Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
Cell Rep ; 37(4): 109883, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34706232

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) impede antitumor immunity; however, the precise mechanisms that regulate their suppressive function remain unresolved. Identifying these mechanisms could lead to therapeutic interventions to boost cancer immunotherapy efficacy. Here, we reveal that ß2 adrenergic receptor (ß2-AR) expression on MDSCs increases with tumor growth and that the ß2-AR stress pathway drives the immune suppressive activity of MDSCs by altering their metabolism. We show that ß2-AR signaling decreases glycolysis and increases oxidative phosphorylation and fatty acid oxidation (FAO). It also increases expression of the fatty acid transporter CPT1A, which is necessary for the FAO-mediated immunosuppressive function of MDSCs. Moreover, we show that ß2-AR signaling increases autophagy and activates the arachidonic acid cycle, both required for increasing the release of the immunosuppressive mediator, PGE2. Our data reveal that ß2-AR signaling triggered by stress is an important physiological regulator of key metabolic pathways in MDSCs, driving their immunosuppressive function.


Subject(s)
Myeloid-Derived Suppressor Cells/metabolism , Neoplasm Proteins/immunology , Neoplasms/immunology , Receptors, Adrenergic, beta-2/immunology , Signal Transduction/immunology , Tumor Microenvironment/immunology , Animals , Lipid Metabolism/genetics , Lipid Metabolism/immunology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasms/genetics , Oxidative Phosphorylation , Receptors, Adrenergic, beta-2/genetics , Tumor Microenvironment/genetics
2.
PLoS One ; 16(5): e0249202, 2021.
Article in English | MEDLINE | ID: mdl-33961631

ABSTRACT

PURPOSE: Agonistic ß2-adrenergic receptor autoantibodies (ß2-agAAb) have been observed in sera of patients with ocular hypertension and open-angle glaucoma (OAG). They target the ß2-receptors on trabecular meshwork, ciliary body and pericytes (Junemann et al. 2018; Hohberger et al. 2019). In addition to their influence on the intraocular pressure, an association to retinal microcirculation is discussed. This study aimed to investigate foveal avascular zone (FAZ) characteristics by en face OCT angiography (OCT-A) in glaucoma suspects and its relationship to ß2-agAAb status in patients with OAG. MATERIAL AND METHODS: Thirty-four patients (28 OAG, 6 glaucoma suspects) underwent standardized, clinical examination including sensory testing as white-on-white perimetry (Octopus G1, mean defect, MD) and structural measures as retinal nerve fibre layer (RNFL) thickness, neuroretinal rim width (BMO-MRW), retinal ganglion cell layer (RGCL) thickness, and inner nuclear layer (INL) thickness with high-resolution OCT. FAZ characteristics were measured by OCT-A scans of superficial vascular plexus (SVP), intermediate capillary plexus (ICP), and deep capillary plexus (DCP). FAZ-R was calculated (area FAZ (SVP)/area FAZ (ICP)). Using cardiomyocyte bioassays we analysed serum samples for the presence of ß2-agAAb. RESULTS: (I) Total mean FAZ area [mm2]: 0.34±0.16 (SVP), 0.24±0.12 (ICP), and 0.49±0.24 (DCP); mean FAZ-R 1.58±0.94. No correlation was seen for FAZ-R with MD, RNFL, BMO-MRW, RGCL thickness and INL thickness (p>0.05). (II) ß2-agAAb have been observed in 91% patients and showed no correlation with MD, RNFL, BMO-MRW, RGCL thickness and INL thickness (p>0.05). (III) FAZ-R correlated significantly with the ß2-agAAb-induced increase of the beat rate of cardiomyocyte (p = 0.028). CONCLUSION: FAZ characteristics did not correlate with any glaucoma associated functional and morphometric follow-up parameter in the present cohort. However, level of ß2-agAAb showed a significantly correlation with FAZ-ratio. We conclude that ß2-agAAb might be a novel biomarker in glaucoma pathogenesis showing association to FAZ-ratio with OCT-A.


