Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Immunother Adv ; 2(1): ltac010, 2022.
Article in English | MEDLINE | ID: mdl-36284839

ABSTRACT

Macrophages are immune cells that are widespread throughout the body and critical for maintaining tissue homeostasis. Their remarkable plasticity allows them to acquire different phenotypes, becoming able either to fight infection (M1-like, classically activated macrophages) or to promote tissue remodeling and repair (M2-like, alternatively activated macrophages). These phenotypes are induced by different cues present in the microenvironment. Among the factors that might regulate macrophage activation are mediators produced by different branches of the nervous system. The regulation exerted by the sympathetic nervous system (SNS) on macrophages (and the immune system in general) is becoming a subject of increasing interest, indeed a great number of articles have been published lately. Catecholamines (noradrenaline and adrenaline) activate α and ß adrenergic receptors expressed by macrophages and shape the effector functions of these cells in contexts as diverse as the small intestine, the lung, or the adipose tissue. Activation of different subsets of receptors seems to produce antagonistic effects, with α adrenergic receptors generally associated with pro-inflammatory functions and ß adrenergic receptors (particularly ß2) related to the resolution of inflammation and tissue remodeling. However, exceptions to this paradigm have been reported, and the factors contributing to these apparently contradictory observations are still far from being completely understood. Additionally, macrophages per se seem to be sources of catecholamines, which is also a subject of some debate. In this review, we discuss how activation of adrenergic receptors modulates macrophage effector functions and its implications for inflammatory responses and tissue homeostasis.

2.
Brain Behav Immun ; 90: 208-215, 2020 11.
Article in English | MEDLINE | ID: mdl-32827702

ABSTRACT

Although several studies demonstrate that stressful situations, such as sleep disturbances, negatively impact the innate and adaptive arms of the immune system, their influence on invariant Natural Killer T (iNKT) cells remains unclear. iNKT cells are CD1d-restricted innate T cells that recognize glycolipid antigens and rapidly produce polarizing cytokines being key players in several immune responses, and a potential target for immunotherapy. iNKT cells differ in several aspects from conventional T lymphocytes, including a unique dependence on CD1d-expressing double-positive (DP) thymocytes for intrathymic maturation. As a consequence of stress, DP thymocytes undergo glucocorticoid-induced apoptosis, which might compromise iNKT developmental pathway. Therefore, we used a paradoxical sleep deprivation (SD) model to determine the impact of sleep disturbance on iNKT cell biology. After 72 h of SD, C57Bl/6 mice exhibited a significant increase in systemic glucocorticoid levels and thymus atrophy. Despite marked decrease in the number of DP thymocytes, the ratio CD1d+/CD1d- was higher in SD mice, and the number of thymic iNKT cells remained unaltered, suggesting that SD did not compromise the iNKT developmental pathway. In contrast, SD reduced hepatic IFN-γ, but not, IL-4-producing iNKT cells, without further effect in the spleen. Despite this fact, SD did not affect stimulation of IFN-γ production by iNKT cells, or cytokine release, in response to α-galactosylceramide, a specific antigen. Furthermore, although SD impaired splenic NK cells activity against tumor cells, it did not affect iNKT cell-specific cytotoxicity. Thus, our study shows that SD-induced stress did not impair the iNKT cells' responses to a cognate antigen.


Subject(s)
Natural Killer T-Cells , Animals , Cytokines , Killer Cells, Natural , Mice , Mice, Inbred C57BL , Sleep, REM , Spleen
3.
Cell Rep ; 28(12): 3120-3130.e5, 2019 Sep 17.
Article in English | MEDLINE | ID: mdl-31533035

ABSTRACT

Noradrenaline (NE), the main neurotransmitter released by sympathetic nerve terminals, is known to modulate the immune response. However, the role of the sympathetic nervous system (SNS) on the development of autoimmune diseases is still unclear. Here, we report that the SNS limits the generation of pathogenic T cells and disease development in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis (MS). ß2-Adrenergic receptor (Adrb2) signaling limits T cell autoimmunity in EAE through a mechanism mediated by the suppression of IL-2, IFN-γ, and GM-CSF production via inducible cAMP early repressor (ICER). Accordingly, the lack of Adrb2 signaling in immune cells is sufficient to abrogate the suppressive effects of SNS activity, resulting in increased pathogenic T cell responses and EAE development. Collectively, these results uncover a suppressive role for the SNS in CNS autoimmunity while they identify potential targets for therapeutic intervention.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Immunity, Cellular , Multiple Sclerosis/immunology , Receptors, Adrenergic, beta-2/immunology , Signal Transduction/immunology , Sympathetic Nervous System/immunology , T-Lymphocytes/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Receptors, Adrenergic, beta-2/genetics , Signal Transduction/genetics , Sympathetic Nervous System/pathology , T-Lymphocytes/pathology
4.
Semin Immunopathol ; 39(2): 165-176, 2017 02.
Article in English | MEDLINE | ID: mdl-27800584

ABSTRACT

Dendritic cells are of paramount importance bridging innate and adaptive immune responses. Depending on the context, after sensing environmental antigens, commensal microorganisms, pathogenic agents, or antigens from the diet, dendritic cells may drive either different effector adaptive immune responses or tolerance, avoiding tissue damage. Although the plasticity of the immune response and the capacity to regulate itself are considered essential to orchestrate appropriate physiological responses, it is known that the nervous system plays a relevant role controlling immune cell function. Dendritic cells present in the skin, the intestine, and lymphoid organs, besides expressing adrenergic receptors, can be reached by neurotransmitters released by sympathetic fibers innervating these tissues. These review focus on how neurotransmitters from the sympathetic nervous system can modulate dendritic cell function and how this may impact the immune response and immune-mediated disorders.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Immune System/cytology , Immune System/physiology , Neuroimmunomodulation , Sympathetic Nervous System/physiology , Animals , Cytokines/metabolism , Humans , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/innervation , Lymphoid Tissue/immunology , Lymphoid Tissue/innervation , Lymphoid Tissue/metabolism , Norepinephrine/metabolism , Receptors, Adrenergic/metabolism , Signal Transduction , Skin/immunology , Skin/innervation , Skin/metabolism
5.
Brain Behav Immun ; 37: 177-86, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24362236

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) has been widely employed as a model to study multiple sclerosis (MS) and indeed has allowed some important advances in our comprehension of MS pathogenesis. Several pieces of evidence suggest that infiltrating Th1 and Th17 lymphocytes are important players leading to CNS demyelination and lesion during the peak of murine EAE. Subsequently, effector T cell responses rapidly decline and the recovery phase of the disease strongly correlates with the expression of anti-inflammatory cytokines and the enrichment of Foxp3+ regulatory T (Treg) cells within the target organ. However, the mechanisms leading to the increased presence of Treg cells and to the remission phase of the disease are still poorly understood. Recent researches demonstrated that chemically induced amino-acid starvation response might suppress CNS immune activity. Here we verified an important participation of the general control nonrepressible 2 (GCN2), a key regulator kinase of the amino-acid starvation response, in the development of the remission phase of EAE in C57BL/6 mice. By immunizing wild type C57BL/6 (WT) and GCN2 knock-out mice (GCN2 KO) with myelin oligodendrocyte glycoprotein peptide (MOG35-55), it was noticed that GCN2 KO mice did not develop the remission phase of the disease and this was associated with higher levels of CNS inflammation and increased presence of effector T cells (Th1/Th17). These animals also showed lower frequency of Treg cells within the CNS as compared to WT animals. Higher expression of indoleamine 2,3-dioxygenase (IDO) and higher frequency of plasmacytoid dendritic cells (pDCs) were found at the peak of the disease in the CNS of WT animals. Our results suggest that the GCN2 kinase-dependent sensing of IDO activity represents an important trigger to the EAE remission phase. The IDO-mediated immunoregulatory events may include the arresting of effector T cell responses and the differentiation/expansion of Treg cells within the target organ.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/enzymology , Protein Serine-Threonine Kinases/physiology , Animals , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Forkhead Transcription Factors/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Remission, Spontaneous , Spinal Cord/pathology , Th1 Cells/metabolism , Th17 Cells/metabolism
6.
Eur J Immunol ; 43(4): 1001-12, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23436577

ABSTRACT

Beta2-adrenergic receptor (B2AR) signaling is known to impair Th1-cell differentiation and function in a cAMP-dependent way, leading to inhibition of cell proliferation and decreased production of IL-2 and IFN-γ. CD4(+) Foxp3(+) Treg cells play a key role in the regulation of immune responses and are essential for maintenance of self-tolerance. Nevertheless, very little is known about adrenergic receptor expression in Treg cells or the influence of noradrenaline on their function. Here we show that Foxp3(+) Treg cells express functional B2AR. B2AR activation in Treg cells leads to increased intracellular cAMP levels and to protein kinase A (PKA)-dependent CREB phosphorylation. We also found that signaling via B2AR enhances the in vitro suppressive activity of Treg cells. B2AR-mediated increase in Treg-cell suppressive function was associated with decreased IL-2 mRNA levels in responder CD4(+) T cells and improved Treg-cell-induced conversion of CD4(+) Foxp3(-) cells into Foxp3(+) induced Treg cells. Moreover, B2AR signaling increased CTLA-4 expression in Treg cells in a PKA-dependent way. Finally, we found that PKA inhibition totally prevented the B2AR-mediated increase in Treg-cell suppressive function. Our data suggest that sympathetic fibers are able to regulate Treg-cell suppressive activity in a positive manner through B2AR signaling.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , CD4 Antigens/metabolism , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Forkhead Transcription Factors/metabolism , Interleukin-2/biosynthesis , Lymphocyte Activation/immunology , Mice , Mice, Knockout
7.
Diabetes ; 61(10): 2534-45, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22688334

ABSTRACT

Adipose-derived mesenchymal stem cells (ADMSCs) display immunosuppressive properties, suggesting a promising therapeutic application in several autoimmune diseases, but their role in type 1 diabetes (T1D) remains largely unexplored. The aim of this study was to investigate the immune regulatory properties of allogeneic ADMSC therapy in T cell-mediated autoimmune diabetes in NOD mice. ADMSC treatment reversed the hyperglycemia of early-onset diabetes in 78% of diabetic NOD mice, and this effect was associated with higher serum insulin, amylin, and glucagon-like peptide 1 levels compared with untreated controls. This improved outcome was associated with downregulation of the CD4(+) Th1-biased immune response and expansion of regulatory T cells (Tregs) in the pancreatic lymph nodes. Within the pancreas, inflammatory cell infiltration and interferon-γ levels were reduced, while insulin, pancreatic duodenal homeobox-1, and active transforming growth factor-ß1 expression were increased. In vitro, ADMSCs induced the expansion/proliferation of Tregs in a cell contact-dependent manner mediated by programmed death ligand 1. In summary, ADMSC therapy efficiently ameliorates autoimmune diabetes pathogenesis in diabetic NOD mice by attenuating the Th1 immune response concomitant with the expansion/proliferation of Tregs, thereby contributing to the maintenance of functional ß-cells. Thus, this study may provide a new perspective for the development of ADMSC-based cellular therapies for T1D.


Subject(s)
Adipocytes/transplantation , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Hyperglycemia/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Adipocytes/immunology , Animals , Cell Proliferation , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Glucagon-Like Peptide 1/blood , Hyperglycemia/blood , Hyperglycemia/immunology , Insulin/blood , Islet Amyloid Polypeptide/blood , Mice , Mice, Inbred NOD , T-Lymphocytes, Regulatory/immunology
8.
J Neuroimmunol ; 227(1-2): 10-7, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20580440

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory immune response directed against myelin antigens of the central nervous system. In its murine model, EAE, Th17 cells play an important role in disease pathogenesis. These cells can induce blood-brain barrier disruption and CNS immune cells activation, due to the capacity to secrete high levels of IL-17 and IL-22 in an IL-6+TGF-ß dependent manner. Thus, using the oral tolerance model, by which 200 µg of MOG 35-55 is given orally to C57BL/6 mice prior to immunization, we showed that the percentage of Th17 cells as well as IL-17 secretion is reduced both in the periphery and also in the CNS of orally tolerated animals. Altogether, our data corroborates with the pathogenic role of IL-17 and IFN-γ in EAE, as its reduction after oral tolerance, leads to an overall reduction of pro-inflammatory cytokines, such as IL-1α, IL-6, IL-9, IL-12p70 and the chemokines MIP-1ß, RANTES, Eotaxin and KC in the CNS. It is noteworthy that this was associated to an increase in IL-10 levels. Thus, our data clearly show that disease suppression after oral tolerance induction, correlates with reduction in target organ inflammation, that may be caused by a reduced Th1/Th17 response.


Subject(s)
Allergens/administration & dosage , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Immune Tolerance , Interleukin-17/antagonists & inhibitors , Lymphocyte Depletion , Nerve Tissue Proteins/administration & dosage , T-Lymphocytes, Helper-Inducer/immunology , Administration, Oral , Allergens/immunology , Amino Acid Sequence , Animals , Encephalomyelitis, Autoimmune, Experimental/therapy , Glycoproteins/administration & dosage , Glycoproteins/immunology , Glycoproteins/therapeutic use , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/immunology , Immunosuppressive Agents/therapeutic use , Inflammation/immunology , Inflammation/pathology , Inflammation/therapy , Interleukin-17/metabolism , Lymphocyte Depletion/methods , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Myelin-Oligodendrocyte Glycoprotein , Nerve Tissue Proteins/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Peptide Fragments/therapeutic use , T-Lymphocytes, Helper-Inducer/pathology
9.
Cell Res ; 19(4): 399-411, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19255592

ABSTRACT

For more than two decades, immunologists have been using the so-called Th1/Th2 paradigm to explain most of the phenomena related to adaptive immunity. The Th1/Th2 paradigm implied the existence of two different, mutually regulated, CD4(+) T helper subsets: Th1 cells, driving cell-mediated immune responses involved in tissue damage and fighting infection against intracellular parasites; and Th2 cells that mediate IgE production and are particularly involved in eosinophilic inflammation, allergy and clearance of helminthic infections. A third member of the T helper set, IL-17-producing CD4(+) T cells, now called Th17 cells, was recently described as a distinct lineage that does not share developmental pathways with either Th1 or Th2 cells. The Th17 subset has been linked to autoimmune disorders, being able to produce IL-17, IL-17F and IL-21 among other inflammatory cytokines. Interestingly, it has been reported that there is not only a cross-regulation among Th1, Th2 and Th17 effector cells but there is also a dichotomy in the generation of Th17 and T regulatory cells. Therefore, Treg and Th17 effector cells arise in a mutually exclusive fashion, depending on whether they are activated in the presence of TGF-beta or TGF-beta plus inflammatory cytokines such as IL-6. This review will address the discovery of the Th17 cells, and recent progress on their development and regulation.


Subject(s)
Interleukin-17/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Down-Regulation , Interleukin-23/metabolism , Interleukin-6/metabolism , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Transcription, Genetic , Transforming Growth Factor beta/metabolism
10.
Neuroimmunomodulation ; 16(1): 19-27, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19077442

ABSTRACT

BACKGROUND: Mice allergic to ovalbumin (OVA) avoid drinking a solution containing this antigen. This was interpreted as related to IgE-dependent mast cell degranulation and sensory C fiber activation. METHODS: We employed pharmacological manipulation to further investigate the mediators involved in immune-induced food aversion. RESULTS: While nonimmunized rats preferred a sweetened OVA solution, immunized rats avoided it. We also employed a paradigm in which rats are conditioned to drink water for two 10-min sessions a day. Tolerant rats presented lower IgE titers, and this manipulation abrogated food aversion. Dexamethasone (1.0 mg/kg) prevented the aversion of OVA-immunized rats to the antigen-containing solution. Combined blockade of H(1) and 5-hydroxytryptamine (5-HT)(2) receptors by promethazine (3.0 mg/kg) plus methysergide (5.0 mg/kg) was unable to alter food aversion. Blockade of 5-HT(3) receptors by ondansetron (1.0 mg/kg) caused a twofold increase in the ingestion of the sweetened OVA solution by immunized rats, suggesting the involvement of 5-HT(3) receptors in food aversion. Finally, we showed that dexamethasone or promethazine plus methysergide, but not ondansetron, effectively prevented the IgE-dependent mast-cell-degranulation-induced increase in vascular permeability in rats. CONCLUSION: We suggest that regardless of whether or not they cause edema, IgE-mediated mast cell degranulation and consequent 5-HT(3) signaling are involved in the process that triggers avoidance to the source of the allergen in allergic rats.


Subject(s)
Feeding Behavior , Food Hypersensitivity/immunology , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Capillary Permeability/drug effects , Capillary Permeability/immunology , Cell Degranulation/drug effects , Cell Degranulation/immunology , Conditioning, Psychological/physiology , Dexamethasone/pharmacology , Disease Models, Animal , Food Hypersensitivity/physiopathology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunosuppressive Agents/pharmacology , Male , Mast Cells/drug effects , Mast Cells/immunology , Methysergide/pharmacology , Ondansetron/pharmacology , Ovalbumin/immunology , Rats , Rats, Wistar , Serotonin/metabolism , Serotonin 5-HT3 Receptor Antagonists , Serotonin Antagonists/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL