Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
2.
Sci Immunol ; 8(86): eabo7975, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37595021

ABSTRACT

Thymic epithelial cells (TECs) produce glucocorticoids, which antagonize negative selection of autoreactive thymocytes and promote a competent T cell antigen-specific repertoire. To characterize their source, we generated a knock-in reporter mouse in which endogenous Cyp11b1, the final enzyme in de novo production of active glucocorticoids, was fluorescently tagged with mScarlet. Here, we find that Cyp11b1 is expressed in medullary TECs (mTECs) but not cortical TECs or other cells in the thymus. A distinct characteristic of mTECs is the presence of Aire, a transcription factor that drives expression of tissue-restricted antigens (TRAs) important for establishing immune tolerance. Cyp11b1 expression was highest in Aire+ mTECs, lower in post-Aire mTECs, and absent in mTECs of Aire-deficient mice. Transcriptomic analyses found that multiple enzymatic biosynthetic pathways are expressed specifically in mTECs and are also Aire dependent. In particular, we found that the thymus expresses messenger RNA for enzymes that catalyze production of many bioactive steroids and that glucocorticoids and sex steroids were secreted by cultured thymi. Expression of the transcripts for these genes and production of their final steroid products were markedly reduced in the absence of Aire. Thus, in addition to its well-established role in inducing TRAs that promote negative selection, Aire has an additional and contrary function of inducing glucocorticoids that antagonize negative selection, which together may expand and enhance the TCR repertoire. Furthermore, because Aire drives expression of multiple enzymes responsible for production of other non-gene-encoded bioactive molecules, it might have yet other roles in thymus development and function.


Subject(s)
Glucocorticoids , Steroid 11-beta-Hydroxylase , Transcription Factors , Animals , Mice , Epithelial Cells , Gene Expression Profiling , Transcription Factors/metabolism , Thymus Gland/metabolism , AIRE Protein
3.
J Clin Invest ; 133(18)2023 09 15.
Article in English | MEDLINE | ID: mdl-37471141

ABSTRACT

Glucocorticoids are steroid hormones with potent immunosuppressive properties. Their primary source is the adrenals, where they are generated via de novo synthesis from cholesterol. In addition, many tissues have a recycling pathway in which glucocorticoids are regenerated from inactive metabolites by the enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1, encoded by Hsd11b1). Here, we find that multiple tumor types express Hsd11b1 and produce active glucocorticoids. Genetic ablation of Hsd11b1 in such cells had no effect on in vitro growth, but reduced in vivo tumor progression, which corresponded with increased frequencies of CD8+ tumor-infiltrating lymphocytes (TILs) expressing activation markers and producing effector cytokines. Tumor-derived glucocorticoids were found to promote signatures of Treg activation and suppress signatures of conventional T cell activation in tumor-infiltrating Tregs. Indeed, CD8+ T cell activation was restored and tumor growth reduced in mice with Treg-specific glucocorticoid receptor deficiency. Importantly, pharmacologic inhibition of 11ß-HSD1 reduced tumor growth to the same degree as gene knockout and rendered immunotherapy-resistant tumors susceptible to PD-1 blockade. Given that HSD11B1 expression is upregulated in many human tumors and that inhibition of 11ß-HSD1 is well tolerated in clinical studies, these data suggest that targeting 11ß-HSD1 may be a beneficial adjunct in cancer therapy.


Subject(s)
Glucocorticoids , Neoplasms , Mice , Humans , Animals , Glucocorticoids/pharmacology , Glucocorticoids/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Receptors, Glucocorticoid/genetics , Gene Knockout Techniques
4.
Front Immunol ; 13: 975858, 2022.
Article in English | MEDLINE | ID: mdl-36119041

ABSTRACT

Sex steroid hormones have major effects on the thymus. Age-related increases in androgens and estrogens and pregnancy-induced increases in progestins all cause dramatic thymic atrophy. Atrophy can also be induced by treatment with exogenous sex steroids and reversed by ablation of endogenous sex steroids. Although these observations are frequently touted as evidence of steroid lymphotoxicity, they are often driven by steroid signaling in thymic epithelial cells (TEC), which are highly steroid responsive. Here, we outline the effects of sex steroids on the thymus and T cell development. We focus on studies that have examined steroid signaling in vivo, aiming to emphasize the actions of endogenous steroids which, via TEC, have remarkable programming effects on the TCR repertoire. Due to the dramatic effects of steroids on TEC, especially thymic involution, the direct effects of sex steroid signaling in thymocytes are less well understood. We outline studies that could be important in addressing these possibilities, and highlight suggestive findings of sex steroid generation within the thymus itself.


Subject(s)
Androgens , Thymocytes , Atrophy , Epithelium , Estrogens , Gonadal Steroid Hormones , Humans , Progestins , Receptors, Antigen, T-Cell
5.
Behav Brain Res ; 433: 114000, 2022 09 05.
Article in English | MEDLINE | ID: mdl-35817135

ABSTRACT

Social isolation is an established risk factor for mental illness and impaired immune function. Evidence suggests that neuroinflammatory processes contribute to mental illness, possibly via cytokine-induced modulation of neural activity. We examined the effects of lipopolysaccharide (LPS) administration and social home cage environment on cognitive performance in the 5-Choice Serial Reaction Time Task (5CSRTT), and their effects on corticosterone and cytokines in serum and brain tissue. Male Long-Evans rats were reared in pairs or in isolation before training on the 5CSRTT. The effects of saline and LPS (150 µg/kg i.p.) administration on sickness behaviour and task performance were then assessed. LPS-induced sickness behaviour was augmented in socially-isolated rats, translating to increased omissions and slower response times in the 5CSRTT. Both social isolation and LPS administration reduced impulsive responding, while discriminative accuracy remained unaffected. With the exception of reduced impulsivity in isolated rats, these effects were not observed following a second administration of LPS, revealing behavioural tolerance to repeated LPS injections. In a separate cohort of animals, social isolation potentiated the ability of LPS to increase serum corticosterone and IL-6, which corresponded to increased IL-6 in the orbitofrontal and medial prefrontal cortices and the nucleus accumbens. Basal IL-4 levels in the nucleus accumbens were reduced in socially-isolated rats. These findings are consistent with the adaptive response of reduced motivational drive following immune challenge, and identify social isolation as an exacerbating factor. Enhanced IL-6 signalling may play a role in mediating the potentiated behavioural response to LPS administration in isolated animals.


Subject(s)
Corticosterone , Lipopolysaccharides , Animals , Cognition/physiology , Cytokines , Humans , Interleukin-6 , Lipopolysaccharides/pharmacology , Male , Rats , Rats, Long-Evans
6.
Nat Rev Immunol ; 21(4): 233-243, 2021 04.
Article in English | MEDLINE | ID: mdl-33149283

ABSTRACT

Glucocorticoids (GCs) are small lipid hormones produced by the adrenals that maintain organismal homeostasis. Circadian and stress-induced changes in systemic GC levels regulate metabolism, cardiovascular and neural function, reproduction and immune activity. Our understanding of GC effects on immunity comes largely from administration of exogenous GCs to treat immune or inflammatory disorders. However, it is increasingly clear that endogenous GCs both promote and suppress T cell immunity. Examples include selecting an appropriate repertoire of T cell receptor (TCR) self-affinities in the thymus, regulating T cell trafficking between anatomical compartments, suppressing type 1 T helper (TH1) cell responses while permitting TH2 cell and, especially, IL-17-producing T helper cell responses, and promoting memory T cell differentiation and maintenance. Furthermore, in addition to functioning at a distance, extra-adrenal (local) production allows GCs to act as paracrine signals, specifically targeting activated T cells in various contexts in the thymus, mucosa and tumours. These pleiotropic effects on different T cell populations during development and immune responses provide a nuanced understanding of how GCs shape immunity.


Subject(s)
Glucocorticoids/immunology , Lymphopoiesis/immunology , Receptors, Glucocorticoid/immunology , T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Lineage , Gene Rearrangement, T-Lymphocyte/genetics , Gene Rearrangement, T-Lymphocyte/immunology , Humans , Immune Tolerance/immunology , Inflammation/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Helper-Inducer/immunology , Thymus Gland
7.
Nucl Recept Signal ; 17: 1550762919899643, 2020.
Article in English | MEDLINE | ID: mdl-35582456