Subject(s)
Autoantibodies/immunology , Glaucoma, Open-Angle/physiopathology , Microcirculation/immunology , Receptors, Adrenergic, beta-2/immunology , Aged , Female , Glaucoma, Open-Angle/immunology , Glaucoma, Open-Angle/pathology , Humans , Intraocular Pressure , Male , Retinal Ganglion Cells/pathology
3.
Cancer Immunol Res ; 9(6): 651-664, 2021 06.
Article in English | MEDLINE | ID: mdl-33762351

ABSTRACT

Metabolic dysfunction and exhaustion in tumor-infiltrating T cells have been linked to ineffectual antitumor immunity and the failure of immune checkpoint inhibitor therapy. We report here that chronic stress plays a previously unrecognized role in regulating the state of T cells in the tumor microenvironment (TME). Using two mouse tumor models, we found that blocking chronic adrenergic stress signaling using the pan ß-blocker propranolol or by using mice lacking the ß2-adrenergic receptor (ß2-AR) results in reduced tumor growth rates with significantly fewer tumor-infiltrating T cells that express markers of exhaustion, with a concomitant increase in progenitor exhausted T cells. We also report that blocking ß-AR signaling in mice increases glycolysis and oxidative phosphorylation in tumor-infiltrating lymphocytes (TIL), which associated with increased expression of the costimulatory molecule CD28 and increased antitumor effector functions, including increased cytokine production. Using T cells from Nur77-GFP reporter mice to monitor T-cell activation, we observed that stress-induced ß-AR signaling suppresses T-cell receptor (TCR) signaling. Together, these data suggest that chronic stress-induced adrenergic receptor signaling serves as a "checkpoint" of immune responses and contributes to immunosuppression in the TME by promoting T-cell metabolic dysfunction and exhaustion. These results also support the possibility that chronic stress, which unfortunately is increased in many patients with cancer following their diagnoses, could be exerting a major negative influence on the outcome of therapies that depend upon the status of TILs and support the use of strategies to reduce stress or ß-AR signaling in combination with immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Experimental/immunology , Receptors, Adrenergic, beta-2/immunology , Tumor Microenvironment/immunology , Adrenergic beta-Antagonists/pharmacology , Animals , Cell Line, Tumor , Cold-Shock Response , Female , Immunotherapy/methods , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Phenotype , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/immunology
4.
Life Sci ; 265: 118864, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33301808

ABSTRACT

Beta-2 adrenergic receptors (ß2-ARs) have important roles in the pathogenesis and treatment of chronic obstructive pulmonary disease (COPD). In recent years, progress has been made in the study of ß2-ARs. Here, we introduce the basic concepts of ß2-ARs, related pathways, as well as application of blockers/agonists of ß2-ARs, and ß2-AR autoantibodies in COPD. Drugs targeting the ß2-AR are being developed rapidly, and we expect them to improve the symptoms and prognosis of COPD patients in the future.


Subject(s)
Pulmonary Disease, Chronic Obstructive/physiopathology , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-2 Receptor Agonists/pharmacology , Adrenergic beta-2 Receptor Antagonists/pharmacology , Autoantibodies/immunology , Drug Development , Humans , Prognosis , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/immunology , Receptors, Adrenergic, beta-2/drug effects , Receptors, Adrenergic, beta-2/immunology
5.
Autoimmun Rev ; 19(6): 102527, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32247028

