Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.173
Filter
Add more filters

Publication year range
1.
Cell ; 156(3): 522-36, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24485458

ABSTRACT

The extended amygdala has dominated research on the neural circuitry of fear and anxiety, but the septohippocampal axis also plays an important role. The lateral septum (LS) is thought to suppress fear and anxiety through its outputs to the hypothalamus. However, this structure has not yet been dissected using modern tools. The type 2 CRF receptor (Crfr2) marks a subset of LS neurons whose functional connectivity we have investigated using optogenetics. Crfr2(+) cells include GABAergic projection neurons that connect with the anterior hypothalamus. Surprisingly, we find that these LS outputs enhance stress-induced behavioral measures of anxiety. Furthermore, transient activation of Crfr2(+) neurons promotes, while inhibition suppresses, persistent anxious behaviors. LS Crfr2(+) outputs also positively regulate circulating corticosteroid levels. These data identify a subset of LS projection neurons that promote, rather than suppress, stress-induced behavioral and endocrinological dimensions of persistent anxiety states and provide a cellular point of entry to LS circuitry.


Subject(s)
Anxiety/physiopathology , Hypothalamus/metabolism , Septum of Brain/physiology , Adrenal Cortex Hormones/metabolism , Amygdala/metabolism , Animals , Behavior, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/physiology , Receptors, Corticotropin-Releasing Hormone/metabolism , Stress, Physiological
2.
Front Neuroendocrinol ; 74: 101146, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39004314

ABSTRACT

Over the past two decades, there has been increasing evidence for the importance of rapid-onset actions of corticosteroid hormones in the brain. Here, we highlight the distinct rapid corticosteroid actions that regulate excitatory and inhibitory synaptic transmission in the hypothalamus, the hippocampus, basolateral amygdala, and prefrontal cortex. The receptors that mediate rapid corticosteroid actions are located at or close to the plasma membrane, though many of the receptor characteristics remain unresolved. Rapid-onset corticosteroid effects play a role in fast neuroendocrine feedback as well as in higher brain functions, including increased aggression and anxiety, and impaired memory retrieval. The rapid non-genomic corticosteroid actions precede and complement slow-onset, long-lasting transcriptional actions of the steroids. Both rapid and slow corticosteroid actions appear to be indispensable to adapt to a continuously changing environment, and their imbalance can increase an individual's susceptibility to psychopathology.


Subject(s)
Adrenal Cortex Hormones , Brain , Synaptic Transmission , Animals , Humans , Brain/metabolism , Brain/drug effects , Brain/physiology , Adrenal Cortex Hormones/metabolism , Adrenal Cortex Hormones/pharmacology , Adrenal Cortex Hormones/physiology , Synaptic Transmission/physiology , Synaptic Transmission/drug effects , Signal Transduction/physiology , Signal Transduction/drug effects
3.
J Virol ; 98(1): e0143623, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38084958

ABSTRACT

Bovine alphaherpesvirus 1 (BoHV-1) infections cause respiratory tract disorders and suppress immune responses, which can culminate in bacterial pneumonia. Following acute infection, BoHV-1 establishes lifelong latency in sensory neurons present in trigeminal ganglia (TG) and unknown cells in pharyngeal tonsil. Latently infected calves consistently reactivate from latency after an intravenous injection of the synthetic corticosteroid dexamethasone (DEX), which mimics the effects of stress. The immediate early transcription unit 1 (IEtu1) promoter drives expression of infected cell protein 0 (bICP0) and bICP4, two key viral transcriptional regulators. The IEtu1 promoter contains two functional glucocorticoid receptor (GR) response elements (GREs), and this promoter is transactivated by GR, DEX, and certain Krüppel transcription factors that interact with GC-rich motifs, including consensus specificity protein 1 (Sp1) binding sites. Based on these observations, we hypothesized that Sp1 stimulates productive infection and transactivates key BoHV-1 promoters. DEX treatment of latently infected calves increased the number of Sp1+ TG neurons and cells in pharyngeal tonsil indicating that Sp1 expression is induced by stress. Silencing Sp1 protein expression with siRNA or mithramycin A, a drug that preferentially binds GC-rich DNA, significantly reduced BoHV-1 replication. Moreover, BoHV-1 infection of permissive cells increased Sp1 steady-state protein levels. In transient transfection studies, GR and Sp1 cooperatively transactivate IEtu1 promoter activity unless both GREs are mutated. Co-immunoprecipitation studies revealed that GR and Sp1 interact in mouse neuroblastoma cells (Neuro-2A) suggesting this interaction stimulates IEtu1 promoter activity. Collectively, these studies suggested that the cellular transcription factor Sp1 enhances productive infection and stress-induced BoHV-1 reactivation from latency.IMPORTANCEFollowing acute infection, bovine alphaherpesvirus 1 (BoHV-1) establishes lifelong latency in sensory neurons in trigeminal ganglia (TG) and pharyngeal tonsil. The synthetic corticosteroid dexamethasone consistently induces BoHV-1 reactivation from latency. The number of TG neurons and cells in pharyngeal tonsil expressing the cellular transcription factor specificity protein 1 (Sp1) protein increases during early stages of dexamethasone-induced reactivation from latency. Silencing Sp1 expression impairs BoHV-1 replication in permissive cells. Interestingly, mithramycin A, a neuroprotective antibiotic that preferentially binds GC-rich DNA, impairs Sp1 functions and reduces BoHV-1 replication suggesting that it is a potential antiviral drug. The glucocorticoid receptor (GR) and Sp1 cooperatively transactivate the BoHV-1 immediate early transcript unit 1 (IEtu1) promoter, which drives expression of infected cell protein 0 (bICP0) and bICP4. Mithramycin A also reduced Sp1- and GR-mediated transactivation of the IEtu1 promoter. These studies revealed that GR and Sp1 trigger viral gene expression and replication following stressful stimuli.


Subject(s)
Herpesviridae Infections , Herpesvirus 1, Bovine , Receptors, Glucocorticoid , Sp1 Transcription Factor , Animals , Cattle , Mice , Adrenal Cortex Hormones/metabolism , Dexamethasone/pharmacology , DNA/metabolism , Herpesvirus 1, Bovine/physiology , Plicamycin/analogs & derivatives , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Transcription Factors/metabolism , Viral Proteins/metabolism , Sp1 Transcription Factor/metabolism
4.
Horm Metab Res ; 56(1): 78-90, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37884032

ABSTRACT

Endocrine-disrupting chemicals (EDCs) are exogenous substances known to interfere with endocrine homeostasis and promote adverse health outcomes. Their impact on the adrenal cortex, corticosteroids and their physiological role in the organism has not yet been sufficiently elucidated. In this review, we collect experimental and epidemiological evidence on adrenal disruption by relevant endocrine disruptors. In vitro data suggest significant alterations of gene expression, cell signalling, steroid production, steroid distribution, and action. Additionally, morphological studies revealed disturbances in tissue organization and development, local inflammation, and zone-specific hyperplasia. Finally, endocrine circuits, such as the hypothalamic-pituitary-adrenal axis, might be affected by EDCs. Many questions regarding the detection of steroidogenesis disruption and the effects of combined toxicity remain unanswered. Not only due to the diverse mode of action of adrenal steroids and their implication in many common diseases, there is no doubt that further research on endocrine disruption of the adrenocortical system is needed.


Subject(s)
Adrenal Cortex , Endocrine Disruptors , Adrenal Cortex/metabolism , Adrenal Cortex Hormones/metabolism , Endocrine Disruptors/toxicity , Hypothalamo-Hypophyseal System , Pituitary-Adrenal System , Steroids/metabolism
5.
Gen Comp Endocrinol ; 352: 114514, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38582175

ABSTRACT

Hormonal influence on hepatic function is a critical aspect of whole-body energy balance in vertebrates. Catecholamines and corticosteroids both influence hepatic energy balance via metabolite mobilization through glycogenolysis and gluconeogenesis. Elasmobranchs have a metabolic organization that appears to prioritize the mobilization of hepatic lipid as ketone bodies (e.g. 3-hydroxybutyrate [3-HB]), which adds complexity in determining the hormonal impact on hepatic energy balance in this taxon. Here, a liver perfusion was used to investigate catecholamine (epinephrine [E]) and corticosteroid (corticosterone [B] and 11-deoxycorticosterone [DOC]) effects on the regulation of hepatic glucose and 3-HB balance in the North Pacific Spiny dogfish, Squalus suckleyi. Further, hepatic enzyme activity involved in ketogenesis (3-hydroxybutyrate dehydrogenase), glycogenolysis (glycogen phosphorylase), and gluconeogenesis (phosphoenolpyruvate carboxykinase) were assessed in perfused liver tissue following hormonal application to discern effects on hepatic energy flux. mRNA transcript abundance key transporters of glucose (glut1 and glut4) and ketones (mct1 and mct2) and glucocorticoid function (gr, pepck, fkbp5, and 11ßhsd2) were also measured to investigate putative cellular components involved in hepatic responses. There were no changes in the arterial-venous difference of either metabolite in all hormone perfusions. However, perfusion with DOC increased gr transcript abundance and decreased flow rate of perfusions, suggesting a regulatory role for this corticosteroid. Phosphoenolpyruvate carboxykinase activity increased following all hormone treatments, which may suggest gluconeogenic function; E also increased 3-hydroxybutyrate dehydrogenase activity, suggesting a function in ketogenesis, and decreased pepck and fkbp5 transcript abundance, potentially showing some metabolic regulation. Overall, we demonstrate hormonal control of hepatic energy balance using liver perfusions at various levels of biological organization in an elasmobranch.