ABSTRACT

Hormone-activated nuclear receptors (NRs) control myriad cellular processes. The classical paradigm for hormone delivery is secretion from endocrine organs and blood-borne distribution to responding cells. However, many hormones can also be synthesized in the same tissues in which responding cells are found (paracrine signaling). In both endocrine and paracrine signaling, numerous factors affect hormone availability to target cell NRs, including hormone access to and sequestration by carrier proteins, transport across cell membranes, metabolism, and receptor availability. These factors can differ dramatically during development, between anatomical locations, and across cell types, and may cause highly variable responses to the same hormone signal. This has been difficult to study because current approaches are unable to quantify cell-intrinsic exposure to NR hormone ligands, precluding assessment of cell-specific hormone access and signaling. We have used the ligand-dependent interaction of the endogenous glucocorticoid (GC) receptor with chromatin as a biosensor that quantifies systemic access of GCs to cells within tissues at the single cell level, showing that tissues are buffered against circulating GCs. This approach also showed highly targeted paracrine GC signaling within the thymus, where GCs promote the positive selection of thymocytes with moderate affinity for self-antigens and the development of a safe and effective T-cell repertoire. We believe that this and complementary biosensor approaches will be useful to identify endocrine and paracrine target cells in situ and quantify their exposure to hormones regardless of the mode of delivery.

8.
J Immunol ; 203(8): 2163-2170, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31527196

ABSTRACT

Glucocorticoid (GC) signaling in thymocytes shapes the TCR repertoire by antagonizing thymocyte negative selection. The transcription factors Nur77 and Helios, which are upregulated in TCR-signaled thymocytes, have been implicated in negative selection. In this study, we found that GCs inhibited Helios and, to a lesser extent, Nur77 upregulation in TCR-stimulated mouse thymocytes. Inhibition was increased by GC preincubation, and reductions in mRNA were prevented by a protein synthesis inhibitor, suggesting that GCs suppress indirectly via an intermediary factor. Upregulation of Helios in TCR-stimulated thymocytes was unaffected by deletion of Nur77, indicating Nur77 and Helios are regulated independently. Whereas CD4+ thymocytes are positively selected in wild-type AND TCR-transgenic B6 mice, loss of GC receptor expression resulted in increased negative selection. Correspondingly, Helios and Nur77 levels were elevated in TCRhiCD4+CD8+ (TCR-signaled) thymocytes. Notably, deletion of Helios fully reversed this negative selection, whereas deletion of Nur77 had no effect on CD4+CD8+ cell numbers but reversed the loss of mature CD4+ thymocytes. Thus, Nur77 and Helios are GC targets that play nonredundant roles in setting the signaling threshold for thymocyte negative selection.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Glucocorticoids/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Thymocytes/drug effects , Transcription Factors/antagonists & inhibitors , Animals , DNA-Binding Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Thymocytes/metabolism , Transcription Factors/metabolism
9.
Cell Rep ; 26(13): 3629-3642.e4, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30917317

ABSTRACT

Glucocorticoids are lipid-soluble hormones that signal via the glucocorticoid receptor (GR), a ligand-dependent transcription factor. Circulating glucocorticoids derive from the adrenals, but it is now apparent that paracrine glucocorticoid signaling occurs in multiple tissues. Effective local glucocorticoid concentrations and whether glucocorticoid delivery can be targeted to specific cell subsets are unknown. We use fluorescence detection of chromatin-associated GRs as biosensors of ligand binding and observe signals corresponding to steroid concentrations over physiological ranges in vitro and in vivo. In the thymus, where thymic epithelial cell (TEC)-synthesized glucocorticoids antagonize negative selection, we find that CD4+CD8+TCRhi cells, a small subset responding to self-antigens and undergoing selection, are specific targets of TEC-derived glucocorticoids and are exposed to 3-fold higher levels than other cells. These results demonstrate and quantitate targeted delivery of paracrine glucocorticoids. This approach may be used to assess in situ nuclear receptor signaling in a variety of physiological and pathological contexts.


Subject(s)
Glucocorticoids/metabolism , Thymus Gland/metabolism , Animals , Biosensing Techniques , Cell Line , Chromatin/metabolism , Drug Delivery Systems , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Single-Cell Analysis , Thymus Gland/cytology
10.
J Immunol ; 200(6): 1988-1994, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29440508

ABSTRACT

Glucocorticoid (GC) signaling in thymocytes counters negative selection and promotes the generation of a self-tolerant yet Ag-responsive T cell repertoire. Whereas circulating GC are derived from the adrenals, GC are also synthesized de novo in the thymus. The significance of this local production is unknown. In this study we deleted 11ß-hydroxylase, the enzyme that catalyzes the last step of GC biosynthesis, in thymic epithelial cells (TEC) or thymocytes. Like GC receptor-deficient T cells, T cells from mice lacking TEC-derived but not thymocyte-derived GC proliferated poorly to alloantigen, had a reduced antiviral response, and exhibited enhanced negative selection. Strikingly, basal expression of GC-responsive genes in thymocytes from mice lacking TEC-derived GC was reduced to the same degree as in GC receptor-deficient thymocytes, indicating that at steady-state the majority of biologically active GC are paracrine in origin. These findings demonstrate the importance of extra-adrenal GC even in the presence of circulating adrenal-derived GC.


Subject(s)
Antigens/metabolism , Epithelial Cells/metabolism , Glucocorticoids/metabolism , Thymocytes/metabolism , Animals , Cells, Cultured , Lymphocyte Activation/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mixed Function Oxygenases/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Glucocorticoid/metabolism , T-Lymphocytes/metabolism
11.
Sci Rep ; 7(1): 698, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28386080

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory condition with variable clinical presentation and disease progression. Importantly, animal models of RA are widely used to examine disease pathophysiology/treatments. Here, we exploited known vendor colony-based differences in endocrine/immune responses to gain insight into inflammatory modulators in arthritis, utilizing the adjuvant-induced arthritis (AA) model. Our previous study found that Sprague-Dawley (SD) rats from Harlan develop more severe AA, have lower corticosteroid binding globulin, and have different patterns of cytokine activation in the hind paw, compared to SD rats from Charles River. Here, we extend these findings, demonstrating that Harlan rats show reduced hypothalamic cytokine responses to AA, compared to Charles River rats, and identify colony-based differences in cytokine profiles in hippocampus and spleen. To go beyond individual measures, probing for networks of variables underlying differential responses, we combined datasets from this and the previous study and performed constrained principal component analysis (CPCA). CPCA revealed that with AA, Charles River rats show activation of chemokine and central cytokine networks, whereas Harlan rats activate peripheral immune/hypothalamic-pituitary-adrenal networks. These data suggest differential underlying disease mechanism(s), highlighting the power of evaluating multiple disease biomarkers, with potential implications for understanding differential disease profiles in individuals with RA.


Subject(s)
Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/metabolism , Endocrine System/immunology , Endocrine System/metabolism , Immune System/immunology , Immune System/metabolism , Animals , Arthritis, Experimental , Arthritis, Rheumatoid/pathology , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Female , Hormones/metabolism , Host Specificity , Inflammation Mediators/metabolism , Male , Organ Specificity , Rats
12.
Horm Behav ; 88: 4-14, 2017 02.
Article in English | MEDLINE | ID: mdl-27818220

ABSTRACT

Circulating glucocorticoids (GCs) are powerful regulators of immunity. Stress-induced GC secretion by the adrenal glands initially enhances and later suppresses the immune response. GC targets include lymphocytes of the adaptive immune system, which are well known for their sensitivity to GCs. Less appreciated, however, is that GCs are locally produced in lymphoid organs, such as the thymus, where GCs play a critical role in selection of the T cell antigen receptor (TCR) repertoire. Here, we review the roles of systemic and locally-produced GCs in T lymphocyte development, which has been studied primarily in laboratory mice. By antagonizing TCR signaling in developing T cells, thymus-derived GCs promote selection of T cells with stronger TCR signaling. This results in increased T cell-mediated immune responses to a range of antigens. We then compare local and systemic GC patterns in mice to those in several bird species. Taken together, these studies suggest that a combination of adrenal and lymphoid GC production might function to adaptively regulate lymphocyte development and selection, and thus antigen-specific immune reactivity, to optimize survival under different environmental conditions. Future studies should examine how lymphoid GC patterns vary across other vertebrates, how GCs function in B lymphocyte development in the bone marrow, spleen, and the avian bursa of Fabricius, and whether GCs adaptively program immunity in free-living animals.


Subject(s)
Adrenal Glands/metabolism , Glucocorticoids/metabolism , Receptors, Glucocorticoid/metabolism , T-Lymphocytes/metabolism , Thymus Gland/metabolism , Animals , Birds/metabolism , Female , Mice
13.
J Leukoc Biol ; 101(4): 893-900, 2017 04.
Article in English | MEDLINE | ID: mdl-28034915

ABSTRACT

The incidence of inflammatory bowel diseases (IBDs) has steadily increased in recent decades-a phenomenon that cannot be explained by genetic mutations alone. Other factors, including the composition of the intestinal microbiome, are potentially important contributors to the increased occurrence of this group of diseases. Previous reports have shown a correlation between early-life antibiotic (Abx) treatment and an increased incidence of IBD. In this report, we investigated the effects of early-life Abx treatments on the pathogenicity of CD4+ T cells using an experimental T cell transfer model of IBD. Our results show that CD4+ T cells isolated from adult mice that had been treated with Abx during gestation and in early life induced a faster onset of IBD in Rag1-deficient mice compared with CD4+ T cells of untreated mice. Ex vivo functional analyses of IBD-inducing CD4+ T cells did not show significant differences in their immunologic potential ex vivo, despite their in vivo phenotype. However, genome-wide gene-expression analysis revealed that these cells displayed dysregulated expression of genes associated with cell-cycle regulation, metabolism, and cellular stress. Analysis of Abx-treated CD4+ T cell donors showed systemically elevated levels of the stress hormone corticosterone throughout life compared with untreated donors. The cohousing of Abx-treated mice with untreated mice decreased serum corticosterone, and a consequent transfer of the cells from cohoused mice into Rag1-deficient mice restored the onset and severity of disease to that of untreated animals. Thus, our results suggest that early-life Abx treatment results in a stress response with high levels of corticosterone that influences CD4+ T cell function.


Subject(s)
Anti-Bacterial Agents/pharmacology , CD4-Positive T-Lymphocytes/immunology , Inflammation/immunology , Inflammation/pathology , Intestinal Mucosa/pathology , Animals , Anti-Bacterial Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , Corticosterone/blood , Cytokines/genetics , Cytokines/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genome , Housing, Animal , Inflammation/blood , Inflammatory Bowel Diseases/blood , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/drug effects , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/metabolism
14.
Brain Behav Immun ; 57: 271-281, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27165988

ABSTRACT

Glucocorticoids (GCs) are circulating adrenal steroid hormones that coordinate physiology, especially the counter-regulatory response to stressors. While systemic GCs are often considered immunosuppressive, GCs in the thymus play a critical role in antigen-specific immunity by ensuring the selection of competent T cells. Elevated thymus-specific GC levels are thought to occur by local synthesis, but the mechanism of such tissue-specific GC production remains unknown. Here, we found metyrapone-blockable GC production in neonatal and adult bone marrow, spleen, and thymus of C57BL/6 mice. This production was primarily via regeneration of adrenal metabolites, rather than de novo synthesis from cholesterol, as we found high levels of gene expression and activity of the GC-regenerating enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1), but not the GC-synthetic enzyme CYP11B1. Furthermore, incubation with physiological concentrations of GC metabolites (11-dehydrocorticosterone, prednisone) induced 11ß-HSD1- and GC receptor-dependent apoptosis (caspase activation) in both T and B cells, showing the functional relevance of local GC regeneration in lymphocyte GC signaling. Local GC production in bone marrow and spleen raises the possibility that GCs play a key role in B cell selection similar to their role in T cell selection. Our results also indicate that local GC production may amplify changes in adrenal GC signaling, rather than buffering against such changes, in the immune system.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Apoptosis , Bone Marrow/metabolism , Glucocorticoids/metabolism , Receptors, Glucocorticoid/metabolism , Spleen/metabolism , Steroid 11-beta-Hydroxylase/metabolism , Thymus Gland/metabolism , Animals , Animals, Newborn , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL
15.
J Biol Chem ; 291(21): 11300-12, 2016 May 20.
Article in English | MEDLINE | ID: mdl-27026706

ABSTRACT

Corticosteroid-binding globulin (CBG) was isolated from chicken serum and identified by mass spectrometry and genomic analysis. This revealed that the organization and synteny of avian and mammalian SerpinA6 genes are conserved. Recombinant zebra finch CBG steroid-binding properties reflect those of the natural protein in plasma and confirm its identity. Zebra finch and rat CBG crystal structures in complex with cortisol resemble each other, but their primary structures share only ∼40% identity, and their steroid-binding site topographies differ in several unexpected ways. Remarkably, a tryptophan that anchors ligands in mammalian CBG steroid-binding sites is replaced by an asparagine. Phylogenetic comparisons show that reptilian CBG orthologs share this unexpected property. Glycosylation of this asparagine in zebra finch CBG does not influence its steroid-binding affinity, but we present evidence that it may participate in protein folding and steroid-binding site formation. Substitutions of amino acids within zebra finch CBG that are conserved only in birds reveal how they contribute to their distinct steroid-binding properties, including their high (nanomolar) affinities for glucocorticoids, progesterone, and androgens. As in mammals, a protease secreted by Pseudomonas aeruginosa cleaves CBG in zebra finch plasma within its reactive center loop and disrupts steroid binding, suggesting an evolutionarily conserved property of CBGs. Measurements of CBG mRNA in zebra finch tissues indicate that liver is the main site of plasma CBG production, and anti-zebra finch CBG antibodies cross-react with CBGs in other birds, extending opportunities to study how CBG regulates the actions of glucocorticoids and sex steroids in these species.


Subject(s)
Avian Proteins/blood , Avian Proteins/genetics , Birds/blood , Birds/genetics , Evolution, Molecular , Transcortin/genetics , Transcortin/metabolism , Adaptation, Physiological , Amino Acid Sequence , Animals , Avian Proteins/chemistry , Chickens/blood , Chickens/genetics , Crystallography, X-Ray , Finches/blood , Finches/genetics , Glycosylation , Models, Molecular , Phylogeny , Rats , Sequence Homology, Amino Acid , Sparrows/blood , Sparrows/genetics , Transcortin/chemistry
16.
Dev Comp Immunol ; 54(1): 116-25, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26366679

ABSTRACT

Glucocorticoids are important for production of functional lymphocytes and immunity. In altricial neonates, adrenal glands are unresponsive and local glucocorticoid synthesis in lymphoid organs may be necessary to support lymphocyte development. Precocial neonates, in contrast, have fully responsive adrenal glucocorticoid production, and lymphoid glucocorticoid synthesis may not be necessary. Here, we found that in altricial zebra finch hatchlings, lymphoid organs had dramatically elevated endogenous glucocorticoid (and precursor) levels compared to levels in circulating blood. Furthermore, while avian adrenals produce corticosterone, finch lymphoid organs had much higher levels of cortisol, an unexpected glucocorticoid in birds. In contrast, precocial Japanese quail and chicken offspring did not have locally elevated lymphoid glucocorticoid levels, nor did their lymphoid organs contain high proportions of cortisol. These results show that lymphoid glucocorticoids differ in identity, concentration, and possibly source, in hatchlings of three different bird species. Locally-regulated glucocorticoids might have species-specific roles in immune development.


Subject(s)
Chickens/immunology , Coturnix/immunology , Finches/immunology , Hydrocortisone/biosynthesis , Lymphoid Tissue/metabolism , Animals , Animals, Newborn , Chickens/growth & development , Chromatography, High Pressure Liquid , Coturnix/growth & development , Finches/growth & development , Hydrocortisone/analysis , Immunoassay , Lymphoid Tissue/immunology , Species Specificity
17.
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
18.
Endocrinology ; 156(2): 511-22, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25406014

ABSTRACT

Glucocorticoids (GCs) are produced by the adrenal glands and circulate in the blood to coordinate organismal physiology. In addition, different tissues may independently regulate their local GC levels via local GC synthesis. Here, we find that in the mouse, endogenous GCs show tissue-specific developmental patterns, rather than mirroring GCs in the blood. Using solid-phase extraction, HPLC, and specific immunoassays, we quantified endogenous steroids and found that in tissues of female and male mice, (1) local GC levels can be much higher than systemic GC levels, (2) local GCs follow age-related patterns different from those of systemic GCs, and (3) local GCs have identities different from those of systemic GCs. For example, whereas corticosterone is the predominant circulating adrenal GC in mice, high concentrations of cortisol were measured in neonatal thymus, bone marrow, and heart. The presence of cortisol was confirmed with liquid chromatography-tandem mass spectrometry. In addition, gene expression of steroidogenic enzymes was detected across multiple tissues, consistent with local GC production. Our results demonstrate that local GCs can differ from GCs in circulating blood. This finding suggests that steroids are widely used as local (paracrine or autocrine) signals, in addition to their classic role as systemic (endocrine) signals. Local GC regulation may even be the norm, rather than the exception, especially during development.


Subject(s)
Growth and Development , Steroids/biosynthesis , Animals , Bone Marrow/metabolism , Brain/metabolism , Female , Gene Expression Regulation, Developmental , Liver/metabolism , Male , Mass Spectrometry , Mice, Inbred C57BL , Myocardium/metabolism , Spleen/metabolism , Steroids/blood , Thymus Gland/metabolism
19.
Eur J Neurosci ; 40(12): 3746-56, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25308904

ABSTRACT

Heightened motor impulsivity and increased novelty-seeking commonly co-occur in psychiatric disorders, including drug addiction. However, the relationship between these two phenomena remains unclear. One-time tests of novelty sensitivity commonly used in preclinical experiments, such as the open-field or novel-object test, fail to capture the fact that novelty-seekers repeatedly experience novel, stimulating situations. The present study therefore investigated whether repeated exposure to a novel, stimulating environment (SE) altered impulsive action. Male Long-Evans rats were trained to perform the five-choice serial reaction time task (5CSRTT) which measures motor impulsivity in the form of premature responding as well as attention and motivation. Animals were then exposed to a novel SE (1 h/day for 16 days) immediately prior to the 5CSRTT. Significant increases in premature responding were observed in a subgroup of reactive animals termed high responders (HR-SE). These rats were not more impulsive at baseline, and levels of impulsivity normalised once exposure to the SE was discontinued. No other aspect of 5CSRTT performance was affected by the SE challenge. We also determined that HR-SE rats were hyperactive in a novel environment. Biochemical analyses revealed changes in gene and protein expression within the dorsal hippocampus of HR-SE rats, including decreases in mRNA encoding the dopamine D1 receptor and brain-derived neurotrophic factor. These results indicate a novel mechanism by which impulsivity and novelty-reactivity interact that may enhance addiction vulnerability synergistically. Furthermore, studying such context-induced impulsivity may provide insight into the process by which environmental load precipitates psychiatric symptoms in impulse control disorders.


Subject(s)
Environment , Impulsive Behavior/physiology , Psychomotor Performance/physiology , Animals , Attention/physiology , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Corticosterone/blood , Hippocampus/physiology , Male , Motivation/physiology , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats, Long-Evans , Receptors, Dopamine D1/metabolism
20.
J Neuroendocrinol ; 25(2): 131-144, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22889357

ABSTRACT

Decreases in testosterone and 17ß-oestradiol (E(2)) are associated with an increased risk for Alzheimer's disease (AD), which has been attributed to an increase in ß-amyloid and tau pathological lesions. Although recent studies have used transgenic animal models to test the effects of sex steroid manipulations on AD-like pathology, almost none have systematically characterised the associations between AD lesions and sex steroid levels in the blood or brain in any mutant model. The present study evaluated age-related changes in testosterone and E(2) concentrations, as well as androgen receptor (AR) and oestrogen receptor (ER) α and ß expression, in brain regions displaying AD pathology in intact male and female 3xTgAD and nontransgenic (ntg) mice. We report for the first time that circulating and brain testosterone levels significantly increase in male 3xTgAD mice with age, but without changes in AR-immunoreactive (IR) cell number in the hippocampal CA1 or medial amygdala. The age-related increase in hippocampal testosterone levels correlated positively with increases in the conformational tau isoform, Alz50. These data suggest that the over-expression of human tau up-regulate the hypothalamic-pituitary-gonadal axis in these mice. Although circulating and brain E(2) levels remained stable with age in both male and female 3xTgAD and ntg mice, ER-IR cell number in the hippocampus and medial amygdala decreased with age in female transgenic mice. Furthermore, E(2) levels were significantly higher in the hippocampus than in serum, suggesting local production of E(2). Although triple transgenic mice mimic AD-like pathology, they do not fully replicate changes in human sex steroid levels, and may not be the best model for studying the effects of sex steroids on AD lesions.


Subject(s)
Alzheimer Disease/blood , Estradiol/blood , Testosterone/blood , Age Factors , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , tau Proteins/genetics , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...