ABSTRACT

Myalgic Encephalomyelitis or Chronic Fatigue Syndrome (CFS/ME) is a complex and severely disabling disease with a prevalence of 0.3% and no approved treatment and therefore a very high medical need. Following an infectious onset patients suffer from severe central and muscle fatigue, chronic pain, cognitive impairment, and immune and autonomic dysfunction. Although the etiology of CFS/ME is not solved yet, there is numerous evidence for an autoantibody mediated dysregulation of the immune and autonomic nervous system. We found elevated ß2 adrenergic receptor (ß2AdR) and M3 acetylcholine receptor antibodies in a subset of CFS/ME patients. As both ß2AdR and M3 acetylcholine receptor are important vasodilators, we would expect their functional disturbance to result in vasoconstriction and hypoxemia. An impaired circulation and oxygen supply could result in many symptoms of ME/CFS. There are consistent reports of vascular dysfunction in ME/CFS. Muscular and cerebral hypoperfusion has been shown in ME/CFS in various studies and correlated with fatigue. Metabolic changes in ME/CFS are also in line with a concept of hypoxia and ischemia. Here we try to develop a unifying working concept for the complex pathomechanism of ME/CFS based on the presence of dysfunctional autoantibodies against ß2AdR and M3 acetylcholine receptor and extrapolate it to the pathophysiology of ME/CFS without an autoimmune pathogenesis.


Subject(s)
Autoantibodies/immunology , Fatigue Syndrome, Chronic/immunology , Fatigue Syndrome, Chronic/pathology , Models, Biological , Receptors, Adrenergic, beta-2/immunology , Humans
6.
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
7.
J Exp Med ; 217(4)2020 04 06.
Article in English | MEDLINE | ID: mdl-32045472

ABSTRACT

In humans, psychological stress has been associated with a higher risk of infectious illness. However, the mechanisms by which the stress pathway interferes with host response to pathogens remain unclear. We demonstrate here a role for the ß2-adrenergic receptor (ß2-AR), which binds the stress mediators adrenaline and noradrenaline, in modulating host response to mouse cytomegalovirus (MCMV) infection. Mice treated with a ß2-AR agonist were more susceptible to MCMV infection. By contrast, ß2-AR deficiency resulted in a better clearance of the virus, less tissue damage, and greater resistance to MCMV. Mechanistically, we found a correlation between higher levels of IFN-γ production by liver natural killer (NK) cells and stronger resistance to MCMV. However, the control of NK cell IFN-γ production was not cell intrinsic, revealing a cell-extrinsic downregulation of the antiviral NK cell response by adrenergic neuroendocrine signals. This pathway reduces host immune defense, suggesting that the blockade of the ß2-AR signaling could be used to increase resistance to infectious diseases.


Subject(s)
Cytomegalovirus Infections/immunology , Down-Regulation/immunology , Immunity, Innate/immunology , Receptors, Adrenergic, beta-2/immunology , Signal Transduction/immunology , Animals , Epinephrine/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Muromegalovirus/immunology , Norepinephrine/immunology
8.
Cell ; 180(1): 64-78.e16, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31923400

ABSTRACT

Enteric-associated neurons (EANs) are closely associated with immune cells and continuously monitor and modulate homeostatic intestinal functions, including motility and nutrient sensing. Bidirectional interactions between neuronal and immune cells are altered during disease processes such as neurodegeneration or irritable bowel syndrome. We investigated the effects of infection-induced inflammation on intrinsic EANs (iEANs) and the role of intestinal muscularis macrophages (MMs) in this context. Using murine models of enteric infections, we observed long-term gastrointestinal symptoms, including reduced motility and loss of excitatory iEANs, which was mediated by a Nlrp6- and Casp11-dependent mechanism, depended on infection history, and could be reversed by manipulation of the microbiota. MMs responded to luminal infection by upregulating a neuroprotective program via ß2-adrenergic receptor (ß2-AR) signaling and mediated neuronal protection through an arginase 1-polyamine axis. Our results identify a mechanism of neuronal death post-infection and point to a role for tissue-resident MMs in limiting neuronal damage.


Subject(s)
Intestinal Mucosa/immunology , Macrophages/immunology , Receptors, Adrenergic, beta-2/metabolism , Adrenergic Agents , Animals , Arginase/metabolism , Caspases, Initiator/immunology , Caspases, Initiator/metabolism , Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Female , Gastrointestinal Diseases , Gastrointestinal Microbiome , Infections , Inflammation/immunology , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Intestines/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Microbiota , Neurons/physiology , Receptors, Adrenergic, beta-2/immunology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Signal Transduction
9.
J Neuroimmune Pharmacol ; 15(1): 74-81, 2020 03.
Article in English | MEDLINE | ID: mdl-30617750

ABSTRACT

Evidence supporting the use of ß2AR agonists in synucleinopathies is rapidly growing. Findings come from different scientific approaches. Molecular and immunological data suggest that adrenergic stimulation may decrease both α-synuclein (α-syn) deposition and pro-inflammatory/neurotoxic molecules release. Small open label clinical trials including a total number of 25 Parkinson's disease (PD) patients, in which the ß2AR agonist salbutamol was added to levodopa, suggest a promising symptomatic benefit. In line with these findings, epidemiological studies investigating the risk of PD development suggest that long term exposure to the agonist salbutamol might be protective, while the antagonist propranolol possibly detrimental. Nonetheless, in both lines of investigation the studies performed so far present important limitations. On the clinical side, large randomized controlled trials are lacking, whereas on the epidemiological side the presence of co-morbid conditions (i.e. smoking and essential tremor) potentially influencing PD risk should taken into consideration. In summary, it is our opinion that ß2AR stimulation in synucleinopathies has a rationale and therefore merits further investigation. Graphical Abstract.


Subject(s)
Adrenergic beta-2 Receptor Agonists/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/immunology , Synucleinopathies/drug therapy , Synucleinopathies/immunology , Adrenergic beta-2 Receptor Agonists/pharmacology , Animals , Catecholamines/immunology , Clinical Trials as Topic/methods , Humans , Receptors, Adrenergic, beta-2/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
10.
J Clin Invest ; 129(12): 5537-5552, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31566578

ABSTRACT

Catecholamines released by sympathetic nerves can activate adrenergic receptors present on nearly every cell type, including myeloid-derived suppressor cells (MDSCs). Using in vitro systems, murine tumor models in wild-type and genetically modified (ß2-AR-/-) mice, and adoptive transfer approaches, we found that the degree of ß2-AR signaling significantly influences MDSC frequency and survival in tumors and other tissues. It also modulates their expression of immunosuppressive molecules such as arginase-I and PD-L1 and alters their ability to suppress the proliferation of T cells. The regulatory functions of ß2-AR signaling in MDSCs were also found to be dependent upon STAT3 phosphorylation. Moreover, we observed that the ß2-AR-mediated increase in MDSC survival is dependent upon Fas-FasL interactions, and this is consistent with gene expression analyses, which reveal a greater expression of apoptosis-related genes in ß2-AR-/- MDSCs. Our data reveal the potential of ß2-AR signaling to increase the generation of MDSCs from both murine and human peripheral blood cells and that the immunosuppressive function of MDSCs can be mitigated by treatment with ß-AR antagonists, or enhanced by ß-AR agonists. This strongly supports the possibility that reducing stress-induced activation of ß2-ARs could help to overcome immune suppression and enhance the efficacy of immunotherapy and other cancer therapies.


Subject(s)
Immune Tolerance , Myeloid-Derived Suppressor Cells/immunology , Receptors, Adrenergic, beta-2/immunology , Animals , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/physiology , Neoplasms/blood supply , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology
11.
Trans Am Clin Climatol Assoc ; 130: 235-245, 2019.
Article in English | MEDLINE | ID: mdl-31516188

ABSTRACT

Significant adverse impact of various forms of psychological stress on susceptibility to infection, altered wound healing, increased prevalence and severity of hypersensitivity diseases, and even increased mortality in cancer patients has been well described. Yet these observations are limited by often unpredictable individual responses to various stressful situations. These associations are further clouded by natural variability among diverse forms of and responses to chronic life stressors and associated comorbid conditions. This is particularly true for inflammatory diseases where gene/external environmental interactions are well-described. What is much less understood is gene-internal environmental (i.e., psychological) interactions that commonly affect disease activity and possible susceptibility. We have used selected single nucleotide polymorphisms of stress hormone and regulatory cytokine receptors to categorize both baseline and stress-associated immune parameters for the a priori classification of individuals with the most stress susceptible immune systems to identify those most responsive to a stress reduction/management-based intervention.


Subject(s)
Hypersensitivity/genetics , Stress, Psychological/genetics , Disease Susceptibility , Genetic Predisposition to Disease , Humans , Hypersensitivity/immunology , Hypersensitivity/psychology , Polymorphism, Single Nucleotide , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/immunology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/immunology , Stress, Psychological/immunology , Stress, Psychological/psychology
12.
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
13.
J Neuroinflammation ; 16(1): 112, 2019 May 28.
Article in English | MEDLINE | ID: mdl-31138227

ABSTRACT

BACKGROUND: Ischemic stroke provokes a neuroinflammatory response and simultaneously promotes release of epinephrine and norepinephrine by the sympathetic nervous system. This increased sympathetic outflow can act on ß2-adrenergic receptors expressed by immune cells such as brain-resident microglia and monocyte-derived macrophages (MDMs), but the effect on post-stroke neuroinflammation is unknown. Thus, we investigated how changes in ß2-adrenergic signaling after stroke onset influence the microglia/MDM stroke response, and the specific importance of microglia/MDM ß2-adrenergic receptors to post-stroke neuroinflammation. METHODS: To investigate the effects of ß2-adrenergic receptor manipulation on post-stroke neuroinflammation, we administered the ß2-adrenergic receptor agonist clenbuterol to mice 3 h after the onset of photothrombotic stroke. We immunostained to quantify microglia/MDM numbers and proliferation and to assess morphology and activation 3 days later. We assessed stroke outcomes by measuring infarct volume and functional motor recovery and analyzed gene expression levels of neuroinflammatory molecules. Finally, we evaluated changes in cytokine expression and microglia/MDM response in brains of mice with selective knockout of the ß2-adrenergic receptor from microglia and monocyte-lineage cells. RESULTS: We report that clenbuterol treatment after stroke onset causes enlarged microglia/MDMs and impairs their proliferation, resulting in reduced numbers of these cells in the peri-infarct cortex by 1.7-fold at 3 days after stroke. These changes in microglia/MDMs were associated with increased infarct volume in clenbuterol-treated animals. In mice that had the ß2-adrenergic receptor specifically knocked out of microglia/MDMs, there was no change in morphology or numbers of these cells after stroke. However, knockdown of ß2-adrenergic receptors in microglia and MDMs resulted in increased expression of TNFα and IL-10 in peri-infarct tissue, while stimulation of ß2-adrenergic receptors with clenbuterol had the opposite effect, suppressing TNFα and IL-10 expression. CONCLUSIONS: We identified ß2-adrenergic receptor signaling as an important regulator of the neuroimmune response after ischemic stroke. Increased ß2-adrenergic signaling after stroke onset generally suppressed the microglia/MDM response, reducing upregulation of both pro- and anti-inflammatory cytokines, and increasing stroke size. In contrast, diminished ß2-adrenergic signaling in microglia/MDMs augmented both pro- and anti-inflammatory cytokine expression after stroke. The ß2-adrenergic receptor may therefore present a therapeutic target for improving the post-stroke neuroinflammatory and repair process.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Brain Ischemia/immunology , Inflammation Mediators/immunology , Receptors, Adrenergic, beta-2/immunology , Signal Transduction/physiology , Stroke/immunology , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects , Stroke/metabolism , Stroke/pathology
14.
Cancer Immunol Immunother ; 68(1): 11-22, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30229289

