disease protection correlated with expansion of IL-10 and IL-35 secreting Treg cells with concomitant suppression of Th17 responses. In contrast, significant increase of Th17 cells in vitreous and retina of control mouse eyes was accompanied by severe choroiditis, massive retinal-folds, and photoreceptor cell damage. These hallmark features of severe uveitis were absent in exosome-treated mice and visual impairment detected by ERG was modest compared to control mice. Absence of toxicity or alloreactivity associated with exosomes thus makes i35-Exosomes attractive therapeutic option for delivering IL-35 into CNS tissues.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocytes, Regulatory/immunology , Exosomes/metabolism , Immunotherapy, Adoptive/methods , Interleukins/metabolism , Neurogenic Inflammation/immunology , Uveitis/immunology , Animals , Autoimmune Diseases/therapy , B-Lymphocytes, Regulatory/transplantation , Cells, Cultured , Disease Models, Animal , Humans , Immune Tolerance , Immunomodulation , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Neurogenic Inflammation/therapy , T-Lymphocytes, Regulatory/immunology
13.
J Immunol ; 205(2): 502-510, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32503894

ABSTRACT

Despite mounting evidence suggesting the involvement of the immune system in regulating brain function, the specific role of immune and inflammatory cells in neurodegenerative diseases remain poorly understood. In this study, we report that depletion of NK cells, a type of innate lymphocytes, alleviates neuroinflammation, stimulates neurogenesis, and improves cognitive function in a triple-transgenic Alzheimer disease (AD) mouse model. NK cells in the brains of triple-transgenic AD mouse model (3xTg-AD) mice exhibited an enhanced proinflammatory profile. Depletion of NK cells by anti-NK1.1 Abs drastically improved cognitive function of 3xTg-AD mice. NK cell depletion did not affect amyloid ß concentrations but enhanced neurogenesis and reduced neuroinflammation. Notably, in 3xTg-AD mice depleted of NK cells, microglia demonstrated a homeostatic-like morphology, decreased proliferative response and reduced expression of neurodestructive proinflammatory cytokines. Together, our results suggest a proinflammatory role for NK cells in 3xTg-AD mice and indicate that targeting NK cells might unlock novel strategies to combat AD.


Subject(s)
Alzheimer Disease/immunology , Killer Cells, Natural/immunology , Neurogenic Inflammation/immunology , Alzheimer Disease/therapy , Animals , Antibodies/metabolism , Antigens, Ly/metabolism , Apoptosis , Cognition , Disease Models, Animal , Humans , Lymphocyte Depletion , Mice , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Neurogenesis , Neurogenic Inflammation/therapy , Recovery of Function
14.
J Immunol Res ; 2020: 6937154, 2020.
Article in English | MEDLINE | ID: mdl-32280719

ABSTRACT

Down Syndrome (DS) is the most common chromosomal disorder. Although DS individuals are mostly perceived as characterized by some distinct physical features, cognitive disabilities, and cardiac defects, they also show important dysregulations of immune functions. While critical information is available for adults with DS, little literature is available on the neuroinflammation in prepubertal DS children. We aimed to evaluate in prepubertal DS children the serum levels of nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), oxidative stress as free oxygen radicals defense (FORD), free oxygen radicals test (FORT), and cytokines playing key roles in neuroinflammation and oxidative processes as TNF-α, TGF-ß, MCP-1, IL-1α, IL-2, IL-6, IL-10, and IL-12. No differences were found in NGF between DS children and controls. However, BDNF was higher in DS subjects compared to controls. We also did not reveal changes in FORD and FORT. Quite interestingly, the serum of DS children disclosed a marked decrease in all analyzed cytokines with evident differences in serum cytokine presence between male and female DS children. In conclusion, the present study evidences in DS prepubertal children a disruption in the neurotrophins and immune system pathways.