Subject(s)
Squalus acanthias , Squalus , Animals , Glucose/metabolism , Squalus/metabolism , Squalus acanthias/metabolism , Hydroxybutyrate Dehydrogenase/metabolism , Phosphoenolpyruvate/metabolism , Liver/metabolism , 3-Hydroxybutyric Acid/pharmacology , 3-Hydroxybutyric Acid/metabolism , Ketone Bodies/metabolism , Gluconeogenesis , Hormones/metabolism , Adrenal Cortex Hormones/metabolism
6.
Int J Mol Sci ; 24(15)2023 Jul 27.
Article in English | MEDLINE | ID: mdl-37569423

ABSTRACT

Placental protein 13 (PP13) is a regulatory protein involved in remodeling the vascular system of the pregnancy and extending the immune tolerance of the mother to the growing fetus. PP13 is localized on the surface of the syncytiotrophoblast. An ex vivo placental model shows that the PP13 is released via placental-associated extracellular vesicles (PEVs) to the maternal uterine vein. This exploratory study aimed to determine PEV-associated PP13 in the maternal circulation as compared to the known soluble fraction since each has a specific communication pathway. Patients admitted to Bnai Zion Medical Center for delivery were recruited, and included 19 preeclampsia (PE) patients (7 preterm PE gestational age < 37 weeks' gestation), 16 preterm delivery (PTD, delivery at GA < 37 weeks' gestation), and 15 matched term delivery controls. Treatment by corticosteroids (Celestone), which is often given to patients with suspected preterm PE and PTD, was recorded. The PEV proteome was purified from the patients' plasma by size exclusion chromatography (SEC) to separate the soluble and PEV-associated PP13. The total level of PP13 (soluble and PEV-associated) was determined using mild detergent that depleted the PEV proteome. PP13 fractions were determined by ELISA with PP13 specific antibodies. ELISA with alkaline phosphatase (PLAP)- and cluster differentiation 63 (CD63)-specific antibodies served to verify the placental origin of the PEVs. SPSS was used for statistical analysis. The patients' medical, pregnancy, and delivery records in all groups were similar except, as expected, that a larger number of PE and PTD patients had smaller babies who were delivered earlier, and the PE patients had hypertension and proteinuria. The SEC analysis detected the presence of PP13 in the cargo of the PEVs and on their surface, in addition to the known soluble fraction. The median soluble PP13 was not significantly different across the PE, PTD, and term delivery control groups. However, after depleting the PEV of their proteome, the total PP13 (soluble and PEV-associated) was augmented in the cases of preterm PE, reaching 2153 pg/mL [IQR 1866-2838] but not in cases of PTD reaching 1576 pg/mL [1011-2014] or term delivery groups reaching 964 pg/mL [875-1636]), p < 0.01. On the surface of the circulating PEV from PTD patients, there was a decrease in PP13. Corticosteroid treatment was accompanied by a massive depletion of PP13 from the PEV, especially in preterm PE patients. This exploratory study is, thus, the first to determine PEV-associated PP13 in maternal circulation, providing a quantitative determination of the soluble and the PEV-associated fractions, and it shows that the latter is the larger. We found an increase in the amount of PP13 carried via the PEV-associated pathway in PE and PTD patients compared to term delivery cases, which was further augmented when the patients were treated with corticosteroids, especially in preterm PE. The signal conveyed by this novel communication pathway warrants further research to investigate these two differential pathways for the liberation of PP13.


Subject(s)
Extracellular Vesicles , Pre-Eclampsia , Female , Humans , Infant , Infant, Newborn , Pregnancy , Adrenal Cortex Hormones/metabolism , Biomarkers/metabolism , Extracellular Vesicles/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Proteome/metabolism
7.
Vet Surg ; 52(3): 361-369, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36571324

ABSTRACT

OBJECTIVE: To determine the effect of concurrent versus delayed treatment with corticosteroid on equine articular tissues also treated with local anesthetic in vitro in the presence of inflammatory mediators. STUDY DESIGN: Controlled laboratory study. ANIMALS: Five geldings, one mare (aged 3-18 years). METHODS: From each horse, 24 synovial and 12 osteochondral explants were cultured in a 12-well plate (2 wells/group, 2 synovial and 1 osteochondral explant/well, total 216 explants in the study). Explants were stimulated in culture medium with 10 µg/ml recombinant equine interleukin-1ß and 10 µg/ml tumor necrosis factor-α for 48 hours, then randomly assigned to six treatments: unstimulated control, stimulated control, triamcinolone acetonide (TA, 10-6  M), mepivacaine hydrochloride (MH, 4.4 mg/ml), MH + TA (concurrent) and MH + TA (delayed). The delayed group was treated with MH and, 6 days later, treated with TA. Every 3 days for 9 days total, medium levels of lactate dehydrogenase (LDH), prostaglandin E2 (PGE2 ), matrix metalloproteinase 13 (MMP-13) and glycosaminoglycan (GAG) were quantified via ELISA. Data were analyzed with mixed-effects models with Tukey's multiple comparisons. RESULTS: Stimulation increased medium PGE2 and MMP-13 and had no effect on LDH or GAG. Treatment with MH increased LDH and decreased PGE2 and MMP-13. Treatment with TA decreased PGE2 and MMP-13. CONCLUSION: There were no differences in cytotoxicity, inflammation or matrix degradation for delayed or concurrent MH and TA treatment groups up to 9 days in culture. CLINICAL SIGNIFICANCE: The lack of an effect of concurrent versus delayed treatment might indicate that concurrent therapy is acceptable.