ABSTRACT

Primary and secondary lymphoid organs are heavily innervated by the autonomic nervous system. Norepinephrine, the primary neurotransmitter secreted by post-ganglionic sympathetic neurons, binds to and activates ß-adrenergic receptors expressed on the surface of immune cells and regulates the functions of these cells. While it is known that both activated and memory CD8+ T-cells primarily express the ß2-adrenergic receptor (ß2-AR) and that signaling through this receptor can inhibit CD8+ T-cell effector function, the mechanism(s) underlying this suppression is not understood. Under normal activation conditions, T-cells increase glucose uptake and undergo metabolic reprogramming. In this study, we show that treatment of murine CD8+ T-cells with the pan ß-AR agonist isoproterenol (ISO) was associated with a reduced expression of glucose transporter 1 following activation, as well as decreased glucose uptake and glycolysis compared to CD8+ T-cells activated in the absence of ISO. The effect of ISO was specifically dependent upon ß2-AR, since it was not seen in adrb2-/- CD8+ T-cells and was blocked by the ß-AR antagonist propranolol. In addition, we found that mitochondrial function in CD8+ T-cells was also impaired by ß2-AR signaling. This study demonstrates that one mechanism by which ß2-AR signaling can inhibit CD8+ T-cell activation is by suppressing the required metabolic reprogramming events which accompany activation of these immune cells and thus reveals a new mechanism by which adrenergic stress can suppress the effector activity of immune cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Receptors, Adrenergic, beta-2/immunology , Signal Transduction/immunology , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Female , Glucose/immunology , Glucose/metabolism , Immune Tolerance/drug effects , Immune Tolerance/immunology , Isoproterenol/pharmacology , Lymphocyte Activation/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Propranolol/pharmacology , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects
15.
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
16.
Brain Behav Immun ; 74: 143-153, 2018 11.
Article in English | MEDLINE | ID: mdl-30172948

ABSTRACT

Acute exercise preferentially mobilizes cytotoxic T-cells, NK-cells and non-classical monocytes to the bloodstream under the influence of hemodynamic forces and/or ß2-adrenergic receptor (ß2-AR) signaling. However, the relative contribution of these mechanisms to the redeployment of the most exercise-responsive cell types is largely unknown. We determined the lymphocyte and monocyte subtypes mobilized to blood during exercise via ß2-AR signaling whilst controlling for ß1-AR mediated reductions in hemodynamic forces. In a randomized, double blind, complete cross-over design, 14 healthy cyclists exercised for 30-minutes at +10% of blood lactate threshold after ingesting: (1) a placebo, (2) a ß1-preferential antagonist (10 mg bisoprolol), or (2) a non-preferential ß1 + ß2-antagonist (80 mg nadolol) across three trials separated by >7-days. Bisoprolol was administered to reduce hemodynamic forces (heart rate and blood pressure) during exercise to levels comparable with nadolol but without blocking ß2-ARs. The mobilization of total NK-cells, terminally differentiated (CD57+) NK-cells, central memory, effector memory and CD45RA+ effector memory CD8+ T-cells; non-classical monocytes; and γδ T-cells were significantly blunted or abrogated under nadolol compared to both bisoprolol and placebo, indicating that the exercise-induced mobilization of these cell types to the blood is largely influenced by ß2-AR signaling. Nadolol failed to inhibit the mobilization of classical monocytes, CD4+ T-cells (and their subsets) or naïve CD8+ T-cells, indicating that these cell types are mobilized with exercise independently of the ß2-AR. We conclude that the preferential mobilization of NK-cells, non-classical monocytes and differentiated subsets of CD8+ T-cells with exercise is largely dependent on catecholamine signaling through the ß2-AR. These findings provide mechanistic insights by which distinct lymphocyte and monocyte subtypes are preferentially mobilized to protect the host from anticipated injury or infection in response to an acute stress response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Exercise/physiology , Killer Cells, Natural/immunology , Monocytes/immunology , Receptors, Adrenergic, beta-2/immunology , Adult , Bisoprolol/pharmacology , CD8-Positive T-Lymphocytes/metabolism , Catecholamines/immunology , Catecholamines/metabolism , Cross-Over Studies , Double-Blind Method , Female , Healthy Volunteers , Humans , Killer Cells, Natural/metabolism , Lymphocyte Activation , Male , Monocytes/metabolism , Nadolol/pharmacology , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction
17.
J Alzheimers Dis ; 64(4): 1091-1097, 2018.
Article in English | MEDLINE | ID: mdl-30010118

ABSTRACT

BACKGROUND: There is a need to assess promising biomarkers for diagnosis and treatment response in real-life settings. Despite the important role of vascular risk factors, cardiovascular biomarkers have played a minor role in dementia research. Agonistic autoantibodies (agAAB) directed against G-protein-coupled receptors (GPCR) are discussed as modulators of pathology and clinical manifestation. OBJECTIVE: 1) Describe prevalence of agAAB directed against GPCR, especially agABB against α1-adrenergic receptors (α1-AR-agAAB) and agABB directed against ß2-adrenergic receptors (ß2-AR-agAAB) and 2) identify factors associated with agAAB in people with dementia during routine care. METHODS: Blood samples and data from 95 subjects who screened positive for dementia from a primary care cohort, analyzed using an enzyme-linked immunosorbent assay (ELISA) for detecting agAAB. Sociodemographic and clinical data were assessed, and medical records checked. RESULTS: In 40 (42%) samples, agAAB was detected, with n = 29 (31%) representing α1-AR-agAAB and n = 21 (22%) ß2-AR-agAAB. There was no association between the presence of any antibody and a formal diagnosis of dementia. However, patients with coronary heart disease were more likely (OR = 4.23) to have α1-AR-agAAB than those without coronary heart disease. There were no associations between agAAB and age, sex, education, or cognitive impairment. DISCUSSION: For the first time, we show that autoantibodies have a significant prevalence in people with dementia in a routine care setting. Previous findings were restricted to highly selective samples. We replicated the association between α1-AR-agAAB in patients with coronary heart diseases but were not able to find other factors associated with the presence of agAAB.


Subject(s)
Autoantibodies/blood , Dementia/blood , Dementia/epidemiology , Receptors, Adrenergic, alpha-1/immunology , Aged , Aged, 80 and over , Cohort Studies , Community Health Planning , Female , Humans , Logistic Models , Male , Psychiatric Status Rating Scales , Psychometrics , Receptors, Adrenergic, beta-2/immunology , Severity of Illness Index
18.
Cardiovasc Res ; 114(11): 1487-1498, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29746700

ABSTRACT

Aims: ß1-adrenergic receptor autoantibodies (ß1-AAs) and ß2-adrenergic receptor autoantibodies (ß2-AAs) are present in patients with heart failure (HF); however, their interrelationship with cardiac structure and function remains unknown. This study explored the effects of the imbalance between ß1-AAs and ß2-AAs on cardiac structure and its underlying mechanisms in HF. Methods and results: Patients with left systolic HF who suffered from coronary heart disease (65.9%) or dilated cardiomyopathy (34.1%) were divided into New York Heart Association Classes I-II (n = 51) and Classes III-IV (n = 37) and compared with healthy volunteers as controls (n = 41). Total immunoglobulin G from HF patient serum comprising ß1-AAs and/or ß2-AAs were determined and purified for in vitro studies from neonatal rat cardiomyocytes (NRCMs). In addition, HF was induced by doxorubicin in mice. We observed that the increased ratio of ß1-AAs/ß2-AAs was associated with worsening HF in patients. Moreover, ß2-AAs from patients with HF suppressed the hyper-shrinking and apoptosis of NRCMS induced by ß1-AAs from some patients. Finally, ß2-AAs alleviated both myocardial damage and ß1-AAs production induced by doxorubicin in mice. Conclusion: ß2-AAs were capable of antagonizing the effects imposed by ß1-AAs both in vitro and in vivo. The imbalance of ß1-AAs and ß2-AAs in patients with HF is a mechanism underlying HF progression, and the increasing ratio of ß1-AAs/ß2-AAs should be considered a clinical assessment factor for the deterioration of cardiac function in patients with HF.


Subject(s)
Autoantibodies/immunology , Heart Failure/immunology , Heart Failure/prevention & control , Myocytes, Cardiac/immunology , Receptors, Adrenergic, beta-1/immunology , Receptors, Adrenergic, beta-2/immunology , Aged , Animals , Apoptosis , Case-Control Studies , Disease Models, Animal , Female , HEK293 Cells , Heart Failure/blood , Heart Failure/pathology , Humans , Male , Mice, Inbred BALB C , Middle Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Necrosis , Rats , Receptors, Adrenergic, beta-1/blood , Receptors, Adrenergic, beta-2/blood , Receptors, G-Protein-Coupled/immunology , Ventricular Function, Left
19.
J Leukoc Biol ; 104(3): 603-614, 2018 09.
Article in English | MEDLINE | ID: mdl-29668114

ABSTRACT

This study tests the hypothesis that in isolated human polymorphonuclear leukocytes (PMN) adrenergic ligands can affect neutrophil extracellular trap (NET) formation. We have previously shown that, in PMN, adrenaline (A), through the activation of adrenergic receptors (AR), reduces stimulus-dependent cell activation; we have, therefore, planned to investigate if AR are involved in NET production. PMN were obtained from venous blood of healthy subject. The ability of adrenergic ligands to affect reactive oxygen species (ROS) production, NET production, and cell migration was investigated in cells cultured under resting conditions or after activation with N-formyl-methionyl-leucyl-phenylalanine (fMLP), LPS, or IL-8. Stimuli-induced NET production measured as ROS, microscopic evaluation, and elastase production was reverted by A and this effect was blocked by the selective ß2 -AR antagonist ICI-118,551. The stimulus-induced ROS generation and migration was prevented by A and by isoprenaline (ISO), and these effects were counteracted only by ICI-118,551 and not by the other two selective ligands for the ß1 and ß3 -AR. Finally, the presence of the ß-ARs on PMN was confirmed, by means of microscopy and flow cytometry. The data of the present study suggest that adrenergic compounds, through the interaction of mainly ß2 -AR, are able to affect neutrophil functions. These data are suggestive of a possible therapeutic role of ß2 -AR ligands (in addition to their classical use), promoting the possible therapeutic relevance of adrenergic system in the modulation of innate immunity proposing their possible use as anti-inflammatory drugs.


Subject(s)
Extracellular Traps/metabolism , Neutrophils/metabolism , Receptors, Adrenergic, beta-2/metabolism , Cells, Cultured , Extracellular Traps/immunology , Humans , Neutrophils/immunology , Receptors, Adrenergic, beta-2/immunology
20.
PLoS One ; 13(3): e0192778, 2018.
Article in English | MEDLINE | ID: mdl-29538413

ABSTRACT

Dementia in general and Alzheimer's disease in particular is increasingly seen in association with autoimmunity being causatively or supportively involved in the pathogenesis. Besides classic autoantibodies (AABs) present in dementia patients, there is the new autoantibody class called functional autoantibodies, which is directed against G-protein coupled receptors (GPCRs; GPCR-AABs) and are seen as pathogenic players. However, less is known about dementia patients' burden with functional autoantibodies. We present here for the first time a study analyzing the prevalence of GPCR-AABs in patients with different dementia forms such as unclassified, Lewy body, vascular and Alzheimer's dementia. We identified the GPCR-AABs' specific targets on the receptors and introduced a neutralization strategy for GPCR-AABs. Patients with Alzheimer's and vascular dementia carried GPCR-AABs targeting the second loop of the alpha1- and the first loop of the beta2-adrenergic receptors (α1-AABs; ß2-AABs). The majority of patients with Lewy body dementia lacked any of the GPCR-AABs. In vitro, the function of the dementia-associated GPCR-AABs could be neutralized by the aptamer BC007. Due to the presence of GPCR-AABs in dementia patients mainly in those suffering from Alzheimer's and vascular dementia, the orchestra of immune players in these dementia forms, so far preferentially represented by the classic autoantibodies, should be supplemented by functional autoantibodies. As dementia-associated functional autoantibodies could be neutralized by the aptamer BC007, the first step was taken for a new in vivo treatment option in dementia patients who were positive for GPCR-AABs.


Subject(s)
Autoantibodies , Autoimmune Diseases , Dementia , Receptors, Adrenergic, beta-2/immunology , Aged , Autoantibodies/blood , Autoantibodies/immunology , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , Dementia/blood , Dementia/immunology , Female , Humans , Male , Middle Aged , Protein Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...