Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
Front Immunol ; 11: 583276, 2020.
Article in English | MEDLINE | ID: mdl-33363533

ABSTRACT

Testicular macrophages (TM) play a central role in maintaining testicular immune privilege and protecting spermatogenesis. Recent studies showed that their immunosuppressive properties are maintained by corticosterone in the testicular interstitial fluid, but the underlying molecular mechanisms are unknown. In this study, we treated mouse bone marrow-derived macrophages (BMDM) with corticosterone (50 ng/ml) and uncovered AMP-activated protein kinase (AMPK) activation as a critical event in M2 polarization at the phenotypic, metabolic, and cytokine production level. Primary TM exhibited remarkably similar metabolic and phenotypic features to corticosterone-treated BMDM, which were partially reversed by AMPK-inhibition. In a murine model of uropathogenic E. coli-elicited orchitis, intraperitoneal injection with corticosterone (0.1mg/day) increased the percentage of M2 TM in vivo, in a partially AMPK-dependent manner. This study integrates the influence of corticosterone on M2 macrophage metabolic pathways, phenotype, and function, and highlights a promising new avenue for the development of innovative therapeutics for orchitis patients.


Subject(s)
Corticosterone/immunology , Escherichia coli Infections/immunology , Immune Tolerance/immunology , Macrophages/immunology , Orchitis/immunology , AMP-Activated Protein Kinases/immunology , AMP-Activated Protein Kinases/metabolism , Animals , Corticosterone/pharmacology , Male , Mice , Mice, Inbred C57BL , Orchitis/metabolism , Phenotype , Testis , Uropathogenic Escherichia coli/immunology
2.
PLoS One ; 15(9): e0239231, 2020.
Article in English | MEDLINE | ID: mdl-32997686

ABSTRACT

It is controversially discussed whether immune-deficient mice experience severity in the absence of infection. Because a comprehensive analysis of the well-being of immune-deficient mice under specific pathogen free conditions is missing, we used a multi-parametric test analyzing, corticosterone, weight, nest building and facial expression over a period of 9 month to determine the well-being of two immune-deficient mouse lines (recombination activating gene 2- and interferon gamma receptor-deficient mice). We do not find evidence for severity when comparing immune-deficient mice to their heterozygous immune-competent littermates. Our data challenge the assumption that immune-deficiency per se regardless of housing conditions causes severity. Based on our study we propose to use objective non-invasive parameters determined by laboratory animal science for decisions concerning severity of immune-deficient mice.


Subject(s)
Corticosterone/genetics , DNA-Binding Proteins/genetics , Interferon-gamma/genetics , Mice, SCID/genetics , Animals , B-Lymphocytes/immunology , Corticosterone/immunology , Humans , Infections/genetics , Infections/immunology , Kidney/metabolism , Kidney/pathology , Mice , Mice, SCID/immunology , Pain/genetics , Pain/pathology , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Signal Transduction/genetics , T-Lymphocytes/immunology , Testosterone/genetics , Interferon gamma Receptor
3.
Theranostics ; 10(20): 9032-9049, 2020.
Article in English | MEDLINE | ID: mdl-32802177

ABSTRACT

Rationale: Herpes simplex virus type 1 (HSV-1) is a neurotropic virus that can cause a variety of clinical syndromes including mucocutaneous disease and HSV-1 encephalitis (HSE). Here, we characterize the molecular mechanisms underlying the susceptibility to HSV-1 under stressful conditions. Methods: Restraint stress and corticosterone (CORT, a primary stress hormone) were respectively used to establish HSV-1 susceptible model in vivo and in vitro. Viral titers were determined by plaque assay. Western blotting, immunofluorescence, transmission electron microscopy (TEM), qRT-PCR, H&E staining, IHC staining and flow cytometry were employed to evaluate virus-related protein expressions and detect the activation of autophagy. Loss- and gain-function assays, co-immunoprecipitation (co-IP) technique and autophagy agonist/antagonist treatments were applied in mechanistic experiments. Results: Restraint stress increased the susceptibility of mouse brain to HSV-1. Similarly, CORT treatment enhanced the susceptibility of neural cells to HSV-1. Furthermore, PML protein level in HSV-1 infected brain tissues and neural cells was remarkably decreased by stress treatment in vivo or CORT treatment in vitro, while its transcriptional level was not affected. Notably, a striking decline in protein expressions of ICP27 and gB was observed in PML-overexpressing cells, which was reversed by CORT treatment. By contrast, protein expression of gB was increased by knockdown with si-PML in virus-infected SH-SY5Y cells. We further discovered that CORT-driven PML degradation was dependent on the activation of autophagy in a ULK1-independent manner, rather than proteasome pathway. Bafilomycin A1 (BaF1) attenuated the augmentation effect of CORT on HSV-1 infection. The expressions of viral proteins were reduced in LC3-depleted cells, and the degradation of PML by CORT-induced autophagy was prevented in cells with LC3 knockdown by RNAi. Interestingly, PML was revealed to interact with the autophagic cargo receptor P62 and the autophagic effector protein LC3. Additionally, CORT failed to increase gB protein level when PML was silenced, providing direct evidence linking autophagic degradation of PML and CORT-induced virus susceptibility. Conclusion: Our results revealed that restraint stress/CORT increased HSV-1 susceptibility by delivering PML into autolysosomes for degradation. The results obtained from in vitro and in vivo models not only demonstrated the adverse effects of stress on HSV-1 infection, but also systematically investigated the underlying molecular mechanisms. These discoveries broaden our understanding of the interplay between host and viruses, and a comprehensive understanding of the role of autophagy in viral infection will provide information for future development of innovative drugs against viral infection.