Subject(s)
Biomarkers/blood , Brain-Derived Neurotrophic Factor/blood , Cytokines/blood , Down Syndrome/diagnosis , Neurogenic Inflammation/diagnosis , Child, Preschool , Down Syndrome/immunology , Female , Humans , Immune System , Male , Nerve Growth Factor/blood , Neurogenic Inflammation/immunology , Puberty , Reactive Oxygen Species/blood , Sex Factors , Signal Transduction
15.
Sci Rep ; 10(1): 5941, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32246039

ABSTRACT

Neuroinflammation is involved in the pathogenesis of amyotrophic lateral sclerosis (ALS), but only limited data are available on systematic peripheral and central immune cell profiles in ALS. We studied detailed immune profiles of 73 ALS patients and 48 healthy controls (controls) in peripheral blood by fluorescence-activated cell sorting as well as cytokine expression profiles in serum. In a subgroup of 16 ALS patients and 10 controls we additionally studied cerebrospinal fluid (CSF) samples. In peripheral blood, T cell subtypes presented a shift towards pro-inflammatory Th 1 and Th 17 cells whereas anti-inflammatory Th2 and T regulatory cells were decreased. Important players in innate immunity including distinct monocyte (Mo) and natural killer (NK) cell subtypes were changed in ALS patients compared to controls. Pro-inflammatory serum cytokines such as interleukin (IL)-1 beta, IL-6 and interferon-gamma (IFN-gamma) were increased and the anti-inflammatory cytokine IL-10 was decreased. Correlation analysis revealed moderate negative correlations between Th1 and Th17 to the ALS functional rating scale revised (ALSFRS-R) and to forced vital capacity. In CSF samples, no relevant alteration of the immune profile was found. In conclusion, the immune profile in ALS was shifted towards a Th1/Th17 cell-mediated pro-inflammatory immune response and correlated to disease severity and progression. Large prospective studies are needed to confirm these findings.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Killer Cells, Natural/immunology , Neurogenic Inflammation/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Aged , Aged, 80 and over , Cytokines/metabolism , Disease Progression , Female , Flow Cytometry , Humans , Male , Middle Aged , Th1-Th2 Balance
16.
J Immunol Res ; 2020: 9469210, 2020.
Article in English | MEDLINE | ID: mdl-32258178

ABSTRACT

Alzheimer's disease (AD) has been clinically characterized by a progressive degeneration of neurons which resulted in a gradual and irreversible cognitive impairment. The accumulation of Aß and τ proteins in the brain contribute to the severity of the disease. Recently, vitexin compound has been the talk amongst researchers due to its pharmacological properties as anti-inflammation and anti-AD. However, the epigenetic mechanism of the compound in regulating the neuroinflammation activity is yet to be fully elucidated. Hence, this review discusses the potential of vitexin compound to have the pharmacoepigenetic property in regulating the neuroinflammation activity in relation to AD. It is with hope that the review would unveil the potential of vitexin as the candidate in treating AD.


Subject(s)
Alzheimer Disease/immunology , Anti-Inflammatory Agents/metabolism , Apigenin/metabolism , Neurogenic Inflammation/immunology , Neurons/pathology , Alzheimer Disease/genetics , Animals , Epigenesis, Genetic , Humans , Neurogenic Inflammation/genetics
17.
Front Immunol ; 11: 180, 2020.
Article in English | MEDLINE | ID: mdl-32153564

ABSTRACT

The transient receptor potential (TRP) cation channels are present in abundance across the gastrointestinal (GI) tract, serving as detectors for a variety of stimuli and secondary transducers for G-protein coupled receptors. The activation of TRP channels triggers neurogenic inflammation with related neuropeptides and initiates immune reactions by extra-neuronally regulating immune cells, contributing to the GI homeostasis. However, under pathological conditions, such as inflammatory bowel disease (IBD), TRP channels are involved in intestinal inflammation. An increasing number of human and animal studies have indicated that TRP channels are correlated to the visceral hypersensitivity (VHS) and immune pathogenesis in IBD, leading to an exacerbation or amelioration of the VHS or intestinal inflammation. Thus, TRP channels are a promising target for novel therapeutic methods for IBD. In this review, we comprehensively summarize the functions of TRP channels, especially their potential roles in immunity and IBD. Additionally, we discuss the contradictory findings of prior studies and offer new insights with regard to future research.