Subject(s)
Anesthetics, Local , Cartilage, Articular , Horses , Animals , Male , Female , Anesthetics, Local/pharmacology , Anesthetics, Local/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 13/pharmacology , Adrenal Cortex Hormones/metabolism , Adrenal Cortex Hormones/pharmacology , Triamcinolone Acetonide/metabolism , Triamcinolone Acetonide/pharmacology , Glycosaminoglycans/analysis , Glycosaminoglycans/metabolism , Glycosaminoglycans/pharmacology
8.
Int J Mol Sci ; 24(1)2022 Dec 29.
Article in English | MEDLINE | ID: mdl-36614027

ABSTRACT

The human adrenal cortex is composed of distinct zones that are the main source of steroid hormone production. The mechanism of adrenocortical cell differentiation into several functionally organized populations with distinctive identities remains poorly understood. Human adrenal disease has been difficult to study, in part due to the absence of cultured cell lines that faithfully represent adrenal cell precursors in the early stages of transformation. Here, Human Adrenocortical Adenoma (HAA1) cell line derived from a patient's macronodular adrenocortical hyperplasia and was treated with histone deacetylase inhibitors (HDACis) and gene expression was examined. We describe a patient-derived HAA1 cell line derived from the zona reticularis, the innermost zone of the adrenal cortex. The HAA1 cell line is unique in its ability to exit a latent state and respond with steroidogenic gene expression upon treatment with histone deacetylase inhibitors. The gene expression pattern of differentiated HAA1 cells partially recreates the roster of genes in the adrenal layer that they have been derived from. Gene ontology analysis of whole genome RNA-seq corroborated increased expression of steroidogenic genes upon HDAC inhibition. Surprisingly, HDACi treatment induced broad activation of the Tumor Necrosis Factor (TNF) alpha pathway. This novel cell line we developed will hopefully be instrumental in understanding the molecular and biochemical mechanisms controlling adrenocortical differentiation and steroidogenesis.


Subject(s)
Adrenal Cortex , Adrenocortical Adenoma , Humans , Zona Reticularis/metabolism , Adrenocortical Adenoma/genetics , Adrenocortical Adenoma/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/metabolism , Adrenal Cortex Hormones/metabolism , Cell Line
9.
Am J Physiol Lung Cell Mol Physiol ; 321(6): L1194-L1205, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34755542

ABSTRACT

Corticosteroid insensitivity in asthma limits the ability to effectively manage severe asthma, which is characterized by persistent airway inflammation, airway hyperresponsiveness (AHR), and airflow obstruction despite corticosteroid treatment. Recent reports indicate that corticosteroid insensitivity is associated with increased interferon-γ (IFN-γ) levels and T-helper (Th) 1 lymphocyte infiltration in severe asthma. Signal transducer and activator of transcription 1 (STAT1) activation by IFN-γ is a key signaling pathway in Th1 inflammation; however, its role in the context of severe allergic airway inflammation and corticosteroid sensitivity remains unclear. In this study, we challenged wild-type (WT) and Stat1-/- mice with mixed allergens (MA) augmented with c-di-GMP [bis-(3'-5')-cyclic dimeric guanosine monophosphate], an inducer of Th1 cell infiltration with increased eosinophils, neutrophils, Th1, Th2, and Th17 cells. Compared with WT mice, Stat1-/- had reduced neutrophils, Th1, and Th17 cell infiltration. To evaluate corticosteroid sensitivity, mice were treated with either vehicle, 1 or 3 mg/kg fluticasone propionate (FP). Corticosteroids significantly reduced eosinophil infiltration and cytokine levels in both c-di-GMP + MA-challenged WT and Stat1-/- mice. However, histological and functional analyses show that corticosteroids did not reduce airway inflammation, epithelial mucous cell abundance, airway smooth muscle mass, and AHR in c-di-GMP + MA-challenged WT or Stat1-/- mice. Collectively, our data suggest that increased Th1 inflammation is associated with a decrease in corticosteroid sensitivity. However, increased airway pathology and AHR persist in the absence of STAT1 indicate corticosteroid insensitivity in structural airway cells is a STAT1 independent process.


Subject(s)
Adrenal Cortex Hormones/metabolism , Inflammation/metabolism , STAT1 Transcription Factor/metabolism , Allergens/metabolism , Animals , Asthma/metabolism , Eosinophils/metabolism , Female , Hypersensitivity/metabolism , Interferon-gamma/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Respiratory Hypersensitivity/metabolism , T-Lymphocytes, Helper-Inducer/metabolism
10.
Gen Comp Endocrinol ; 307: 113756, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33741310

ABSTRACT

Anadromous sea lamprey (Petromyzon marinus) larvae undergo a months-long true metamorphosis during which they develop seawater (SW) tolerance prior to downstream migration and SW entry. We have previously shown that intestinal Na+/K+-ATPase (NKA) activity increases during metamorphosis and is critical to the osmoregulatory function of the intestine in SW. The present study investigated the role of 11-deoxycortisol (S) in controlling NKA in the anterior (AI) and posterior (PI) intestine during sea lamprey metamorphosis. In a tissue profile, nka mRNA and protein were most abundant in the gill, kidney, and AI. During metamorphosis, AI nka mRNA increased 10-fold, whereas PI nka mRNA did not change. Specific corticosteroid receptors were found in the AI, which had a higher binding affinity for S compared to 11-deoxycorticosterone (DOC). In vivo administration of S in mid-metamorphic lamprey upregulated NKA activity 3-fold in the AI and PI, whereas administration of DOC did not affect intestinal NKA activity. During a 24 h SW challenge test, dehydration of white muscle moisture was rescued by prior treatment with S, which was associated with increased intestinal nka mRNA and NKA activity. These results indicate that intestinal osmoregulation in sea lamprey is a target for control by S during metamorphosis and the development of SW tolerance.