Subject(s)
Autophagy/immunology , Corticosterone/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Promyelocytic Leukemia Protein/immunology , Animals , Brain/immunology , Cell Line , Chlorocebus aethiops , HeLa Cells , Humans , Male , Mice , Mice, Inbred BALB C , Neurons/immunology , Vero Cells , Viral Proteins/immunology , Virus Replication/immunology
4.
Gut Microbes ; 11(6): 1531-1546, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32573321

ABSTRACT

Although dysbiosis in the gut microbiota is known to be involved in several inflammatory diseases, whether any specific bacterial taxa control host response to inflammatory stimuli is still elusive. Here, we hypothesized that dysbiotic indigenous taxa could be involved in modulating host response to inflammatory triggers. To test this hypothesis, we conducted experiments in germ-free (GF) mice and in mice colonized with dysbiotic taxa identified in conventional (CV) mice subjected to chemotherapy-induced mucositis. First, we report that the absence of microbiota decreased inflammation and damage in the small intestine after administration of the chemotherapeutic agent 5-fluorouracil (5-FU). Also, 5-FU induced a shift in CV microbiota resulting in higher amounts of Enterobacteriaceae, including E. coli, in feces and small intestine and tissue damage. Prevention of Enterobacteriaceae outgrowth by treating mice with ciprofloxacin resulted in diminished 5-FU-induced tissue damage, indicating that this bacterial group is necessary for 5-FU-induced inflammatory response. In addition, monocolonization of germ-free (GF) mice with E. coli led to reversal of the protective phenotype during 5-FU chemotherapy. E. coli monocolonization decreased the basal plasma corticosterone levels and blockade of glucocorticoid receptor in GF mice restored inflammation upon 5-FU treatment. In contrast, treatment of CV mice with ciprofloxacin, that presented reduction of Enterobacteriaceae and E. coli content, induced an increase in corticosterone levels. Altogether, these findings demonstrate that Enterobacteriaceae outgrowth during dysbiosis impacts inflammation and tissue injury in the small intestine. Importantly, indigenous Enterobacteriaceae modulates host production of the anti-inflammatory steroid corticosterone and, consequently, controls inflammatory responsiveness in mice.


Subject(s)
Corticosterone/metabolism , Dysbiosis/microbiology , Enterobacteriaceae/growth & development , Animals , Antineoplastic Agents/adverse effects , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Bacteria/isolation & purification , Corticosterone/immunology , Dysbiosis/etiology , Dysbiosis/immunology , Dysbiosis/metabolism , Enterobacteriaceae/genetics , Fluorouracil/adverse effects , Gastrointestinal Microbiome/drug effects , Humans , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/microbiology , Male , Mice
5.
PLoS One ; 14(9): e0222856, 2019.
Article in English | MEDLINE | ID: mdl-31539413

ABSTRACT

Immune responses have been mostly studied at a specific time in anuran species. However, time-changes related to immunomodulation associated with glucocorticoid (GC) alterations following stressors and GC treatment are complex. The present study describes time-related changes in immune response and corticosterone (CORT) plasma levels following restraint challenge, short, mid and long-term captivity, and CORT exogenous administration by transdermal application (TA) in Rhinella ornata toads. We observed increased neutrophil: lymphocyte ratios after restraint challenge and CORT TA, without changes following short and mid-term captivity. Plasma bacterial killing ability was sustained in all treatments, except long-term captivity, with decreased values after 90 days under such conditions. Phagocytic activity of peritoneal cells increased after mid-term captivity, and the phytohemagglutinin swelling response was impaired in those animals treated with CORT TA for 20 consecutive days. Plasma CORT levels increased or were sustained after restraint challenge (depending on initial values), decreased following mid and long-term captivity (for those animals showing high CORT in the field) and increased after 20 days of CORT TA. By performing assessments of time-changes in immune processes and CORT plasma levels in R. ornata, we demonstrate immuno-enhancing effects following restraint, short and mid-term stressors, while long-term stressors and CORT TA promoted immunosuppression in these toads.


Subject(s)
Bufonidae/immunology , Corticosterone/immunology , Immunomodulation/immunology , Restraint, Physical , Stress, Physiological/immunology , Administration, Cutaneous , Animals , Bufonidae/physiology , Corticosterone/administration & dosage , Corticosterone/blood , Lymphocytes/immunology , Male , Neutrophils/immunology , Phagocytes/immunology , Time Factors
6.
J Exp Med ; 216(11): 2479-2491, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31467037

ABSTRACT

Hypothalamic-pituitary-adrenal (HPA) axis dysfunction contributes to numerous human diseases and disorders. We developed a high-affinity monoclonal antibody, CTRND05, targeting corticotropin-releasing factor (CRF). In mice, CTRND05 blocks stress-induced corticosterone increases, counteracts effects of chronic variable stress, and induces other phenotypes consistent with suppression of the HPA axis. CTRND05 induces skeletal muscle hypertrophy and increases lean body mass, effects not previously reported with small-molecule HPA-targeting pharmacologic agents. Multiorgan transcriptomics demonstrates broad HPA axis target engagement through altering levels of known HPA-responsive transcripts such as Fkbp5 and Myostatin and reveals novel HPA-responsive pathways such as the Apelin-Apelin receptor system. These studies demonstrate the therapeutic potential of CTRND05 as a suppressor of the HPA axis and serve as an exemplar of a potentially broader approach to target neuropeptides with immunotherapies, as both pharmacologic tools and novel therapeutics.


Subject(s)
Antibodies, Monoclonal/pharmacology , Corticotropin-Releasing Hormone/antagonists & inhibitors , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Stress, Physiological/drug effects , Animals , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Corticosterone/immunology , Corticosterone/metabolism , Corticotropin-Releasing Hormone/immunology , Gene Expression Profiling/methods , Humans , Hypothalamo-Hypophyseal System/immunology , Hypothalamo-Hypophyseal System/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype , Pituitary-Adrenal System/immunology , Pituitary-Adrenal System/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Stress, Physiological/immunology
7.
Neurotherapeutics ; 16(4): 1335-1349, 2019 10.
Article in English | MEDLINE | ID: mdl-31338703

ABSTRACT

Neuropeptide Y (NPY) has been demonstrated to exert stress buffering effects and promote resilience. Non-invasive intranasal (IN) application of NPY to rodents is able to mitigate traumatic stress-induced behavioral changes as well as dysfunction of the hypothalamic-pituitary-adrenal (HPA) axis. However, it is unknown whether IN NPY could prevent the behavioral, pro-inflammatory and neurochemical responses to peripheral immune activation by the Toll-like receptor 4 (TLR4) stimulant lipopolysaccharide (LPS). Therefore, we analyzed the effects of IN NPY (100 µg) on the behavioral sickness response (reduced locomotion and exploration) and the underlying molecular mechanisms, 3 h and 21 h after intraperitoneal injections of LPS (0.03 mg/kg) in male C57BL/6N mice. The acute behavioral sickness response was significantly dampened by pretreatment with IN NPY 3 h after LPS injection. This effect was accompanied by diminished weight loss and lowered plasma corticosterone (CORT) levels 21 h after LPS injection. In contrast, acute circulating cytokine levels and hypothalamic cytokine mRNA expression remained unaltered by IN NPY, which indicates that the peripheral and cerebral immune response to LPS was left undisturbed. Our findings are in agreement with the reported activity of NPY to dampen the response of the HPA axis to stress. We propose that IN NPY ablates sickness behavior at a site beyond the peripheral and cerebral cytokine response, an action that is associated with reduced activity of the HPA axis as determined by decreased plasma CORT.These results indicate that IN NPY administration may be relevant to the management of neuropsychiatric disorders arising from immune-induced neuroendocrine dysfunction.


Subject(s)
Hypothalamo-Hypophyseal System/drug effects , Illness Behavior/drug effects , Immunity, Cellular/drug effects , Lipopolysaccharides/toxicity , Neuropeptide Y/administration & dosage , Pituitary-Adrenal System/drug effects , Administration, Intranasal , Animals , Corticosterone/blood , Corticosterone/immunology , Hypothalamo-Hypophyseal System/immunology , Hypothalamo-Hypophyseal System/metabolism , Illness Behavior/physiology , Immunity, Cellular/physiology , Male , Mice , Mice, Inbred C57BL , Pituitary-Adrenal System/immunology , Pituitary-Adrenal System/metabolism
8.
Nat Commun ; 9(1): 4525, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30375380

ABSTRACT

Malaria reduces host fitness and survival by pathogen-mediated damage and inflammation. Disease tolerance mechanisms counter these negative effects without decreasing pathogen load. Here, we demonstrate that in four different mouse models of malaria, adrenal hormones confer disease tolerance and protect against early death, independently of parasitemia. Surprisingly, adrenalectomy differentially affects malaria-induced inflammation by increasing circulating cytokines and inflammation in the brain but not in the liver or lung. Furthermore, without affecting the transcription of hepatic gluconeogenic enzymes, adrenalectomy causes exhaustion of hepatic glycogen and insulin-independent lethal hypoglycemia upon infection. This hypoglycemia is not prevented by glucose administration or TNF-α neutralization. In contrast, treatment with a synthetic glucocorticoid (dexamethasone) prevents the hypoglycemia, lowers cerebral cytokine expression and increases survival rates. Overall, we conclude that in malaria, adrenal hormones do not protect against lung and liver inflammation. Instead, they prevent excessive systemic and brain inflammation and severe hypoglycemia, thereby contributing to tolerance.


Subject(s)
Adrenal Glands/metabolism , Brain/immunology , Cytokines/immunology , Hormones/immunology , Hypoglycemia/immunology , Liver/immunology , Lung/immunology , Malaria/immunology , Adrenal Glands/immunology , Adrenalectomy , Animals , Blood Glucose/drug effects , Brain/drug effects , Corticosterone/immunology , Corticosterone/metabolism , Cytokines/drug effects , Dexamethasone/pharmacology , Disease Models, Animal , Epinephrine/immunology , Epinephrine/metabolism , Glucocorticoids/immunology , Glucocorticoids/pharmacology , Glycogen/metabolism , Hydrocortisone/immunology , Hydrocortisone/metabolism , Inflammation , Liver/drug effects , Lung/drug effects , Mice , Mineralocorticoids/immunology , Mineralocorticoids/metabolism , Norepinephrine/immunology , Norepinephrine/metabolism , Plasmodium berghei , Plasmodium chabaudi , Survival Rate
9.
Sci Rep ; 8(1): 10188, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29976948

ABSTRACT

Increased glucocorticoid concentrations have been shown to favor resilience towards autoimmune phenomena. Here, we addressed whether experimentally induced elevations in circulating glucocorticoids mitigate the abnormalities exhibited by an experimental model of Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcus (PANDAS). This is a pathogenic hypothesis linking repeated exposures to Group-A-beta-hemolytic streptococcus (GAS), autoantibodies targeting selected brain nuclei and neurobehavioral abnormalities. To persistently elevate glucocorticoid concentrations, we supplemented lactating SJL/J mice with corticosterone (CORT; 80 mg/L) in the drinking water. Starting in adolescence (postnatal day 28), developing offspring were exposed to four injections - at bi-weekly intervals - of a GAS homogenate and tested for behavioral, immunological, neurochemical and molecular alterations. GAS mice showed increased perseverative behavior, impaired sensorimotor gating, reduced reactivity to a serotonergic agonist and inflammatory infiltrates in the anterior diencephalon. Neonatal CORT persistently increased circulating glucocorticoids concentrations and counteracted these alterations. Additionally, neonatal CORT increased peripheral and CNS concentrations of the anti-inflammatory cytokine IL-9. Further, upstream regulator analysis of differentially expressed genes in the striatum showed that the regulatory effect of estradiol is inhibited in GAS-treated mice and activated in GAS-treated mice exposed to CORT. These data support the hypothesis that elevations in glucocorticoids may promote central immunomodulatory processes.


Subject(s)
Autoimmune Diseases/immunology , Corpus Striatum/immunology , Corticosterone/immunology , Obsessive-Compulsive Disorder/immunology , Streptococcal Infections/immunology , Stress, Psychological/immunology , Animals , Animals, Newborn , Autoimmune Diseases/blood , Autoimmune Diseases/diagnosis , Autoimmune Diseases/microbiology , Behavior Observation Techniques , Behavior, Animal , Corpus Striatum/metabolism , Corticosterone/administration & dosage , Corticosterone/blood , Disease Models, Animal , Female , Gene Expression Profiling , Interleukin-9/immunology , Interleukin-9/metabolism , Lactation , Male , Mice , Mice, Inbred Strains , Obsessive-Compulsive Disorder/blood , Obsessive-Compulsive Disorder/diagnosis , Obsessive-Compulsive Disorder/microbiology , Streptococcal Infections/blood , Streptococcal Infections/diagnosis , Streptococcal Infections/microbiology , Streptococcus/pathogenicity , Stress, Psychological/blood
10.
J Neurosci ; 38(9): 2328-2340, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29382712

ABSTRACT

Repeated social defeat (RSD) stress promotes the release of bone marrow-derived monocytes into circulation that are recruited to the brain, where they augment neuroinflammation and cause prolonged anxiety-like behavior. Physiological stress activates the sympathetic nervous system and hypothalamic-pituitary-adrenal gland (HPA) axis, and both of these systems play a role in the physiological, immunological, and behavioral responses to stress. The purpose of this study was to delineate the role of HPA activation and corticosterone production in the immunological responses to stress in male C57BL/6 mice. Here, surgical (adrenalectomy) and pharmacological (metyrapone) interventions were used to abrogate corticosterone signaling during stress. We report that both adrenalectomy and metyrapone attenuated the stress-induced release of monocytes into circulation. Neither intervention altered the production of monocytes during stress, but both interventions enhanced retention of these cells in the bone marrow. Consistent with this observation, adrenalectomy and metyrapone also prevented the stress-induced reduction of a key retention factor, CXCL12, in the bone marrow. Corticosterone depletion with metyrapone also abrogated the stress-induced glucocorticoid resistance of myeloid cells. In the brain, these corticosterone-associated interventions attenuated stress-induced microglial remodeling, neurovascular expression of the adhesion molecule intercellular cell adhesion molecule-1, prevented monocyte accumulation and neuroinflammatory signaling. Overall, these results indicate that HPA activation and corticosterone production during repeated social defeat stress are critical for monocyte release into circulation, glucocorticoid resistance of myeloid cells, and enhanced neurovascular cell adhesion molecule expression.SIGNIFICANCE STATEMENT Recent studies of stress have identified the presence of monocytes that show an exaggerated inflammatory response to immune challenge and are resistant to the suppressive effects of glucocorticoids. Increased presence of these proinflammatory monocytes has been implicated in neuropsychiatric symptoms and the development of chronic cardiovascular, autoimmune, and metabolic disorders. In the current study, we show novel evidence that corticosterone produced during stress enhances the release of proinflammatory monocytes from the bone marrow into circulation, augments their recruitment to the brain and the induction of a neuroinflammatory profile. Overproduction of corticosterone during stress is also the direct cause of glucocorticoid resistance, a key phenotype in individuals exposed to chronic stress. Inhibiting excess corticosterone production attenuates these inflammatory responses to stress.


Subject(s)
Corticosterone/immunology , Intercellular Adhesion Molecule-1/biosynthesis , Monocytes/immunology , Neuroimmunomodulation/immunology , Stress, Psychological/immunology , Animals , Bone Marrow Cells/immunology , Cell Movement/immunology , Corticosterone/metabolism , Glucocorticoids/pharmacology , Hypothalamo-Hypophyseal System/physiology , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/immunology , Pituitary-Adrenal System/physiology , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
11.
J Comp Physiol B ; 188(2): 345-358, 2018 03.
Article in English | MEDLINE | ID: mdl-28988304

ABSTRACT

Knowledge of endocrine stress responses can be advantageous for understanding how animals respond to their environment. One tool in wildlife endocrinology is to measure the adrenocortical activity as a parameter of disturbance of animals. Fecal glucocorticoid metabolites (GCMs) provide a noninvasive assessment of adrenocortical activity. Using an adrenocorticotropic hormone (ACTH) challenge administered to 28 captive coyotes (Canis latrans), we measured the levels of plasma cortisol, and fecal cortisol and corticosterone metabolites (i.e., GCMs). Our goal was to determine the dose-response in the plasma and fecal samples following the injection and determine if there were effects of sex, age, and time of day. Specifically, animals were anesthetized for ~ 90 min with treatment animals intravenously injected with exogenous ACTH and control animals receiving saline. We collected blood samples prior to injection and at 4 different time points post-injection. We also collected fecal samples 2 days pre- and 2 days post-injection to measure fecal GCMs and determine if an endocrine stress response could be detected in fecal samples. We found a definite response in cortisol levels in the plasma for coyotes to the ACTH challenge. There was a response in fecal corticosterone 1 day post-injection, but the control males showed a similar response indicating a handling effect. Fecal cortisol levels did not indicate a response to the ACTH challenge, and were significantly lower than corticosterone concentrations. We also found significant sex, but not age or diurnal, differences in fecal GCMs. Radioimmunoassays for fecal corticosterone levels appeared to be a reliable indicator of physiological stress in coyotes.


Subject(s)
Adrenocorticotropic Hormone/pharmacokinetics , Corticosterone/metabolism , Coyotes/physiology , Feces/chemistry , Hydrocortisone/metabolism , Radioimmunoassay/methods , Adrenocorticotropic Hormone/blood , Animals , Antibodies/immunology , Corticosterone/immunology , Female , Hydrocortisone/immunology , Male , Sex Characteristics
12.
Ross Fiziol Zh Im I M Sechenova ; 103(3): 217-29, 2017 Mar.
Article in Russian | MEDLINE | ID: mdl-30199203

ABSTRACT

The review represents a modern concept about cells-molecular basis of mechanisms of neuro-immune interactions, the data on the effects of destabilizing factors (electric pain stimulation, rotation, cold and psychoemotional stress) on the functioning of neurons and immune cells. It must be underlined, that under the stress conditions take place the alterations of ligand-receptors interactions on the membrane of lymphocyte. In particular the reaction of these cells to regulating signal - application of Interleikin-1 grow up after mild stress, but it falls down after an influence of severe stress factors. Special attention is paid to the role of the orexinergic system in mechanism of realization of CNS reactions to application of antigens. In the present work the possible methods of correction of imbalance in functional interactions between nervous and immune systems, caused by different destabilizing factors, are reviewed.


Subject(s)
Central Nervous System/immunology , Immune System/metabolism , Neuroimmunomodulation/genetics , Signal Transduction/immunology , Stress, Psychological/immunology , Animals , Cell Differentiation/drug effects , Central Nervous System/metabolism , Central Nervous System/physiopathology , Corticosterone/blood , Corticosterone/immunology , DNA/pharmacology , Gene Expression Regulation , Humans , Immune System/physiopathology , Immunologic Factors/pharmacology , Interleukin-1alpha/blood , Interleukin-1alpha/immunology , Interleukin-1beta/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Neuroimmunomodulation/drug effects , Neurons/immunology , Neurons/metabolism , Orexin Receptors/genetics , Orexin Receptors/immunology , Orexins/genetics , Orexins/immunology , Signal Transduction/genetics , Stress, Psychological/genetics , Stress, Psychological/physiopathology , Stress, Psychological/prevention & control , Thymocytes/cytology , Thymocytes/drug effects , Thymocytes/immunology
13.
J Neuroimmunol ; 290: 9-14, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26711562

ABSTRACT

We aimed to evaluate the effect of paradoxical sleep deprivation on the cellular migration during inflammation, the peritoneal macrophage phenotype and the infectious stimulus outcomes. A/J mice were inoculated with thioglycollate and exposed to paradoxical sleep deprivation. Sleep-deprived animals presented decreased cell migration compared to controls. Nitric oxide production was reduced in macrophages from sleep-deprived mice compared to controls. Cell surface analysis showed that sleep deprivation reduced F4/80(+)/CD80(low) peritoneal cell population induced by thioglycollate injection. Sleep-deprived mice were not more susceptible to infection than control mice. Our findings challenge the general perception that sleep loss always increases infection susceptibility.


Subject(s)
Cell Movement/immunology , Macrophages, Peritoneal/immunology , Sleep Deprivation/immunology , Animals , Corticosterone/blood , Corticosterone/immunology , Macrophages, Peritoneal/metabolism , Male , Mice , Sleep Deprivation/blood
14.
Endocrinology ; 156(12): 4604-17, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26402842

ABSTRACT

Sprague Dawley rats from different vendor colonies display divergent responses in a variety of experimental paradigms. An adjuvant-induced arthritis (AA) model of human rheumatoid arthritis was used to examine immune and endocrine responses to inflammatory challenge in Sprague Dawley rats from Charles River and Harlan colonies. Rats were injected with either complete Freund's adjuvant or physiological saline (control), weights, and paw volumes measured over 15 days, and blood and tissue were collected 16 days post-injection. Overall, Harlan rats developed more severe AA than Charles River rats. In addition, despite comparable corticosterone levels, corticosteroid binding globulin levels were lower in Harlan compared with Charles River rats in the absence of inflammation, suggesting that a lower corticosterone reservoir in Harlan rats may underlie their greater susceptibility to inflammation. With increasing AA severity, there was an increase in plasma corticosterone (total and free) and a decrease in corticosteroid binding globulin in both Charles River and Harlan rats. However, contrasting patterns of cytokine activation were observed in the hind paw, suggesting a reliance on different cytokine networks at different stages of inflammation, with Charles River rats exhibiting increased TNF-α, monocyte chemotactic protein-1 (MCP-1), keratinocyte chemoattractant/growth-regulated oncogene (KC/GRO), and IL-1ß in the absence of clinical signs of arthritis, whereas Harlan had increased TNF-α, monocyte chemotactic protein-1, and IL-6 with mild to moderate arthritis. These colony-specific differences in endocrine and immune responses to AA in Sprague Dawley rats must be considered when comparing data from different laboratories and could be exploited to provide insight into physiological changes and therapeutic outcomes in arthritis and other inflammatory disorders.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Rats, Sprague-Dawley/immunology , Adjuvants, Immunologic/toxicity , Animals , Arthritis, Experimental/chemically induced , Arthritis, Rheumatoid/chemically induced , Chemokine CCL2/immunology , Chemokine CXCL1/immunology , Corticosterone/immunology , Disease Models, Animal , Female , Freund's Adjuvant/toxicity , Inflammation/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Rats , Severity of Illness Index , Transcortin/immunology , Tumor Necrosis Factor-alpha/immunology
15.
Mucosal Immunol ; 8(3): 559-71, 2015 May.
Article in English | MEDLINE | ID: mdl-25336169

ABSTRACT

Scavenger receptor B-I (SR-BI) is a multirecognition receptor that regulates cholesterol trafficking and cardiovascular inflammation. Although it is expressed by neutrophils (PMNs) and lung-resident cells, no role for SR-BI has been defined in pulmonary immunity. Herein, we report that, compared with SR-BI(+/+) counterparts, SR-BI(-/-) mice suffer markedly increased mortality during bacterial pneumonia associated with higher bacterial burden in the lung and blood, deficient induction of the stress glucocorticoid corticosterone, higher serum cytokines, and increased organ injury. SR-BI(-/-) mice had significantly increased PMN recruitment and cytokine production in the infected airspace. This was associated with defective hematopoietic cell-dependent clearance of lipopolysaccharide from the airspace and increased cytokine production by SR-BI(-/-) macrophages. Corticosterone replacement normalized alveolar neutrophilia but not alveolar cytokines, bacterial burden, or mortality, suggesting that adrenal insufficiency derepresses PMN trafficking to the SR-BI(-/-) airway in a cytokine-independent manner. Despite enhanced alveolar neutrophilia, SR-BI(-/-) mice displayed impaired phagocytic killing. Bone marrow chimeras revealed this defect to be independent of the dyslipidemia and adrenal insufficiency of SR-BI(-/-) mice. During infection, SR-BI(-/-) PMNs displayed deficient oxidant production and CD11b externalization, and increased surface L-selectin, suggesting defective activation. Taken together, SR-BI coordinates several steps in the integrated neutrophilic host defense response to pneumonia.


Subject(s)
Klebsiella Infections/immunology , Lung/immunology , Neutrophils/immunology , Pneumonia, Bacterial/immunology , Scavenger Receptors, Class B/immunology , Adrenal Glands/immunology , Adrenal Glands/pathology , Animals , Bacterial Load , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , CD11b Antigen/genetics , CD11b Antigen/immunology , Corticosterone/biosynthesis , Corticosterone/immunology , Cytokines/biosynthesis , Cytokines/immunology , Gene Expression Regulation , Klebsiella Infections/genetics , Klebsiella Infections/mortality , Klebsiella Infections/pathology , Klebsiella pneumoniae/immunology , L-Selectin/genetics , L-Selectin/immunology , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Lung/pathology , Male , Mice , Mice, Knockout , Neutrophils/pathology , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/mortality , Pneumonia, Bacterial/pathology , Scavenger Receptors, Class B/deficiency , Scavenger Receptors, Class B/genetics , Signal Transduction , Survival Analysis
16.
Eur J Immunol ; 45(3): 807-17, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25487064

ABSTRACT

Potentially life-threatening enterocolitis is the most frequent complication in children with colonic aganglionosis (Hirschsprung disease, HSCR), and little is known about the mechanisms leading to enterocolitis. Splenic lymphopenia has been reported in the Endothelin Receptor B (Ednrb)-null mouse model of HSCR that develops enterocolitis. In this study, we sought to identify molecular mechanisms underlying this immune phenotype. We employed the Ednrb(-/-) mouse, and the knockout of its ligand, Edn3 (Edn3(-/-)). The major finding is that enterocolitis in the Ednrb(-/-) and Edn3(-/-) mice lead to thymic involution, splenic lymphopenia, and suppression of B lymphopoiesis as a consequence of colonic aganglionosis, not an intrinsic Edn3-Ednrb signaling defect directly affecting the lymphoid organs. We showed that adoptive transfer of Ednrb(-/-) marrow repopulated the RAG2-null mice marrow, thymus and spleen without development of enterocolitis. We identified the glucocorticoid corticosterone, as a potential mediator of the immune phenotype. This previously unrecognized pattern of immune abnormalities in mouse is nearly identical to lymphoid depletion in neonatal sepsis during severe physiological stress, suggesting that the mouse model used here could be also used for sepsis studies.


Subject(s)
Endothelin-3/immunology , Enterocolitis/immunology , Hirschsprung Disease/immunology , Receptor, Endothelin B/immunology , Animals , Colon/immunology , Colon/pathology , Corticosterone/genetics , Corticosterone/immunology , Disease Models, Animal , Endothelin-3/genetics , Enterocolitis/genetics , Enterocolitis/pathology , Hirschsprung Disease/genetics , Hirschsprung Disease/pathology , Mice , Mice, Knockout , Receptor, Endothelin B/genetics , Spleen/immunology , Spleen/pathology , Thymus Gland/immunology , Thymus Gland/pathology
17.
J Immunol ; 193(6): 2743-52, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25114100

ABSTRACT

Autoimmune neuroinflammation, including multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), a prototype for T cell-mediated autoimmunity, is believed to result from immune tolerance dysfunction leading to demyelination and substantial neurodegeneration. We previously showed that CNS-restricted expression of hepatocyte growth factor (HGF), a potent neuroprotective factor, reduced CNS inflammation and clinical deficits associated with EAE. In this study, we demonstrate that systemic HGF treatment ameliorates EAE through the development of tolerogenic dendritic cells (DCs) with high expression levels of glucocorticoid-induced leucine zipper (GILZ), a transcriptional repressor of gene expression and a key endogenous regulator of the inflammatory response. RNA interference-directed neutralization of GILZ expression by DCs suppressed the induction of tolerance caused by HGF. Finally, adoptive transfer of HGF-treated DCs from wild-type but not GILZ gene-deficient mice potently mediated functional recovery in recipient mice with established EAE through effective modulation of autoaggressive T cell responses. Altogether, these results show that by inducing GILZ in DCs, HGF reproduces the mechanism of immune regulation induced by potent immunomodulatory factors such as IL-10, TGF-ß1, and glucocorticoids and therefore that HGF therapy may have potential in the treatment of autoimmune dysfunctions.


Subject(s)
Corticosterone/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Hepatocyte Growth Factor/pharmacology , T-Lymphocytes/immunology , Transcription Factors/biosynthesis , Adoptive Transfer , Animals , Autoimmunity/immunology , Cell Proliferation , Cells, Cultured , Central Nervous System/immunology , Corticosterone/blood , Dendritic Cells/immunology , Dendritic Cells/transplantation , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Hepatocyte Growth Factor/biosynthesis , Immune Tolerance/genetics , Inflammation/immunology , Interleukin-10/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/immunology , RNA Interference , RNA, Small Interfering , Transcription Factors/genetics , Transforming Growth Factor beta1/immunology
18.
Article in English | MEDLINE | ID: mdl-25086332

ABSTRACT

Yolk testosterone concentrations vary in response to environmental conditions and different testosterone contents can subsequently modify the phenotypic traits of offspring. Apart from effects on growth, proactive behaviour and secondary sexual characteristics, the possible negative impacts of maternal testosterone on the immune system are often considered a limitation for its deposition. The effects of maternal testosterone can be modulated by postnatal environmental conditions, such as the availability of food resources. However, the majority of studies considering the effects of maternal testosterone on the immune system have been conducted under optimum conditions. We evaluated the influence of genetic selection for high (HET) and low (LET) egg testosterone content in Japanese quail on immune responsiveness of offspring to phytohaemagglutinin (PHA) and lipopolysaccharide (LPS) stimulation under severe protein restriction. Protein restriction negatively influenced body weight and performance in the PHA-test. We observed an increase in Cort (corticosterone) and He/Ly (heterophil/lymphocyte ratio) after LPS, while no changes occurred in total IgY levels in the protein-restricted group. HET quails showed higher body mass and total IgY levels and lower He/Ly ratio than LET quails, while the PHA index and Cort concentration did not differ between lines. No interactions were found between protein restriction and genetic line. In conclusion, the immune response was not compromised under conditions of severe protein restriction in the faster growing HET line compared with the LET line. We hypothesise that the immune responsiveness of birds with higher yolk testosterone may be linked with other maternally-derived substances in a context-dependent manner.


Subject(s)
Egg Yolk/immunology , Egg Yolk/metabolism , Quail/immunology , Quail/metabolism , Testosterone/immunology , Testosterone/metabolism , Animals , Body Weight/immunology , Corticosterone/immunology , Corticosterone/metabolism , Diet, Protein-Restricted/methods , Environment , Female , Immunoglobulins/immunology , Lipopolysaccharides/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Phenotype , Phytohemagglutinins/immunology , Selection, Genetic/immunology
19.
Int Arch Allergy Immunol ; 163(4): 297-306, 2014.
Article in English | MEDLINE | ID: mdl-24776388

ABSTRACT

BACKGROUND: Psychological stress is associated with the aggravation of asthma symptoms. Glucocorticoids (GC), which are stress hormones released upon exposure to stress, have the potential to shift immune responses towards a predominant Th2 response by priming antigen-presenting cells to produce lower levels of IL-12 as well as reducing the development of regulatory T cells. However, the involvement of GC in psychological stress-induced exacerbations of allergic asthma has not yet been clarified. METHODS: Sensitized mice were exposed to restraint stress followed by forced swimming stress, during which a GC receptor antagonist or a GC synthesis inhibitor was administered, and then antigen was inhaled. Corticosterone levels in the blood were measured in stressed and nonstressed mice. After antigen inhalation, the airway responses to aerosolized methacholine, epithelial mucus secretion and airway inflammation were evaluated, and the IL-13 contents in bronchoalveolar lavage fluid were measured. RESULTS: The exposure to stress significantly increased corticosterone levels. Allergic airway responses and the increase of IL-13 contents evoked by antigen inhalation were significantly higher in stressed mice than in nonstressed mice. The administration of a GC receptor antagonist and a GC synthesis inhibitor during stress exposure significantly reduced the exacerbation of the airway responses and the increase of IL-13 contents in stressed mice challenged with antigen. CONCLUSIONS: These results indicate that the increased release of GC upon exposure to stress has a priming effect on the aggravation of allergic airway responses following the exposure, suggesting a pathophysiological role for the neuroendocrine axis in linking psychological stress to asthma exacerbations.


Subject(s)
Asthma/immunology , Asthma/psychology , Glucocorticoids/blood , Stress, Psychological/immunology , Animals , Asthma/physiopathology , Bronchial Hyperreactivity/physiopathology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Corticosterone/blood , Corticosterone/immunology , Corticosterone/pharmacology , Disease Models, Animal , Female , Glucocorticoids/immunology , Glucocorticoids/pharmacology , Inflammation/psychology , Interleukin-12/immunology , Interleukin-13/immunology , Male , Mice , Mice, Inbred BALB C , Mifepristone/pharmacology , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/biosynthesis , Respiratory System/physiopathology , Sex Factors , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology
20.
Sci Signal ; 7(314): ra20, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24570488

ABSTRACT

Intestinal crypt epithelial cells synthesize glucocorticoids, steroid hormones that protect against inflammatory bowel disease. To investigate how intestinal glucocorticoids are regulated during chronic inflammation, we induced chronic colitis in mice by exposing them to the chemical dextran sulfate sodium (DSS). We found that intestinal glucocorticoid secretion and expression of the genes Cyp11a1 and Cyp11b1 (which encode enzymes that synthesize glucocorticoids) were initially stimulated, but declined during the chronic phase, whereas tumor necrosis factor (TNF) and inflammatory cytokines secreted by T helper type 1 (TH1) and TH17 cells continuously increased in abundance in the inflamed colon. This suggested that inadequate intestinal glucocorticoid synthesis is a feature of chronic intestinal inflammation. We screened for cytokines that regulated intestinal glucocorticoid synthesis and found that TNF suppressed corticosterone secretion and Cyp11a1 and Cyp11b1 expression in an intestinal crypt epithelial cell line. TNF suppressed steroidogenesis by activating the transcription factors c-Jun and nuclear factor κB (NF-κB), which both interacted with the transcription factor NR5A2 and repressed Cyp11a1 reporter activity. This repression was relieved by expression of a dominant-negative form of c-Jun amino-terminal kinase 1 (JNK1), inhibitor of NF-κB, or by a JNK inhibitor. Furthermore, the dominant-negative TNF inhibitor XPro1595 inhibited c-Jun and NF-κB activation in mice, restored intestinal Cyp11a1 and Cyp11b1 expression, reduced colonic cell death, and rescued chronic colitis caused by DSS. Thus, during chronic colitis, TNF suppresses intestinal steroidogenic gene expression by inhibiting the activity of NR5A2, thus decreasing glucocorticoid synthesis and sustaining chronic inflammation.


Subject(s)
Colitis/metabolism , Glucocorticoids/biosynthesis , Intestinal Mucosa/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Cholesterol Side-Chain Cleavage Enzyme/biosynthesis , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/immunology , Chronic Disease , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Colitis/pathology , Corticosterone/biosynthesis , Corticosterone/genetics , Corticosterone/immunology , Dextran Sulfate/toxicity , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/immunology , Glucocorticoids/genetics , Glucocorticoids/immunology , Humans , Intestines/immunology , Intestines/pathology , Mice , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/immunology , Mitogen-Activated Protein Kinase 8/metabolism , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/immunology , Steroid 11-beta-Hydroxylase/biosynthesis , Steroid 11-beta-Hydroxylase/genetics , Steroid 11-beta-Hydroxylase/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...