Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 817
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
Trends Immunol ; 45(5): 371-380, 2024 May.
Article in English | MEDLINE | ID: mdl-38653601

ABSTRACT

Peripheral sensory neurons recognize diverse noxious stimuli, including microbial products and allergens traditionally thought to be targets of the mammalian immune system. Activation of sensory neurons by these stimuli leads to pain and itch responses as well as the release of neuropeptides that interact with their cognate receptors expressed on immune cells, such as dendritic cells (DCs). Neuronal control of immune cell function through neuropeptide release not only affects local inflammatory responses but can impact adaptive immune responses through downstream effects on T cell priming. Numerous neuropeptide receptors are expressed by DCs but only a few have been characterized, presenting opportunities for further investigation of the pathways by which cutaneous neuroimmune interactions modulate host immunity.


Subject(s)
Sensory Receptor Cells , Skin , Humans , Animals , Sensory Receptor Cells/immunology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Skin/immunology , Neuropeptides/metabolism , Neuropeptides/immunology , Dendritic Cells/immunology , Neuroimmunomodulation , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/immunology
3.
Fish Shellfish Immunol ; 121: 142-151, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34998986

ABSTRACT

Crustacean cardioactive peptide (CCAP) is a pleiotropic neuropeptide, but its immunomodulatory role is not clear. Herein, the mud crab Scylla paramamosain provides a primitive model to study crosstalk between the neuroendocrine and immune systems. In this study, in situ hybridization showed that Sp-CCAP positive signal localized in multiple cells in the nervous tissue, while its conjugate receptor (Sp-CCAPR) positive signal mainly localized in the semigranular cells of hemocytes. The Sp-CCAP mRNA expression level in the thoracic ganglion was significantly up-regulated after lipopolysaccharide (LPS) stimulation, but the Sp-CCAP mRNA expression level was up-regulated firstly and then down-regulated after the stimulation of polyriboinosinic polyribocytidylic acid [Poly (I:C)]. After the injection of Sp-CCAP synthesis peptide, the phagocytosis ability of hemocytes was significantly higher than that of synchronous control group. Simultaneously, the mRNA expression of phagocytosis related gene (Sp-Rab5), nuclear transcription factor NF-κB homologues (Sp-Relish), C-type lectin (Sp-CTL-B), prophenoloxidase (Sp-proPO), pro-inflammatory cytokines factor (Sp-TNFSF, Sp-IL16) and antimicrobial peptides (Sp-ALF1 and Sp-ALF5) in the hemocytes were also significantly up-regulated at different time points after the injection of Sp-CCAP synthetic peptide, but Sp-TNFSF, Sp-ALF1 and Sp-ALF5 were down-regulated significantly at 24h. In addition, RNA interference of Sp-CCAP suppressed the phagocytic activity of hemocytes and inhibited the mRNA expression of Sp-Rab5, Sp-Relish, Sp-CTL-B, Sp-TNFSF, Sp-IL16 and Sp-ALF5 in the hemocytes, and ultimately weakened the ability of hemolymph bacteria clearance of mud crab. Taken together, these results revealed that CCAP induced innate immune and increased the anti-infection ability in the mud crab.


Subject(s)
Arthropod Proteins/immunology , Brachyura , Immunity, Innate , Neuropeptides , Animals , Brachyura/genetics , Brachyura/immunology , Interleukin-16 , Neuropeptides/immunology , Phylogeny , Poly I-C/pharmacology , RNA, Messenger/genetics
4.
Mar Drugs ; 20(2)2022 Feb 17.
Article in English | MEDLINE | ID: mdl-35200674

ABSTRACT

Antimicrobial peptides (AMPs) are found widespread in nature and possess antimicrobial and immunomodulatory activities. Due to their multifunctional properties, these peptides are a focus of growing body of interest and have been characterized in several fish species. Due to their similarities in amino-acid composition and amphipathic design, it has been suggested that neuropeptides may be directly involved in the innate immune response against pathogen intruders. In this review, we report the molecular characterization of the fish-specific AMP piscidin1, the production of an antibody raised against this peptide and the immunohistochemical identification of this peptide and enkephalins in the neuroepithelial cells (NECs) in the gill of several teleost fish species living in different habitats. In spite of the abundant literature on Piscidin1, the biological role of this peptide in fish visceral organs remains poorly explored, as well as the role of the neuropeptides in neuroimmune interaction in fish. The NECs, by their role as sensors of hypoxia changes in the external environments, in combination with their endocrine nature and secretion of immunomodulatory substances would influence various types of immune cells that contain piscidin, such as mast cells and eosinophils, both showing interaction with the nervous system. The discovery of piscidins in the gill and skin, their diversity and their role in the regulation of immune response will lead to better selection of these immunomodulatory molecules as drug targets to retain antimicrobial barrier function and for aquaculture therapy in the future.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Fish Proteins/metabolism , Neuropeptides/metabolism , Animals , Antimicrobial Cationic Peptides/immunology , Aquaculture , Fish Proteins/immunology , Fishes , Gills/metabolism , Humans , Immunity, Innate/immunology , Neuropeptides/immunology , Skin/metabolism
5.
Immunol Rev ; 282(1): 232-247, 2018 03.
Article in English | MEDLINE | ID: mdl-29431202

ABSTRACT

This review presents evidence that the skin mast cell, in particular the MCTC subtype, is the primary effector cell in urticaria. Mast cells are located in the upper dermis, the ideal situation for wheal formation and sensory nerve stimulation. Increased numbers of mast cells are found in both lesional and non-lesional skin in CSU and inducible urticaria. Mast cell degranulation in the area of wheals has been demonstrated repeatedly by light and electron microscopy. Histamine, PGD2 and tryptase are found in the venous blood draining wheal formation. The last 2 are specific for mast cells rather than basophils. Mast cell reactivity is increased in active urticaria by local inflammatory cytokines and neuropeptides. Mast cell cytokines and neuropeptides, particularly nerve growth factor, induce a Th2 type inflammation that is particularly obvious at the sites of whealing. In conclusion, autoimmunity, either of Type 1 viz. IgE antibodies to local autoallergens, or Type 2b, viz. IgG autoantibodies to IgE or its receptor, are considered to be the most frequent causes of CSU. In both cases, the mast cell is likely to be the axial cell in producing the wheals.


Subject(s)
Inflammation Mediators/immunology , Mast Cells/immunology , Neuropeptides/immunology , Urticaria/immunology , Animals , Autoimmunity , Cytokines/metabolism , Histamine/metabolism , Humans , Tryptases/metabolism
6.
Nat Immunol ; 10(3): 249-56, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19198592

ABSTRACT

After being infected by the fungus Drechmeria coniospora, Caenorhabditis elegans produces antimicrobial peptides in its epidermis, some regulated by a signaling cascade involving a p38 mitogen-activated protein kinase. Here we show that infection-induced expression of peptides of the Caenacin family occurred independently of the p38 pathway. The caenacin (cnc) genes enhanced survival after fungal infection, and neuronal expression of the transforming growth factor-beta homolog DBL-1 promoted cnc-2 expression in the epidermis in a dose-dependent paracrine way. Our results lead to a model in which antifungal defenses are coordinately regulated by a cell-autonomous p38 cascade and a distinct cytokine-like transforming growth factor-beta signal from the nervous system, each of which controls distinct sets of antimicrobial peptide-encoding genes in the epidermis.


Subject(s)
Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans/immunology , Epidermis/immunology , Neuroimmunomodulation , Neuropeptides/immunology , Transforming Growth Factor beta/immunology , Animals , Animals, Genetically Modified , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/metabolism , Epidermis/metabolism , Gene Expression Regulation , Hypocreales/pathogenicity , Multigene Family , Neuropeptides/metabolism , RNA, Helminth/metabolism , Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Nat Immunol ; 10(11): 1208-14, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19767757

ABSTRACT

In response to invading microorganisms, macrophages engage in phagocytosis and rapidly release reactive oxygen species (ROS), which serve an important microbicidal function. However, how phagocytosis induces ROS production remains largely unknown. CARD9, a caspase-recruitment domain (CARD)-containing protein, is important for resistance to fungal and bacterial infection. The mechanism of CARD9-mediated bacterial clearance is still mostly unknown. Here we show that CARD9 is required for killing intracellular bacteria in macrophages. CARD9 associated with the GDP-dissociation inhibitor LyGDI in phagosomes after bacterial and fungal infection and binding of CARD9 suppressed LyGDI-mediated inhibition of the GTPase Rac1, thereby leading to ROS production and bacterial killing in macrophages. Thus, our studies identify a key pathway that leads to microbe-elicited ROS production.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Macrophages/immunology , Neuropeptides/immunology , Phagosomes/immunology , Proteins/immunology , Reactive Oxygen Species/immunology , rac GTP-Binding Proteins/immunology , Animals , CARD Signaling Adaptor Proteins , Candida albicans/immunology , Cell Line , Guanine Nucleotide Dissociation Inhibitors/immunology , Guanine Nucleotide Dissociation Inhibitors/metabolism , Immunity, Innate , Listeria monocytogenes/immunology , Macrophages/microbiology , Mice , Mice, Knockout , Neuropeptides/metabolism , Phagosomes/microbiology , Proteins/metabolism , Reactive Oxygen Species/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein , rho-Specific Guanine Nucleotide Dissociation Inhibitors
8.
Ann Neurol ; 87(6): 869-884, 2020 06.
Article in English | MEDLINE | ID: mdl-32196746

ABSTRACT

OBJECTIVE: Assess occurrence of the dendritic spine scaffolding protein Drebrin as a pathophysiologically relevant autoantibody target in patients with recurrent seizures and suspected encephalitis as leading symptoms. METHODS: Sera of 4 patients with adult onset epilepsy and suspected encephalitis of unresolved etiology and equivalent results in autoantibody screening were subjected to epitope identification. We combined a wide array of approaches, ranging from immunoblotting, immunoprecipitation, mass spectrometry, subcellular binding pattern analyses in primary neuronal cultures, and immunohistochemistry in brains of wild-type and Drebrin knockout mice to in vitro analyses of impaired synapse formation, morphology, and aberrant neuronal excitability by antibody exposure. RESULTS: In the serum of a patient with adult onset epilepsy and suspected encephalitis, a strong signal at ∼70kDa was detected by immunoblotting, for which mass spectrometry revealed Drebrin as the putative antigen. Three other patients whose sera also showed strong immunoreactivity around 70kDa on Western blotting were also anti-Drebrin-positive. Seizures, memory impairment, and increased protein content in cerebrospinal fluid occurred in anti-Drebrin-seropositive patients. Alterations in cerebral magnetic resonance imaging comprised amygdalohippocampal T2-signal increase and hippocampal sclerosis. Diagnostic biopsy revealed T-lymphocytic encephalitis in an anti-Drebrin-seropositive patient. Exposure of primary hippocampal neurons to anti-Drebrin autoantibodies resulted in aberrant synapse composition and Drebrin distribution as well as increased spike rates and the emergence of burst discharges reflecting network hyperexcitability. INTERPRETATION: Anti-Drebrin autoantibodies define a chronic syndrome of recurrent seizures and neuropsychiatric impairment as well as inflammation of limbic and occasionally cortical structures. Immunosuppressant therapies should be considered in this disorder. ANN NEUROL 2020;87:869-884.


Subject(s)
Autoantibodies/immunology , Encephalitis/immunology , Neuropeptides/immunology , Seizures/immunology , Adult , Aged , Animals , Encephalitis/diagnostic imaging , Epitopes/immunology , Female , Hippocampus/immunology , Hippocampus/pathology , Humans , Magnetic Resonance Imaging , Male , Mental Disorders/immunology , Mental Disorders/psychology , Mice, Knockout , Middle Aged , Neuroimaging , Seizures/diagnostic imaging , Synapses/immunology , Young Adult
9.
J Immunol ; 202(9): 2529-2534, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30936294

ABSTRACT

Systemic lupus erythematosus severity correlates with elevated serum levels of type I IFNs, cytokines produced in large quantities by plasmacytoid dendritic cells (pDC) in response to engagement of TLR7 and TLR9 with endocytosed nucleic acids. B cell adaptor for PI3K (BCAP) promoted many aspects of TLR7-driven lupus-like disease, including Isg15 and Ifit1 expression in blood and an immature pDC phenotype associated with higher IFN production. BCAP-/- mice produced significantly less serum IFN-α than wild-type mice after injection of TLR9 agonist, and BCAP promoted TLR7 and TLR9-induced IFN-α production specifically in pDC. TLR-induced IFN-α production in pDC requires DOCK2-mediated activation of Rac1 leading to activation of IKKα, a mechanism we show was dependent on BCAP. BCAP-/- pDC had decreased actin polymerization and Rac1 activation and reduced IKKα phosphorylation upon TLR9 stimulation. We show a novel role for BCAP in promoting TLR-induced IFN-α production in pDC and in systemic lupus erythematosus pathogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Dendritic Cells/immunology , Interferon-alpha/immunology , Lupus Erythematosus, Systemic/immunology , Membrane Glycoproteins/immunology , Plasma Cells/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/pathology , Female , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/immunology , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/immunology , Interferon-alpha/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Neuropeptides/genetics , Neuropeptides/immunology , Plasma Cells/pathology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 9/genetics , Ubiquitins/genetics , Ubiquitins/immunology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/immunology
10.
Int J Mol Sci ; 22(7)2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33915818

ABSTRACT

The regulation of infection and inflammation by a variety of host peptides may represent an evolutionary failsafe in terms of functional degeneracy and it emphasizes the significance of host defense in survival. Neuropeptides have been demonstrated to have similar antimicrobial activities to conventional antimicrobial peptides with broad-spectrum action against a variety of microorganisms. Neuropeptides display indirect anti-infective capacity via enhancement of the host's innate and adaptive immune defense mechanisms. However, more recently concerns have been raised that some neuropeptides may have the potential to augment microbial virulence. In this review we discuss the dual role of neuropeptides, perceived as a double-edged sword, with antimicrobial activity against bacteria, fungi, and protozoa but also capable of enhancing virulence and pathogenicity. We review the different ways by which neuropeptides modulate crucial stages of microbial pathogenesis such as adhesion, biofilm formation, invasion, intracellular lifestyle, dissemination, etc., including their anti-infective properties but also detrimental effects. Finally, we provide an overview of the efficacy and therapeutic potential of neuropeptides in murine models of infectious diseases and outline the intrinsic host factors as well as factors related to pathogen adaptation that may influence efficacy.


Subject(s)
Infections/immunology , Neuropeptides/immunology , Animals , Humans , Infections/microbiology , Infections/therapy , Molecular Targeted Therapy , Virulence
11.
Int J Mol Sci ; 22(22)2021 Nov 16.
Article in English | MEDLINE | ID: mdl-34830245

ABSTRACT

Although histamine is a well-known itch mediator, histamine H1-receptor blockers often lack efficacy in chronic itch. Recent molecular and cellular based studies have shown that non-histaminergic mediators, such as proteases, neuropeptides and cytokines, along with their cognate receptors, are involved in evocation and modulation of itch sensation. Many of these molecules are produced and secreted by immune cells, which act on sensory nerve fibers distributed in the skin to cause itching and sensitization. This understanding of the connections between immune cell-derived mediators and sensory nerve fibers has led to the development of new treatments for itch. This review summarizes current knowledge of immune cell-derived itch mediators and neuronal response mechanisms, and discusses therapeutic agents that target these systems.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Histamine/immunology , Immunologic Factors/therapeutic use , Pruritus/immunology , Receptors, Histamine H1/immunology , Sensory Receptor Cells/immunology , Antibodies, Monoclonal/therapeutic use , Cytokines/antagonists & inhibitors , Cytokines/immunology , Cytokines/metabolism , Gene Expression , Histamine/metabolism , Histamine Antagonists/therapeutic use , Humans , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/pathology , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/pathology , Neuropeptides/antagonists & inhibitors , Neuropeptides/immunology , Neuropeptides/metabolism , Peptide Hydrolases/immunology , Peptide Hydrolases/metabolism , Protease Inhibitors/therapeutic use , Pruritus/drug therapy , Pruritus/genetics , Pruritus/pathology , Receptors, Histamine H1/genetics , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/pathology , Skin/drug effects , Skin/immunology , Skin/innervation , Skin/pathology
12.
Eur J Immunol ; 49(11): 1984-1997, 2019 11.
Article in English | MEDLINE | ID: mdl-31327163

ABSTRACT

The body is innervated by a meshwork of heterogeneous peripheral neurons (including sensory neurons) which project virtually to all the organs. Peripheral neurons have been studied extensively in the context of their primary function of initiation of voluntary and involuntary movement, transmission of sensations and induction of appropriate behavioral response such as withdrawal to avoid tissue injury or scratching to remove irritating molecules. More recently, breakthrough articles have shown that, on top of their primary function of signal transmission to the spinal cord and brain, peripheral neurons (including afferent neurons) could directly sense environmental alarms and consequently regulate the development of various type of immune responses through the release of neuropeptides or growth factors. In this review, we discuss recent advances in the neural regulation of the immune response, both in physiological and pathological contexts by taking into account the type of organs (lungs, skin and gut), subtypes of peripheral neurons (sympathetic, nociceptive and intrinsic gut neurons) or immune cells and strains of pathogens studied. We also highlight future challenges in the field and potential therapeutic innovations targeting neuro-immune interactions.


Subject(s)
Gastrointestinal Tract/immunology , Immunity, Mucosal , Peripheral Nervous System/immunology , Sensory Receptor Cells/immunology , Skin/immunology , Synaptic Transmission/immunology , Animals , Bacterial Infections/immunology , Bacterial Infections/metabolism , Bacterial Infections/microbiology , Brain/immunology , Brain/metabolism , Cytokines/immunology , Cytokines/metabolism , Gastrointestinal Tract/metabolism , Humans , Immunity, Innate , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Lung/immunology , Lung/metabolism , Neuropeptides/immunology , Neuropeptides/metabolism , Nociception/physiology , Peripheral Nervous System/metabolism , Sensory Receptor Cells/metabolism , Skin/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism
13.
J Neuroinflammation ; 17(1): 356, 2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33239034

ABSTRACT

The existence of the neural control of mast cell functions has long been proposed. Mast cells (MCs) are localized in association with the peripheral nervous system (PNS) and the brain, where they are closely aligned, anatomically and functionally, with neurons and neuronal processes throughout the body. They express receptors for and are regulated by various neurotransmitters, neuropeptides, and other neuromodulators. Consequently, modulation provided by these neurotransmitters and neuromodulators allows neural control of MC functions and involvement in the pathogenesis of mast cell-related disease states. Recently, the roles of individual neurotransmitters and neuropeptides in regulating mast cell actions have been investigated extensively. This review offers a systematic review of recent advances in our understanding of the contributions of neurotransmitters and neuropeptides to mast cell activation and the pathological implications of this regulation on mast cell-related disease states, though the full extent to which such control influences health and disease is still unclear, and a complete understanding of the mechanisms underlying the control is lacking. Future validation of animal and in vitro models also is needed, which incorporates the integration of microenvironment-specific influences and the complex, multifaceted cross-talk between mast cells and various neural signals. Moreover, new biological agents directed against neurotransmitter receptors on mast cells that can be used for therapeutic intervention need to be more specific, which will reduce their ability to support inflammatory responses and enhance their potential roles in protecting against mast cell-related pathogenesis.


Subject(s)
Mast Cells/immunology , Neurons/immunology , Neuropeptides/immunology , Neurotransmitter Agents/immunology , Receptors, Neurotransmitter/immunology , Animals , Brain/immunology , Brain/metabolism , Humans , Mast Cells/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Receptors, Neurotransmitter/metabolism
14.
Nat Immunol ; 9(12): 1415-24, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18854822

ABSTRACT

Communication between the immune and nervous systems, each of which is able to react rapidly to environmental stimuli, may confer a survival advantage. However, precisely how the nervous system influences the immune response and whether neural modulation of immune function is biologically important are not well understood. Here we report that neuronal exocytosis of neuropeptides from dense core vesicles suppressed the survival of Caenorhabditis elegans and their clearance of infection with the human bacterial pathogen Pseudomonas aeruginosa. This immunomodulatory function was mediated by INS-7, an insulin-like neuropeptide whose induction was associated with Pseudomonas virulence. INS-7 secreted from the nervous system functioned in a non-cell autonomous way to activate the insulin pathway and alter basal and inducible expression of immunity-related genes in intestinal cells.


Subject(s)
Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans/immunology , Immunity, Innate , Neurosecretory Systems/immunology , Peptide Hormones/immunology , Signal Transduction/immunology , Animals , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/metabolism , Exocytosis/immunology , Insulin/metabolism , Neuropeptides/immunology , Neuropeptides/metabolism , Peptide Hormones/metabolism , Pseudomonas Infections/immunology , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Secretory Vesicles/immunology , Secretory Vesicles/metabolism
15.
Exp Dermatol ; 29(8): 703-725, 2020 08.
Article in English | MEDLINE | ID: mdl-32682334

ABSTRACT

Anagen stage hair follicles (HFs) exhibit "immune privilege (IP)" from the level of the bulge downwards to the bulb. Both passive and active IP mechanisms protect HFs from physiologically undesired immune responses and limit immune surveillance. IP is relative, not absolute, and is primarily based on absent, or greatly reduced, intra-follicular antigen presentation via MHC class I and II molecules, along with prominent expression of "no danger" signals like CD200 and the creation of an immunoinhibitory signalling milieu generated by the secretory activities of HFs. Perifollicular mast cells, Tregs and other immunocytes may also contribute to HF IP maintenance in healthy human skin. Collapse of anagen hair bulb IP is an essential prerequisite for the development of alopecia areata (AA). In AA, lesional HFs are rapidly infiltrated by NKG2D + T cells and natural killer (NK) cells, while perifollicular mast cells acquire a profoundly pro-inflammatory phenotype and interact with autoreactive CD8+ T cells. Using animal models, significant functional evidence has accumulated that demonstrates the dominance of the immune system in AA pathogenesis. Purified CD8+T-cell and NK cell populations alone, which secrete fγ, suffice to induce the AA phenotype, while CD4+T-cells aggravate it, and Tregs and iNKT cells may provide relative protection against AA development. While IP collapse may be induced by exogenous agents, inherent IP deficiencies might confer increased susceptibility to AA for some individuals. Thus, a key goal for effective AA management is the re-establishment of a functional HF IP, which will also provide superior protection from disease relapse.


Subject(s)
Alopecia Areata/immunology , Alopecia Areata/therapy , Cytokines/immunology , Hair Follicle/immunology , Immune Privilege , Alopecia Areata/metabolism , Animals , B7-H1 Antigen/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Mast Cells/immunology , Neuropeptides/immunology , Signal Transduction , T-Lymphocytes, Regulatory/immunology
16.
Fish Shellfish Immunol ; 101: 244-251, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32272259

ABSTRACT

Short neuropeptide F (sNPF), a highly conserved neuropeptide, displays pleiotropic functions on multiple aspects of physiological processes, such as feeding, metabolic stress, locomotion, circadian clock and reproduction. However, to date there has no any report on the possible immunoregulation of sNPF in crustaceans. In the present study, we found that the Sp-sNPF was mainly expressed in the nervous tissue in the mud crab Scylla paramamosain, while the sNPF receptor gene (Sp-sNPF-R) was expressed in a wide variety of tissues, including the hepatopancreas. In situ hybridization further showed that the Sp-sNPF-R positive signal mainly localized in the F-cells of the hepatopancreas. Moreover, the Sp-sNPF-R transcription could be significantly up-regulated after the challenge of bacteria-analog LPS or virus-analog Poly (I:C). Both in vitro and in vivo experiments showed that the synthetic sNPF peptide significantly increased the gene expressions of sNPF-R, nuclear factor-κB (NF-κB) signaling genes and antimicrobial peptides (AMPs) in the hepatopancreas. Simultaneously, the administration of sNPF peptide in vitro also increased the concentration of nitric oxide (NO) and the bacteriostasis of the culture medium of hepatopancreas. These results indicated that sNPF up-regulated hepatopancreas immune responses, which may bring new insight into the neuroendocrine-immune regulatory system in crustacean species, and could potentially provide a new strategy for disease prevention and control for mud crab aquaculture.


Subject(s)
Arthropod Proteins/immunology , Brachyura/immunology , Hepatopancreas/immunology , Immunity, Innate/genetics , Neuropeptides/immunology , Animals , Brachyura/genetics , Female
17.
J Immunol ; 200(11): 3697-3710, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29669783

ABSTRACT

Vasoactive intestinal peptide (VIP) is a neuropeptide that exerts various vascular and cardioprotective functions and regulates immune function and inflammatory response at multiple levels. However, its role in inflammatory cardiovascular disorders is largely unknown. Myocarditis and atherosclerosis are two inflammatory and autoimmune cardiovascular diseases that cause important adverse circulatory events. In this study, we investigate the therapeutic effects of VIP in various well-established preclinical models of experimental autoimmune myocarditis and atherosclerosis. Intraperitoneal injection of VIP during the effector phase of experimental autoimmune myocarditis in susceptible BALB/c mice significantly reduced its prevalence, ameliorated signs of heart hypertrophy and injury, attenuated myocardial inflammatory infiltration, and avoided subsequent profibrotic cardiac remodeling. This effect was accompanied by a reduction of Th17-driven cardiomyogenic responses in peripheral lymphoid organs and in the levels of myocardial autoantibodies. In contrast, acute and chronic atherosclerosis was induced in apolipoprotein E-deficient mice fed a hyperlipidemic diet and subjected to partial carotid ligation. Systemic VIP treatment reduced the number and size of atherosclerotic plaques in carotid, aorta, and sinus in hypercholesterolemic mice. VIP reduced Th1-driven inflammatory responses and increased regulatory T cells in atherosclerotic arteries and their draining lymph nodes. VIP also regulated cholesterol efflux in macrophages and reduced the formation of foam cells and their presence in atherosclerotic plaques. Finally, VIP inhibited proliferation and migration of smooth muscle cells and neointima formation in a mouse model of complete carotid ligation. These findings encourage further studies aimed to assess whether VIP can be used as a pharmaceutical agent to treat heart inflammation and atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Autoimmune Diseases/immunology , Autoimmunity/immunology , Inflammation/immunology , Myocarditis/immunology , Vasoactive Intestinal Peptide/immunology , Animals , Apolipoproteins E/immunology , Autoantibodies/immunology , Disease Models, Animal , Female , Lymph Nodes/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscle, Smooth/immunology , Myocardium/immunology , Neuropeptides/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
18.
Int J Mol Sci ; 21(21)2020 Oct 22.
Article in English | MEDLINE | ID: mdl-33105700

ABSTRACT

Members of neuropeptide B/W signaling system have been predominantly detected and mapped within the CNS. In the rat, this system includes neuropeptide B (NPB), neuropeptide W (NPW) and their specific receptor NPBWR1. This signaling system has a wide spectrum of functions including a role in modulation of inflammatory pain and neuroendocrine functions. Expression of NPB, NPW and NPBWR1 in separate heart compartments, dorsal root ganglia (DRG) and stellate ganglia was proven by RT-qPCR, Western blot (WB) and immunofluorescence. Presence of mRNA for all tested genes was detected within all heart compartments and ganglia. The presence of proteins preproNPB, preproNPW and NPBWR1 was confirmed in all the chambers of heart by WB. Expression of preproNPW and preproNPB was proven in cardiac ganglionic cells obtained by laser capture microdissection. In immunofluorescence analysis, NPB immunoreactivity was detected in nerve fibers, some nerve cell bodies and smooth muscle within heart and both ganglia. NPW immunoreactivity was present in the nerve cell bodies and nerve fibers of heart ganglia. Weak nonhomogenous staining of cardiomyocytes was present within heart ventricles. NPBWR1 immunoreactivity was detected on cardiomyocytes and some nerve fibers. We confirmed the presence of NPB/W signaling system in heart, DRG and stellate ganglia by proteomic and genomic analyses.


Subject(s)
Myocardium/metabolism , Neuropeptides/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Animals , Fluorescent Antibody Technique , Ganglia, Spinal/metabolism , Gene Expression , Male , Neuropeptides/immunology , Neuropeptides/metabolism , Rats, Zucker , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/immunology , Reproducibility of Results , Signal Transduction , Stellate Ganglion/metabolism
19.
Immunology ; 156(2): 136-146, 2019 02.
Article in English | MEDLINE | ID: mdl-30317558

ABSTRACT

Phagocytosis, macropinocytosis and antigen presentation by dendritic cells (DC) requires reorganization of the actin cytoskeleton. Drebrin (Dbn1) is an actin binding and stabilizing protein with roles in endocytosis, formation of dendrite spines in neurons and coordinating cell-cell synapses in immune cells. However, its role in DC phagocytosis and antigen presentation is unknown. These studies now report that silencing of Dbn1 in DC resulted in restrained cell surface display of receptors, most notably MHC class I and II and co-stimulatory molecules. This, as expected, resulted in impaired antigen-specific T-cell activation and proliferation. Studies additionally revealed that knockdown of Dbn1 in DC impaired macropinocytosis and phagocytosis. However, there was a concomitant increase in fluid-phase uptake, suggesting that Dbn1 is responsible for the differential control of macropinocytosis versus micropinocytosis activities. Taken together, these findings now reveal that Dbn1 plays a major role in coordinating the actin cytoskeletal activities responsible for antigen presentation in DC.


Subject(s)
Antigen Presentation , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class I/immunology , Neuropeptides/immunology , Phagocytosis , Animals , Cytoskeleton/genetics , Cytoskeleton/immunology , Dendritic Cells/cytology , Gene Knockout Techniques , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class II/genetics , Immunological Synapses/genetics , Immunological Synapses/immunology , Lymphocyte Activation/genetics , Mice , Mice, Transgenic , Neuropeptides/genetics , T-Lymphocytes/cytology , T-Lymphocytes/immunology
20.
J Cell Sci ; 130(15): 2530-2540, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28600322

ABSTRACT

Lamellipodia are sheet-like cell protrusions driven by actin polymerization mainly through Rac1, a GTPase molecular switch. In Fcγ receptor-mediated phagocytosis of IgG-opsonized erythrocytes (IgG-Es), Rac1 activation is required for lamellipodial extension along the surface of IgG-Es. However, the significance of Rac1 deactivation in phagosome formation is poorly understood. Our live-cell imaging and electron microscopy revealed that RAW264 macrophages expressing a constitutively active Rac1 mutant showed defects in phagocytic cup formation, while lamellipodia were formed around IgG-Es. Because activated Rac1 reduced the phosphorylation levels of myosin light chains, failure of the cup formation is probably due to inhibition of actin/myosin II contractility. Reversible photo-manipulation of the Rac1 switch in macrophages fed with IgG-Es could phenocopy two lamellipodial motilities: outward-extension and cup-constriction by Rac1 ON and OFF, respectively. In conjunction with fluorescence resonance energy transfer imaging of Rac1 activity, we provide a novel mechanistic model of phagosome formation spatiotemporally controlled by Rac1 switching within a phagocytic cup.


Subject(s)
Macrophages/immunology , Models, Immunological , Neuropeptides/immunology , Phagosomes/immunology , Receptors, IgG/immunology , rac1 GTP-Binding Protein/immunology , Animals , Mice , Phosphorylation/immunology , RAW 264.7 Cells
SELECTION OF CITATIONS
SEARCH DETAIL