Subject(s)
Petromyzon , Adrenal Cortex Hormones/metabolism , Animals , Gills/metabolism , Intestines , Osmoregulation , Petromyzon/metabolism , Seawater , Sodium-Potassium-Exchanging ATPase/metabolism
11.
Article in English | MEDLINE | ID: mdl-34237466

ABSTRACT

The neuroendocrine mechanism underlying stress responses in vertebrates is hypothesized to be highly conserved and evolutionarily ancient. Indeed, elements of this mechanism, from the brain to steroidogenic tissue, are present in all vertebrate groups; yet, evidence of the function and even identity of some elements of the hypothalamus-pituitary-adrenal/interrenal (HPA/I) axis is equivocal among the most basal vertebrates. The purpose of this review is to discuss the functional evolution of the HPA/I axis in vertebrates with a focus on our understanding of this neuroendocrine mechanism in the most ancient vertebrates: the agnathan (i.e., hagfish and lamprey) and chondrichthyan fishes (i.e., sharks, rays, and chimeras). A review of the current literature presents evidence of a conserved HPA/I axis in jawed vertebrates (i.e., gnathostomes); yet, available data in jawless (i.e., agnathan) and chondrichthyan fishes are limited. Neuroendocrine regulation of corticosteroidogenesis in agnathans and chondrichthyans appears to function through similar pathways as in bony fishes and tetrapods; however, key elements have yet to be identified and the involvement of melanotropins and gonadotropin-releasing hormone in the stress axis in these ancient fishes warrants further investigation. Further, the identities of physiological glucocorticoids are uncertain in hagfishes, chondrichthyans, and even coelacanths. Resolving these and other knowledge gaps in the stress response of ancient fishes will be significant for advancing knowledge of the evolutionary origins of the vertebrate stress response.


Subject(s)
Adrenal Cortex Hormones/metabolism , Hagfishes/physiology , Hypothalamo-Hypophyseal System , Lampreys/physiology , Stress, Physiological , Animals , Biological Evolution , Corticosterone/analogs & derivatives , Corticosterone/metabolism , Cortodoxone/metabolism , Glucocorticoids/metabolism , Hagfishes/genetics , Lampreys/genetics , Neurosecretory Systems/physiology , Phylogeny , Pituitary-Adrenal System , Vertebrates
12.
Am J Otolaryngol ; 42(6): 103058, 2021.
Article in English | MEDLINE | ID: mdl-33932624

ABSTRACT

OBJECTIVE: We have evaluated that the deposition patterns of corticosteroid nasal spray in the sinonasal cavity of both post-operated human cases, which were further compared with a computed tomography-based sinonasal airway model. METHODS: Fifty-one patients with chronic rhinosinusitis following an endoscopic sinus surgery were enrolled in this study. Nasal spray mometasone furoate hydrate (Nasonex®) containing 0.1% indigocarmine was applied to the patients' nasal cavities and the sinonasal cavity was observed by endoscopy and video documentation. A single plaster sinonasal model was used to quantify the sinonasal deposition of nasal sprays containing 10% red ink solution using 12 round paper strips. RESULTS: The predominant areas of the spray deposition of the operated sinonasal cavities were recognized in the ethmoid sinus and the olfactory cleft in the human study. The droplets were mainly deposited in the inferior turbinate followed by the posterior part of the ethmoid sinus, the olfactory cleft, and anterior part of the ethmoid sinus in a sinonasal model. CONCLUSION: The corticosteroid nasal spray efficiently reached the olfactory cleft and the ethmoid sinus in post-operative conditions, which was demonstrated by post-operated human cases and a computed tomography-based sinonasal airway model.


Subject(s)
Adrenal Cortex Hormones/administration & dosage , Adrenal Cortex Hormones/metabolism , Coloring Agents/administration & dosage , Coloring Agents/metabolism , Endoscopy/methods , Indigo Carmine/administration & dosage , Indigo Carmine/metabolism , Mometasone Furoate/administration & dosage , Mometasone Furoate/metabolism , Nasal Sprays , Paranasal Sinuses/metabolism , Paranasal Sinuses/surgery , Rhinitis/surgery , Silicones , Sinusitis/surgery , Adolescent , Adult , Aged , Chronic Disease , Female , Humans , Male , Middle Aged , Models, Anatomic , Paranasal Sinuses/diagnostic imaging , Rhinitis/metabolism , Sinusitis/metabolism , Tomography, X-Ray Computed , Young Adult
13.
Int J Mol Sci ; 22(10)2021 May 18.
Article in English | MEDLINE | ID: mdl-34069759

ABSTRACT

Sexual dimorphism involves differences between biological sexes that go beyond sexual characteristics. In mammals, differences between sexes have been demonstrated regarding various biological processes, including blood pressure and predisposition to develop hypertension early in adulthood, which may rely on early events during development and in the neonatal period. Recent studies suggest that corticosteroid signaling pathways (comprising glucocorticoid and mineralocorticoid signaling pathways) have distinct tissue-specific expression and regulation during this specific temporal window in a sex-dependent manner, most notably in the kidney. This review outlines the evidence for a gender differential expression and activation of renal corticosteroid signaling pathways in the mammalian fetus and neonate, from mouse to human, that may favor mineralocorticoid signaling in females and glucocorticoid signaling in males. Determining the effects of such differences may shed light on short term and long term pathophysiological consequences, markedly for males.