Subject(s)
Gastrointestinal Tract/metabolism , Inflammatory Bowel Diseases/immunology , Neurogenic Inflammation/immunology , Neurogenic Inflammation/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Homeostasis/immunology , Humans , Mice , Neuropeptides/metabolism , T-Lymphocytes/immunology , Visceral Pain/metabolism
18.
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
19.
Int Immunopharmacol ; 81: 106284, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32058931

ABSTRACT

Chronic intermittent hypoxia (CIH) is the main symptom of obstructive sleep apnea syndrome (OSAS) and causes neural damage and cognitive deficits via neuroinflammation. Toll-like receptors (TLRs), especially TLR2, play an important role in neuroinflammation. However, the mechanisms by which TLR2 participates in CIH-induced cognitive deficits remain unclear. In this study, wild-type (WT) and TLR2 knock out (KO) mice were exposed to CIH for 8 weeks, and their social novelty discrimination, spatial learning and memory were severely compromised. Additionally, seriously damaged neurons and abnormally activated glia were observed in the CA1 and dentate gyrus (DG) areas of the hippocampus. Mechanistically, knocking out the TLR2 gene significantly alleviated these pathological changes and improved the behavioral performance. Together, these findings demonstrate that the TLR2-MyD88 signaling pathway might play an important role in CIH-induced cognitive deficits.


Subject(s)
Cognitive Dysfunction/metabolism , Hypoxia/metabolism , Neurogenic Inflammation/metabolism , Neurons/pathology , Sleep Apnea, Obstructive/metabolism , Toll-Like Receptor 2/metabolism , Animals , Apoptosis , Behavior, Animal , Cognitive Dysfunction/immunology , Disease Models, Animal , Humans , Hypoxia/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenic Inflammation/immunology , Signal Transduction , Sleep Apnea, Obstructive/immunology , Toll-Like Receptor 2/genetics
20.
PLoS One ; 15(1): e0227817, 2020.
Article in English | MEDLINE | ID: mdl-31971954

ABSTRACT

Besides monocyte (MO)-derived macrophages (MACs), self-renewing tissue-resident macrophages (trMACs) maintain the intracutaneous MAC pool in murine skin. Here, we have asked whether the same phenomenon occurs in human skin using organ-cultured, full-thickness skin detached from blood circulation and bone marrow. Skin stimulation ex vivo with the neuropeptide substance P (SP), mimicking neurogenic skin inflammation, significantly increased the number of CD68+MACs in the papillary dermis without altering intracutaneous MAC proliferation or apoptosis. Since intraluminal CD14+MOs were undetectable in the non-perfused dermal vasculature, new MACs must have differentiated from resident intracutaneous progenitor cells in human skin. Interestingly, CD68+MACs were often seen in direct cell-cell-contact with cells expressing both, the hematopoietic stem cell marker CD34 and SP receptor (neurokinin-1 receptor [NK1R]). These cell-cell contacts and CD34+cell proliferation were up-regulated in SP-treated skin samples. Collectively, our study provides the first evidence that resident MAC progenitors, from which mature MACs can rapidly differentiate within the tissue, do exist in normal adult human skin. That these NK1R+trMAC-progenitor cells quickly respond to a key stress-associated neuroinflammatory stimulus suggests that this may satisfy increased local MAC demand under conditions of wounding/stress.


Subject(s)
Macrophages/immunology , Neurogenic Inflammation/immunology , Skin/immunology , Stem Cells/immunology , Substance P/immunology , Adult , Antigens, CD/immunology , Antigens, CD34/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Apoptosis , Cell Differentiation , Female , Humans , Macrophages/cytology , Organ Culture Techniques , Skin/cytology , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...