Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Exp Med ; 217(4)2020 04 06.
Article in English | MEDLINE | ID: mdl-32045471

ABSTRACT

Natural killer (NK) cells are innate lymphocytes that exhibit adaptive features, such as clonal expansion and memory, during viral infection. Although activating receptor engagement and proinflammatory cytokines are required to drive NK cell clonal expansion, additional stimulatory signals controlling their proliferation remain to be discovered. Here, we describe one such signal that is provided by the adrenergic nervous system, and demonstrate that cell-intrinsic adrenergic signaling is required for optimal adaptive NK cell responses. Early during mouse cytomegalovirus (MCMV) infection, NK cells up-regulated Adrb2 (which encodes the ß2-adrenergic receptor), a process dependent on IL-12 and STAT4 signaling. NK cell-specific deletion of Adrb2 resulted in impaired NK cell expansion and memory during MCMV challenge, in part due to a diminished proliferative capacity. As a result, NK cell-intrinsic adrenergic signaling was required for protection against MCMV. Taken together, we propose a novel role for the adrenergic nervous system in regulating circulating lymphocyte responses to viral infection.


Subject(s)
Adrenergic Neurons/immunology , Cytomegalovirus Infections/immunology , Killer Cells, Natural/immunology , Signal Transduction/immunology , Animals , Cell Proliferation/physiology , Cytokines/immunology , Immunologic Memory/immunology , Interleukin-12/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Muromegalovirus/immunology , Receptors, Adrenergic, beta-2/immunology , STAT4 Transcription Factor/immunology , Up-Regulation/immunology
2.
Neurochem Int ; 130: 104320, 2019 11.
Article in English | MEDLINE | ID: mdl-30342959

ABSTRACT

Since the beginning of the last century, substantial evidence has suggested that various aspects of the immune system are influenced by the activity of the nervous system. However, the cellular and molecular basis for the neural control of immune responses has emerged only in the past decade. Recent studies have shown that adrenergic nerves control trafficking of immune cells through cell-type-specific mechanisms. Activation of the ß2-adrenergic receptor expressed on lymphocytes enhances signals mediated by a particular set of chemokine receptors, and consequently inhibits their exit from lymph nodes. This mechanism is involved in the diurnal variation of adaptive immune responses and the progression of inflammatory diseases. In the present review, we focus on the role of adrenergic nerves in the control of lymphocyte trafficking and adaptive immune responses in physiological and pathological conditions.


Subject(s)
Adaptive Immunity/physiology , Adrenergic Neurons/metabolism , Lymphocytes/metabolism , Receptors, Adrenergic, beta-2/metabolism , Adrenergic Neurons/immunology , Animals , Circadian Rhythm/physiology , Humans , Lymphocytes/immunology , Protein Transport/physiology , Receptors, Adrenergic, beta-2/immunology
3.
Nat Commun ; 9(1): 2723, 2018 07 13.
Article in English | MEDLINE | ID: mdl-30006573

ABSTRACT

Regulating immunity is a leading target for cancer therapy. Here, we show that the anti-tumor immune response can be modulated by the brain's reward system, a key circuitry in emotional processes. Activation of the reward system in tumor-bearing mice (Lewis lung carcinoma (LLC) and B16 melanoma) using chemogenetics (DREADDs), resulted in reduced tumor weight. This effect was mediated via the sympathetic nervous system (SNS), manifested by an attenuated noradrenergic input to a major immunological site, the bone marrow. Myeloid derived suppressor cells (MDSCs), which develop in the bone marrow, became less immunosuppressive following reward system activation. By depleting or adoptively transferring the MDSCs, we demonstrated that these cells are both necessary and sufficient to mediate reward system effects on tumor growth. Given the central role of the reward system in positive emotions, these findings introduce a physiological mechanism whereby the patient's psychological state can impact anti-tumor immunity and cancer progression.


Subject(s)
Carcinoma, Lewis Lung/drug therapy , Clozapine/analogs & derivatives , Immunologic Factors/pharmacology , Melanoma, Experimental/drug therapy , Myeloid-Derived Suppressor Cells/drug effects , Reward , Ventral Tegmental Area/drug effects , Adrenergic Neurons/drug effects , Adrenergic Neurons/immunology , Adrenergic Neurons/pathology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Clozapine/pharmacology , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/immunology , Dopaminergic Neurons/pathology , Immunity, Innate/drug effects , Injections, Intraventricular , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/pathology , Norepinephrine/metabolism , Stereotaxic Techniques , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/immunology , Sympathetic Nervous System/pathology , Tumor Burden/drug effects , Ventral Tegmental Area/immunology , Ventral Tegmental Area/pathology
4.
Science ; 359(6379): 1056-1061, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29496881

ABSTRACT

The type 2 inflammatory response is induced by various environmental and infectious stimuli. Although recent studies identified group 2 innate lymphoid cells (ILC2s) as potent sources of type 2 cytokines, the molecular pathways controlling ILC2 responses are incompletely defined. Here we demonstrate that murine ILC2s express the ß2-adrenergic receptor (ß2AR) and colocalize with adrenergic neurons in the intestine. ß2AR deficiency resulted in exaggerated ILC2 responses and type 2 inflammation in intestinal and lung tissues. Conversely, ß2AR agonist treatment was associated with impaired ILC2 responses and reduced inflammation in vivo. Mechanistically, we demonstrate that the ß2AR pathway is a cell-intrinsic negative regulator of ILC2 responses through inhibition of cell proliferation and effector function. Collectively, these data provide the first evidence of a neuronal-derived regulatory circuit that limits ILC2-dependent type 2 inflammation.


Subject(s)
Adaptive Immunity , Adrenergic Neurons/immunology , Immunity, Innate , Lymphocytes/metabolism , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-2 Receptor Agonists/pharmacology , Animals , Humans , Inflammation/immunology , Intestines/immunology , Lung/immunology , Mice , Mice, Inbred C57BL , Nerve Net/immunology , Receptors, Adrenergic, beta-2/genetics , Signal Transduction
5.
Neuropharmacology ; 71: 299-307, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23602988

ABSTRACT

Antidepressants that inhibit the recapture of noradrenaline have variable effects in chronic pain which may be related to the complex role of noradrenaline in pain modulation. Whereas at the spinal cord noradrenaline blocks nociceptive transmission, both antinociception and pronociception were reported after noradrenaline release in the brain. To study the role of noradrenaline in pain modulatory areas of the brain, we elected the dorsal reticular nucleus (DRt), a key pain facilitatory area located at the medulla oblongata. Three studies were performed. First, we show that the infusion in the DRt of nomifensine, which increases local extracellular levels of noradrenaline as shown by in vivo microdialysis, also enhances pain behavioral responses during both phases of the formalin test, a classic inflammatory pain model. Then, we demonstrate that the formalin test triggers the release of noradrenaline in the DRt in a biphasic pattern that matches the two phases of the test. Finally, we show that reducing noradrenaline release into the DRt, using an HSV-1 vector which decreases the expression of tyrosine hydroxylase in noradrenergic DRt-projecting neurons, attenuates pain behavioral responses in both phases of the formalin test. The increased noradrenaline levels induced by the infusion of nomifensine at the DRt, along with the hyperalgesic effects of noradrenaline released at the DRt upon noxious stimulation, indicates that noradrenaline may enhance pain facilitation from the brain. It is important to evaluate if antidepressants that inhibit noradrenaline recapture enhance pain facilitation from the brain herein attenuating their analgesic effects.


Subject(s)
Adrenergic Neurons/metabolism , Disease Models, Animal , Encephalitis/physiopathology , Hyperalgesia/metabolism , Norepinephrine/metabolism , Reticular Formation/metabolism , Up-Regulation/drug effects , Adrenergic Neurons/drug effects , Adrenergic Neurons/immunology , Adrenergic Neurons/pathology , Animals , Behavior, Animal/drug effects , Gene Knockdown Techniques , Hyperalgesia/etiology , Hyperalgesia/immunology , Hyperalgesia/pathology , Locus Coeruleus/drug effects , Locus Coeruleus/immunology , Locus Coeruleus/metabolism , Locus Coeruleus/pathology , Male , Microdialysis , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nomifensine , Norepinephrine/agonists , Norepinephrine/antagonists & inhibitors , Pain Measurement/drug effects , Rats , Rats, Wistar , Reticular Formation/drug effects , Reticular Formation/immunology , Reticular Formation/pathology , Synaptic Transmission/drug effects , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
6.
J Immunol ; 190(9): 4717-24, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23543756

ABSTRACT

Effective host defense requires a robust, yet self-limited response to pathogens. A poorly calibrated response can lead to either bacterial dissemination due to insufficient inflammation or organ injury due to excessive inflammation. Recent evidence suggests that the cholinergic anti-inflammatory reflex helps calibrate the immune response. However, the influence of peripheral noradrenergic neurons, which are primarily sympathetic neurons, in regulating immunity remains incompletely characterized. Using a model of 6-hydroxydopamine-mediated noradrenergic nerve ablation, we show that elimination of noradrenergic neurons improves survival during Klebsiella pneumoniae peritonitis (67 versus 23%, p < 0.005) in mice. The survival benefit results from enhanced MCP-1-dependent monocyte recruitment and a subsequent decrease in bacterial loads. Splenectomy eliminated both the survival benefit of 6-hydroxydopamine and monocyte recruitment, suggesting that monocytes recruited to the peritoneum originate in the spleen. These results suggest that noradrenergic neurons regulate the immune response through two pathways. First, sympathetic nerve-derived norepinephrine directly restrains MCP-1 production by peritoneal macrophages during infection. Second, norepinephrine derived from the vagally innervated splenic nerve regulates splenic monocyte egress. Removal of these two modulators of the immune response enhances antibacterial immunity and improves survival. These results may have implications for how states of catecholamine excess influence the host response to bacterial infections.


Subject(s)
Adrenergic Neurons/immunology , Klebsiella Infections/immunology , Klebsiella pneumoniae/immunology , Monocytes/immunology , Peritonitis/immunology , Adrenergic Neurons/metabolism , Adrenergic Neurons/microbiology , Animals , Cell Movement/immunology , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Cytokines/immunology , Cytokines/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Klebsiella Infections/metabolism , Klebsiella Infections/microbiology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mast Cells/immunology , Mast Cells/metabolism , Mast Cells/microbiology , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Monocytes/microbiology , Peritonitis/metabolism , Peritonitis/microbiology , Spleen/immunology , Spleen/metabolism , Spleen/microbiology
7.
Immunity ; 37(2): 290-301, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22863835

ABSTRACT

The multistep sequence leading to leukocyte migration is thought to be locally regulated at the inflammatory site. Here, we show that broad systemic programs involving long-range signals from the sympathetic nervous system (SNS) delivered by adrenergic nerves regulate rhythmic recruitment of leukocytes in tissues. Constitutive leukocyte adhesion and migration in murine bone marrow (BM) and skeletal-muscle microvasculature fluctuated with circadian peak values at night. Migratory oscillations, altered by experimental jet lag, were implemented by perivascular SNS fibers acting on ß-adrenoreceptors expressed on nonhematopoietic cells and leading to tissue-specific, differential circadian oscillations in the expression of endothelial cell adhesion molecules and chemokines. We showed that these rhythms have physiological consequences through alteration of hematopoietic cell recruitment and overall survival in models of septic shock, sickle cell vaso-occlusion, and BM transplantation. These data provide unique insights in the leukocyte adhesion cascade and the potential for time-based therapeutics for transplantation and inflammatory diseases.


Subject(s)
Cell Movement/immunology , Circadian Rhythm/immunology , Leukocytes/immunology , Sympathetic Nervous System/immunology , Adrenergic Fibers/immunology , Adrenergic Fibers/metabolism , Adrenergic Neurons/immunology , Adrenergic Neurons/metabolism , Anemia, Sickle Cell/immunology , Animals , Bone Marrow/metabolism , Bone Marrow Transplantation/immunology , Cell Adhesion/immunology , Chemokines/metabolism , Disease Models, Animal , Flow Cytometry , Green Fluorescent Proteins , Humans , Immunohistochemistry , Intercellular Adhesion Molecule-1/genetics , Isoproterenol/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Adrenergic, beta/metabolism , Shock, Septic/immunology , Sympathetic Nervous System/cytology , Sympathetic Nervous System/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...