Subject(s)
Adrenal Cortex Hormones/metabolism , Kidney/embryology , Aldosterone/metabolism , Animals , Blood Pressure/physiology , Gene Expression Regulation, Developmental/genetics , Glucocorticoids/metabolism , Humans , Hypertension/metabolism , Kidney/metabolism , Mineralocorticoids/metabolism , Organogenesis , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , Sex Characteristics , Signal Transduction/physiology
14.
J Neuroinflammation ; 17(1): 318, 2020 Oct 25.
Article in English | MEDLINE | ID: mdl-33100225

ABSTRACT

BACKGROUND: We previously found that high-dose methylprednisolone increased the incidence of critical illness-related corticosteroid insufficiency (CIRCI) and mortality in rats with traumatic brain injury (TBI), whereas low-dose hydrocortisone but not methylprednisolone exerted protective effects. However, the receptor-mediated mechanism remains unclear. This study investigated the receptor-mediated mechanism of the opposite effects of different glucocorticoids on the survival of paraventricular nucleus (PVN) cells and the incidence of CIRCI after TBI. METHODS: Based on controlled cortical impact (CCI) and treatments, male SD rats (n = 300) were randomly divided into the sham, CCI, CCI + GCs (methylprednisolone 1 or 30 mg/kg/day; corticosterone 1 mg/kg/day), CCI + methylprednisolone+RU486 (RU486 50 mg/kg/day), and CCI + corticosterone+spironolactone (spironolactone 50 mg/kg/day) groups. Blood samples were collected 7 days before and after CCI. Brain tissues were collected on postinjury day 7 and processed for histology and western blot analysis. RESULTS: We examined the incidence of CIRCI, mortality, apoptosis in the PVN, the receptor-mediated mechanism, and downstream signaling pathways on postinjury day 7. We found that methylprednisolone and corticosterone exerted opposite effects on the survival of PVN cells and the incidence of CIRCI by activating different receptors. High-dose methylprednisolone increased the nuclear glucocorticoid receptor (GR) level and subsequently increased cell loss in the PVN and the incidence of CIRCI. In contrast, low-dose corticosterone but not methylprednisolone played a protective role by upregulating mineralocorticoid receptor (MR) activation. The possible downstream receptor signaling mechanism involved the differential effects of GR and MR on the activity of the Akt/CREB/BDNF pathway. CONCLUSION: The excessive activation of GR by high-dose methylprednisolone exacerbated apoptosis in the PVN and increased CIRCI. In contrast, refilling of MR by corticosterone protects PVN neurons and reduces the incidence of CIRCI by promoting GR/MR rebalancing after TBI.


Subject(s)
Brain Injuries, Traumatic/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Steroid/metabolism , Adrenal Cortex Hormones/metabolism , Animals , Brain Injuries, Traumatic/pathology , Cell Survival/physiology , Critical Illness/therapy , Glucocorticoids/pharmacology , Male , Methylprednisolone/pharmacology , Paraventricular Hypothalamic Nucleus/pathology , Rats , Rats, Sprague-Dawley
15.
Cell Immunol ; 356: 104177, 2020 10.
Article in English | MEDLINE | ID: mdl-32795666

ABSTRACT

Alpha1-antitrypsin (AAT) is a serum protease inhibitor that rises during inflammation and healthy pregnancies. Plasma-derived AAT, indicated for genetic AAT deficiency, is presently being explored for additional medical indications. Unlike corticosteroids, some anti-inflammatory activities of AAT involve NF-κB-dependent outcomes, e.g., induction of IL-1R antagonist. AAT activities were compared to dexamethasone (DEX), using various in-vitro cells assays, animal studies, and NF-κB-p65 localization and activity studies. Results demonstrate a cytokine shift towards resolution in AAT-treated cells, as opposed to pan-suppression in DEX-treated cells. AAT enhanced, while DEX suppressed LPS-induced IL-1Ra production and re-epithelialization. When drugs were combined, AAT allowed the immunosuppressive DEX activities, while DEX at medium to high levels antagonized beneficial AAT effects. Interestingly, lower levels of DEX maintained the immunosuppressive effect, while allowing upregulation of IL-1Ra. Therefore, AAT may represent a distinct endogenous anti-inflammatory, resolution-promoting agent that may improve tissue well-being while preventing undesired corticostroids side effects.


Subject(s)
Adrenal Cortex Hormones/metabolism , Inflammation/metabolism , alpha 1-Antitrypsin/metabolism , A549 Cells , Adrenal Cortex Hormones/physiology , Animals , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Dexamethasone/metabolism , Dexamethasone/pharmacology , Humans , Interleukin 1 Receptor Antagonist Protein/metabolism , Mice , NF-kappa B/metabolism , RAW 264.7 Cells , Signal Transduction/genetics , THP-1 Cells , Tumor Necrosis Factor-alpha/metabolism , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/pharmacology , alpha 1-Antitrypsin/physiology
16.
Respir Res ; 21(1): 279, 2020 Oct 21.
Article in English | MEDLINE | ID: mdl-33087119

ABSTRACT

BACKGROUND: Tezepelumab is a human monoclonal antibody that blocks the activity of the epithelial cytokine thymic stromal lymphopoietin. The efficacy, safety and oral corticosteroid-sparing potential of tezepelumab are being investigated in two ongoing, phase 3, randomized, double-blind, placebo-controlled studies (NAVIGATOR [NCT03347279] and SOURCE [NCT03406078]). DESTINATION (NCT03706079) is a long-term extension (LTE) of these studies. METHODS: DESTINATION is a randomized, double-blind, placebo-controlled LTE study in adults (18-80 years old) and adolescents (12-17 years old) with severe, uncontrolled asthma who are receiving treatment with medium- or high-dose inhaled corticosteroids plus at least one additional controller medication with or without oral corticosteroids. The study population will comprise patients who complete the 52- and 48-week NAVIGATOR and SOURCE studies, respectively. Patients who were randomized to receive tezepelumab 210 mg every 4 weeks (Q4W) in either predecessor study will continue to receive this regimen for 1 year; those who were previously randomized to receive placebo will be re-randomized (1:1) to receive either tezepelumab 210 mg Q4W or placebo for 1 year. Patients will receive their prescribed controller medications throughout DESTINATION and study physicians will have the opportunity to down- or up-titrate dosage of these medications, if appropriate. The primary objective is to evaluate the long-term safety and tolerability of tezepelumab over 104 weeks (inclusive of the treatment period of either predecessor study). The secondary objective is to assess the long-term effect of tezepelumab on asthma exacerbations. Patients recruited from SOURCE will be followed up post-treatment for 12 weeks. Patients recruited from NAVIGATOR who complete 100 weeks of tezepelumab treatment will be eligible for either 12 weeks of follow-up or a 36-week extended follow-up during which the clinical benefit of tezepelumab after treatment cessation will be investigated. DISCUSSION: DESTINATION will evaluate the long-term safety, tolerability and efficacy of tezepelumab versus placebo with continued dosing for up to 2 years. DESTINATION will also evaluate the clinical effect of tezepelumab after treatment cessation. This LTE study aims to elucidate the long-term safety implications of receiving tezepelumab and to assess its potential long-term treatment benefits in patients with severe, uncontrolled asthma. TRIAL REGISTRATION: NCT03706079 (ClinicalTrials.gov). Registered 15 October 2018.


Subject(s)
Anti-Asthmatic Agents/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Asthma/drug therapy , Severity of Illness Index , Administration, Inhalation , Adolescent , Adrenal Cortex Hormones/administration & dosage , Adrenal Cortex Hormones/adverse effects , Adrenal Cortex Hormones/metabolism , Adult , Aged , Aged, 80 and over , Anti-Asthmatic Agents/adverse effects , Anti-Asthmatic Agents/metabolism , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/metabolism , Asthma/diagnosis , Asthma/metabolism , Cytokines/metabolism , Double-Blind Method , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Male , Middle Aged , Young Adult
17.
Handb Exp Pharmacol ; 261: 25-37, 2020.
Article in English | MEDLINE | ID: mdl-31375923

ABSTRACT

Drug development and pharmacotherapy of rare pediatric diseases have significantly expanded over the last decade, in part due to incentives and financial support provided by governments, regulators, and nonprofit foundations. Duchenne muscular dystrophy (DMD) is among the most common rare pediatric disorders, and clinical trials of therapeutic approaches have seen dramatic expansion. Pharmacotherapeutic standard of care has been limited to off-label prescription of high-dose, daily corticosteroids (prednisone, deflazacort). Deflazacort received FDA approval for DMD in 2016, although the price increases associated with formal FDA approval and the severe side effects associated with corticosteroid use have limited patient/physician uptake and insurance coverage in the USA. In Europe, EMA has given conditional marketing authorization for prescription of Translarna (a stop codon read-through drug prescribed to ~10% of DMD patients), although there is not yet evidence of clinical efficacy. The FDA awarded conditional approval to etiplirsen, an exon-skipping oligonucleotide drug, based on accelerated pathways (increased dystrophin production in patient muscle). Evidence of clinical efficacy remains the focus of post-marketing studies. There are many innovative pharmacotherapies under clinical development for DMD (Phase I, II, and III clinical trials). All are "disease modifying" in the sense that none seek to replace the full-length, normal DMD gene or dystrophin protein, but instead either seek to introduce an abnormal "Becker-like" version of the gene or protein or target pathophysiological pathways downstream of the primary defect. It is envisioned that the most significant benefit to DMD patients will be through multidrug approaches simultaneously aiming to introduce partially functional dystrophin in patient muscle while also targeting both chronic inflammation and the fibrofatty replacement of muscle.


Subject(s)
Muscular Dystrophy, Duchenne , Adrenal Cortex Hormones/genetics , Adrenal Cortex Hormones/metabolism , Adrenal Cortex Hormones/pharmacology , Child , Dystrophin/genetics , Dystrophin/metabolism , Dystrophin/physiology , Exons/physiology , Humans
18.
Handb Exp Pharmacol ; 261: 131-151, 2020.
Article in English | MEDLINE | ID: mdl-32076895

ABSTRACT

The management of atopic diseases such as severe asthma, severe atopic dermatitis, and severe food allergy in childhood is challenging. In particular, there are safety concerns regarding the use of high-dose corticosteroids. The recent development of biologicals and their approval for the treatment of children offer a new, very promising, and more personalized therapy option. Omalizumab, mepolizumab, and dupilumab are currently approved as add-on treatments of severe asthma in children and have been shown to be effective in improvement of asthma control and reduction of exacerbations. Dupilumab is the only biological approved for the treatment of atopic dermatitis in adolescents so far. It has been demonstrated to significantly improve symptoms of atopic dermatitis.However, safety data for biologicals used in atopic diseases in children and adolescents are still very limited. Biologicals are generally considered to be safe in adults. These data are often extrapolated to children. Additionally, data for long-term use are lacking. Thus, the safety profiles of those biologicals cannot yet be conclusively assessed.


Subject(s)
Asthma , Biological Products , Dermatitis, Atopic , Adolescent , Adrenal Cortex Hormones/chemistry , Adrenal Cortex Hormones/genetics , Adrenal Cortex Hormones/metabolism , Adult , Child , Humans , Omalizumab/chemistry
19.
Int J Mol Sci ; 21(18)2020 Sep 14.
Article in English | MEDLINE | ID: mdl-32938015

ABSTRACT

Some fish have acquired the ability to breathe air, but these fish can no longer flush their gills effectively when out of water. Hence, they have developed characteristic means for defense against external stressors, including thirst (osmolarity/ions) and toxicity. Amphibious fish, extant air-breathing fish emerged from water, may serve as models to examine physiological responses to these stressors. Some of these fish, including mudskipper gobies such as Periophthalmodon schlosseri, Boleophthalmus boddarti and our Periophthalmus modestus, display distinct adaptational behaviors to these factors compared with fully aquatic fish. In this review, we introduce the mudskipper goby as a unique model to study the behaviors and the neuro/endocrine mechanisms of behavioral responses to the stressors. Our studies have shown that a local sensation of thirst in the buccal cavity-this being induced by dipsogenic hormones-motivates these fish to move to water through a forebrain response. The corticosteroid system, which is responsive to various stressors, also stimulates migration, possibly via the receptors in the brain. We suggest that such fish are an important model to deepen insights into the stress-related neuro/endocrine-behavioral effects.


Subject(s)
Adrenal Cortex Hormones/metabolism , Amphibians/physiology , Cheek/physiology , Fishes/physiology , Neurosecretory Systems/physiology , Sensation/physiology , Amphibians/metabolism , Animals , Brain/metabolism , Brain/physiology , Fishes/metabolism , Models, Animal
20.
J Environ Sci Health B ; 55(2): 91-102, 2020.
Article in English | MEDLINE | ID: mdl-31524045

ABSTRACT

Information regarding the enantioselective endocrine disruption of chiral herbicides is scarce. This study assessed the disrupting effects of eight typical chiral herbicides on corticosteroids (including glucocorticoids and mineralocorticoids). Enantioselectivity of eight chiral herbicides were evaluated for their agonistic/antagonistic effects on glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) with CHOK1 cell line using reporter gene assay. Their influence on the production of corticosteroids were further investigated in H295R cell line using enzyme-linked immunosorbent assay (ELISA). None of the racemates or enantiomers of eight chiral herbicides exhibited GR or MR agonistic activity at non-cytotoxic concentrations. However, rac-propisochlor and S-imazamox antagonized cortisol-induced transactivation of GR by 21.79% and 38.73% at the concentration of 1.0 × 10-7 M and 1.0 × 10-6 M, respectively, and R-napropamide remarkably attenuated aldosterone-induced MR transactivation by 68.78% at 1.0 × 10-6 M. The secretion of cortisol was significantly restrained after treated with 1.0 × 10-6 M rac-propisochlor and rac-/R-napropamide at the concentration of 1.0 × 10-6 M by 26.49%, 30.10% and 35.27%, respectively, while this glucocorticoid was remarkably induced by 1.0 × 10-5 M rac-diclofop-methyl and its two enantiomers at the concentration of 1.0 × 10-5 M by 75.60%, 100.1% and 68.78%, respectively. Exposure to rac-propisochlor (1.0 × 10-6 M), S-diclofop-methyl (1.0 × 10-5 M) or rac-/S-/R- acetochlor (1.0 × 10-6 M) and rac-/S-/R-lactofen (1.0 × 10-6 M) inhibited the secretion of aldosterone by approximately 40%. Our findings suggested that chiral herbicides disrupted corticosteroid homeostasis in an enantioselective way. Therefore, more comprehensive screening is required to better understand the ecological and health risks of chiral pesticides.


Subject(s)
Adrenal Cortex Hormones/metabolism , Herbicides/chemistry , Herbicides/toxicity , Aldosterone/metabolism , Animals , CHO Cells , Cell Line , Cricetulus , Endocrine Disruptors/chemistry , Endocrine Disruptors/toxicity , Humans , Hydrocortisone/metabolism , Mineralocorticoid Receptor Antagonists/chemistry , Mineralocorticoid Receptor Antagonists/toxicity , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/agonists , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL