Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 404
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Endocrinol Invest ; 41(2): 233-240, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28730425

ABSTRACT

PURPOSE: Gossypol, a naturally occurring compound in cottonseeds, has anticancer effects against several tumor cell lines. It has been extensively studied in clinical trials and is well tolerated with a favorable safety profile. AT-101, a derivative of R (-)-gossypol, binds to Bcl-2 family proteins and induces apoptosis in vitro. Although transsphenoidal surgical excision of the pituitary corticotroph adenoma is the gold standard of care, it is not successful all the time. Medical therapy for Cushing's disease still remains a challenge for the clinicians. We aimed to investigate the cytotoxic and apoptotic effects of AT-101 in mouse pituitary corticotroph tumor AtT20 cells. METHODS: Cytotoxic effect of AT-101 was assessed by XTT cell viability assay. Apoptosis was shown by measuring DNA fragmentation and Caspase-3/7 activity. Changes in mRNA expressions of apoptosis-related genes were investigated by qPCR array after treatment with AT-101. ACTH was measured by ACTH-EIA Kit. RESULTS: AT-101 induced cytotoxicity and apoptosis in AtT20 cells. mRNA levels of pro-apoptotic genes such as TNFR-SF-10B, Bid, PYCARD, Caspase-8, Caspase-3, and Caspase-7 were induced by 2.0-, 1.5-, 1.7-, 1.5-, 1.6-, and 2-fold, respectively, in AtT20 cells by AT-101 treatment. Moreover, some of the anti-apoptotic genes such as BCL2L10, NAIP1, and PAK-7 were reduced by 2.1-, 2.3-, 4.0-fold, respectively, in AtT20 cells. AT-101 also decreased ACTH secretion significantly. CONCLUSION: AT-101 induces apoptosis in mouse pituitary corticotroph tumor cells.


Subject(s)
ACTH-Secreting Pituitary Adenoma/drug therapy , Adenoma/drug therapy , Adrenocorticotropic Hormone/antagonists & inhibitors , Apoptosis/drug effects , Cell Proliferation/drug effects , Gossypol/analogs & derivatives , Pituitary Neoplasms/drug therapy , ACTH-Secreting Pituitary Adenoma/metabolism , ACTH-Secreting Pituitary Adenoma/pathology , Adenoma/metabolism , Adenoma/pathology , Adrenocorticotropic Hormone/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis Regulatory Proteins/metabolism , Gossypol/pharmacology , Mice , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Tumor Cells, Cultured
2.
Endocr J ; 62(2): 145-51, 2015.
Article in English | MEDLINE | ID: mdl-25342092

ABSTRACT

Combination treatment with intravenous immunoglobulin (IVIG) plus prednisolone, newly designed for children with severe Kawasaki disease (KD), reduces coronary artery abnormalities significantly. As prednisolone is administered for approximately 20 days in this regimen, we examined whether adrenal function of the treated patients is suppressed. A prospective study was performed at one medical institution in 21 children with KD (age range 0.3-10.4 years, median 3.1 years) who were treated with the regimen between February and June, 2012. We assessed cortisol and ACTH values before the initiation and after the cessation of prednisolone administration as well as peak cortisol and ACTH values at corticotropin-releasing hormone (CRH) stimulation tests, which were repeated 0, 2, and 6 months after the treatment. Morning cortisol and ACTH values after the cessation of prednisolone treatment were suppressed. Peak cortisol values at the first CRH stimulation test ranged from 5.1 to 25.4 µg/dL and were less than 20 µg/dL in 17 of 21 patients, but were restored to more than 14.6 µg/dL in all patients by 6 months after the prednisolone treatment. A significant positive correlation was observed between cortisol values at 09:00 h after the prednisolone treatment and peak cortisol values at the following CRH stimulation test (r = 0.727, p < 0.001). We conclude that adrenal suppression can occur in a high proportion of children with KD treated with IVIG plus prednisolone, despite rather short duration and relatively small amounts of administered glucocorticoids.


Subject(s)
Adrenal Glands/drug effects , Adrenal Insufficiency/chemically induced , Anti-Inflammatory Agents/adverse effects , Glucocorticoids/adverse effects , Immunoglobulins, Intravenous/adverse effects , Mucocutaneous Lymph Node Syndrome/drug therapy , Prednisolone/adverse effects , Adrenal Glands/metabolism , Adrenal Glands/physiopathology , Adrenal Insufficiency/epidemiology , Adrenal Insufficiency/prevention & control , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/metabolism , Anti-Inflammatory Agents/therapeutic use , Child , Child, Preschool , Combined Modality Therapy/adverse effects , Female , Glucocorticoids/therapeutic use , Humans , Hydrocortisone/adverse effects , Hydrocortisone/antagonists & inhibitors , Hydrocortisone/blood , Hydrocortisone/metabolism , Hydrocortisone/therapeutic use , Immunoglobulins, Intravenous/therapeutic use , Incidence , Infant , Japan/epidemiology , Male , Mucocutaneous Lymph Node Syndrome/blood , Mucocutaneous Lymph Node Syndrome/physiopathology , Mucocutaneous Lymph Node Syndrome/therapy , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiopathology , Prednisolone/therapeutic use , Prospective Studies , Risk
3.
Am J Physiol Endocrinol Metab ; 307(9): E754-63, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25184992

ABSTRACT

Elevated levels of adrenocorticotrophic hormone (ACTH) mobilize granulocytes from bone marrow into the blood, although these neutrophils are refractory to a full migratory response into inflamed tissues. Here, we investigated the dependence of glucocorticoid receptor activation and glucocorticoid-regulated protein annexin A1 (ANXA1) on ACTH-induced neutrophilia and the phenotype of blood neutrophil after ACTH injection, focusing on adhesion molecule expressions and locomotion properties. ACTH injection (5 µg ip, 4 h) induced neutrophilia in wild-type (WT) mice and did not alter the elevated numbers of neutrophils in RU-38486 (RU)-pretreated or ANXA1(-/-) mice injected with ACTH. Neutrophils from WT ACTH-treated mice presented higher expression of Ly6G⁺ANXA1(high), CD18(high), CD62L(high), CD49(high), CXCR4(high), and formyl-peptide receptor 1 (FPR1(low)) than those observed in RU-pretreated or ANXA1(-/-) mice. The membrane phenotype of neutrophils collected from WT ACTH-treated mice was paralleled by elevated fractions of rolling and adherent leukocytes to the cremaster postcapillary venules together with impaired neutrophil migration into inflamed air pouches in vivo and in vitro reduced formyl-methionyl-leucyl-phenylalanine (fMLP) or stromal-derived factor-1 (SDF-1α)-induced chemotaxis. In an 18-h senescence protocol, neutrophils from WT ACTH-treated mice had a higher proportion of ANXAV(low)/CXCR4(low), and they were less phagocytosed by peritoneal macrophages. We conclude that alterations on HPA axis affect the pattern of membrane receptors in circulating neutrophils, which may lead to different neutrophil phenotypes in the blood. Moreover, ACTH actions render circulating neutrophils to a phenotype with early reactivity, such as in vivo leukocyte-endothelial interactions, but with impaired locomotion and clearance.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Annexin A1/metabolism , Leukopoiesis , Neutrophils/metabolism , Receptors, Corticotropin/metabolism , Stress, Physiological , Stress, Psychological/immunology , Adrenocorticotropic Hormone/administration & dosage , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/blood , Animals , Annexin A1/blood , Annexin A1/genetics , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Corticosterone/blood , Corticosterone/metabolism , Hormone Antagonists/pharmacology , Leukopoiesis/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred BALB C , Mice, Knockout , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology , Phagocytosis/drug effects , Receptors, Corticotropin/agonists , Receptors, Corticotropin/antagonists & inhibitors , Receptors, Corticotropin/blood , Stress, Physiological/drug effects , Stress, Psychological/blood , Stress, Psychological/metabolism , Stress, Psychological/pathology , Surface Properties/drug effects , Up-Regulation/drug effects
4.
Endocr Pract ; 20(1): 84-93, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24126229

ABSTRACT

OBJECTIVE: To review and evaluate medical therapies for Cushing's disease (CD), with an emphasis on recent clinical trial experience with pasireotide and mifepristone, and to discuss the therapeutic potential and appropriate selection of these compounds in this patient population. METHODS: Recently published Phase III trial data for each compound are reviewed and assessed, and relative benefits and risks are examined and compared where possible. RESULTS: Mifepristone and pasireotide are both potentially beneficial for CD patients but have greatly dissimilar mechanisms of action and adverse event (AE) profiles. Pasireotide acts at the level of the pituitary adenoma, reducing cortisol levels through inhibition of adrenocorticotropic hormone (ACTH) release. However, pasireotide reduces insulin secretion and incretin hormone response and is associated with significant risk for new or worsening hyperglycemia. Mifepristone ameliorates the signs and symptoms of hypercortisolemia via glucocorticoid receptor (GR2) blockade, but this approach raises serum cortisol levels and increases risk for adrenal insufficiency (AI), hypokalemia, and endometrial thickening. While response to pasireotide can be monitored via measurements of serum, urine, or late-night salivary cortisol, evaluation of response to mifepristone is solely based on changes in clinical parameters (e.g., hyperglycemia, hypertension, body weight/composition). CONCLUSION: Management of persistent CD is challenging, and the decision to initiate medical treatment hinges on many factors. Pasireotide may be a more attractive option for most patients due to its action at the underlying tumor and the ability to monitor biochemical responses. However, mifepristone may be more appropriate when it is necessary to avoid or minimize risk for hyperglycemia-related complications.


Subject(s)
Mifepristone/therapeutic use , Pituitary ACTH Hypersecretion/drug therapy , Somatostatin/analogs & derivatives , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/metabolism , Humans , Mifepristone/pharmacology , Receptors, Glucocorticoid/antagonists & inhibitors , Somatostatin/pharmacology , Somatostatin/therapeutic use
5.
Neuroendocrinology ; 98(3): 233-41, 2013.
Article in English | MEDLINE | ID: mdl-24080898

ABSTRACT

Clusterin is a sulfated glycoprotein abundantly expressed in the pituitary gland and hypothalamus of mammals. However, its physiological role in neuroendocrine function is largely unknown. In the present study, we investigated the effects of intracerebroventricular (ICV) administration of clusterin on plasma pituitary hormone levels in normal rats. Single ICV injection of clusterin provoked neurohormonal changes seen under acute stress condition: increased plasma adrenocorticotropic hormone (ACTH), corticosterone, GH and prolactin levels and decreased LH and FSH levels. Consistently, hypothalamic and pituitary clusterin expression levels were upregulated following a restraint stress, suggesting an involvement of endogenous clusterin in stress-induced neurohormonal changes. In the pituitary intermediate lobe, clusterin was coexpressed with proopiomelanocortin (POMC), a precursor of ACTH. Treatment of clusterin in POMC expressing AtT-20 pituitary cells increased basal and corticotropin-releasing hormone (CRH)-stimulated POMC promoter activities and intracellular cAMP levels. Furthermore, clusterin treatment triggered ACTH secretion from AtT-20 cells in a CRH-dependent manner, indicating that increased clusterin under stressful conditions may augment CRH-stimulated ACTH production and release. In summary, hypothalamic and pituitary clusterin may function as a modulator of neurohormonal responses under stressful conditions.


Subject(s)
Clusterin/physiology , Hypothalamus/metabolism , Neurotransmitter Agents/biosynthesis , Pituitary Gland/metabolism , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/biosynthesis , Adrenocorticotropic Hormone/metabolism , Animals , Clusterin/administration & dosage , Clusterin/blood , Hypothalamus/drug effects , Injections, Intraventricular , Male , Neurotransmitter Agents/antagonists & inhibitors , Neurotransmitter Agents/metabolism , Pituitary Gland/drug effects , Pro-Opiomelanocortin/antagonists & inhibitors , Pro-Opiomelanocortin/biosynthesis , Pro-Opiomelanocortin/metabolism , Rats , Rats, Sprague-Dawley , Stress, Psychological/blood , Stress, Psychological/prevention & control , Stress, Psychological/psychology , Up-Regulation/physiology
6.
Neurosci Lett ; 746: 135618, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33429002

ABSTRACT

Stress is a common seizure trigger in persons with epilepsy. The body's physiological response to stress is mediated by the hypothalamic-pituitary-adrenal (HPA) axis and involves a hormonal cascade that includes corticotropin releasing hormone (CRH), adrenocorticotropin releasing hormone (ACTH) and the release of cortisol (in humans and primates) or corticosterone (in rodents). The prolonged exposure to stress hormones may not only exacerbate pre-existing medical conditions including epilepsy, but may also increase the predisposition to psychiatric comorbidities. Hyperactivity of the HPA axis negatively impacts the structure and function of the temporal lobe of the brain, a region that is heavily involved in epilepsy and mood disorders like anxiety and depression. Seizures themselves damage temporal lobe structures, further disinhibiting the HPA axis, setting off a vicious cycle of neuronal damage and increasing susceptibility for subsequent seizures and psychiatric comorbidity. Treatments targeting the HPA axis may be beneficial both for epilepsy and for associated stress-related comorbidities such as anxiety or depression. This paper will highlight the evidence demonstrating dysfunction in the HPA axis associated with epilepsy which may contribute to the comorbidity of psychiatric disorders and epilepsy, and propose treatment strategies that may dually improve seizure control as well as alleviate stress related psychiatric comorbidities.


Subject(s)
Epilepsy/metabolism , Hypothalamo-Hypophyseal System/metabolism , Mental Disorders/metabolism , Pituitary-Adrenal System/metabolism , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/metabolism , Animals , Anticonvulsants/administration & dosage , Corticosterone/antagonists & inhibitors , Corticosterone/metabolism , Desoxycorticosterone/antagonists & inhibitors , Desoxycorticosterone/metabolism , Epilepsy/drug therapy , Epilepsy/epidemiology , Humans , Hypothalamo-Hypophyseal System/drug effects , Mental Disorders/drug therapy , Mental Disorders/epidemiology , Pituitary-Adrenal System/drug effects , Treatment Outcome
7.
J Neurosci ; 29(5): 1404-13, 2009 Feb 04.
Article in English | MEDLINE | ID: mdl-19193887

ABSTRACT

Inflammation-induced activation of the hypothalamic-pituitary-adrenal (HPA) axis has been suggested to depend on prostaglandins, but the prostaglandin species and the prostaglandin-synthesizing enzymes that are responsible have not been fully identified. Here, we examined HPA axis activation in mice after genetic deletion or pharmacological inhibition of prostaglandin E(2)-synthesizing enzymes, including cyclooxygenase-1 (Cox-1), Cox-2, and microsomal prostaglandin E synthase-1 (mPGES-1). After immune challenge by intraperitoneal injection of lipopolysaccharide, the rapid stress hormone responses were intact after Cox-2 inhibition and unaffected by mPGES-1 deletion, whereas unselective Cox inhibition blunted these responses, implying the involvement of Cox-1. However, mPGES-1-deficient mice showed attenuated transcriptional activation of corticotropin-releasing hormone (CRH) that was followed by attenuated plasma concentrations of adrenocorticotropic hormone and corticosterone. Cox-2 inhibition similarly blunted the delayed corticosterone response and further attenuated corticosterone release in mPGES-1 knock-out mice. The expression of the c-fos gene, an index of synaptic activation, was maintained in the paraventricular hypothalamic nucleus and its brainstem afferents both after unselective and Cox-2 selective inhibition as well as in Cox-1, Cox-2, and mPGES-1 knock-out mice. These findings point to a mechanism by which (1) neuronal afferent signaling via brainstem autonomic relay nuclei and downstream Cox-1-dependent prostaglandin release and (2) humoral, CRH transcription-dependent signaling through induced Cox-2 and mPGES-1 elicited PGE(2) synthesis, shown to occur in brain vascular cells, play distinct, but temporally supplementary roles for the stress hormone response to inflammation.


Subject(s)
Dinoprostone/biosynthesis , Hypothalamo-Hypophyseal System/enzymology , Hypothalamo-Hypophyseal System/immunology , Pituitary-Adrenal System/enzymology , Pituitary-Adrenal System/immunology , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/metabolism , Animals , Corticosterone/antagonists & inhibitors , Corticosterone/metabolism , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/administration & dosage , Hypothalamo-Hypophyseal System/drug effects , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/biosynthesis , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Pituitary-Adrenal System/drug effects , Prostaglandin-E Synthases
8.
Am J Physiol Endocrinol Metab ; 299(4): E567-75, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20663987

ABSTRACT

We have previously shown that urocortin 2 (Ucn 2), a member of the corticotropin-releasing factor (CRF) peptide family that binds to CRF type 2 receptor, is expressed in proopiomelanocortin (POMC) cells of rat pituitary and that its secretion and expression are increased by CRF in both the anterior and intermediate lobes and suppressed by glucocorticoids in the anterior lobe. We have also shown that Ucn 2 secreted by POMC cells acts on gonadotrophs expressing CRF type 2 receptors and inhibits the expression and secretion of gonadotropins. In the present study, we examined whether pituitary Ucn 2 is involved in stress-induced inhibition of gonadotropin secretion. A 90-min period of immobilization stress increased POMC mRNA expression without influencing Ucn 2 mRNA expression and suppressed luteinizing hormone (LH) ß-subunit mRNA expression in the anterior lobe and plasma LH levels, while it increased both POMC and Ucn 2 mRNA expression in the intermediate lobe of the pituitary. Pretreatment with anti-CRF IgG blocked immobilization-induced increases in plasma ACTH and corticosterone and in POMC mRNA expression in both pituitary lobes and Ucn 2 mRNA expression in the intermediate pituitary. It also blocked immobilization-induced suppression of plasma LH and LH ß-subunit mRNA expression. Pretreatment with anti-Ucn 2 IgG blocked immobilization-induced suppression of plasma LH and LH ß-subunit expression without affecting immobilization-induced ACTH and corticosterone release and POMC or Ucn 2 mRNA expression. These results suggest that CRF suppresses the secretion and expression of LH probably through pituitary Ucn 2 in stress.


Subject(s)
Corticotropin-Releasing Hormone/physiology , Luteinizing Hormone, beta Subunit/physiology , Pituitary Gland/physiopathology , Pro-Opiomelanocortin/physiology , Stress, Physiological/physiology , Urocortins/physiology , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/blood , Animals , Corticosterone/antagonists & inhibitors , Corticosterone/blood , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/immunology , Immunization/methods , Immunoglobulin G/pharmacology , Luteinizing Hormone, beta Subunit/antagonists & inhibitors , Luteinizing Hormone, beta Subunit/genetics , Luteinizing Hormone, beta Subunit/metabolism , Male , Pituitary Gland/metabolism , Pro-Opiomelanocortin/genetics , RNA/chemistry , RNA/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Urocortins/genetics
9.
Drugs ; 79(9): 935-956, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31098899

ABSTRACT

Endogenous Cushing's syndrome is a chronic disease associated with increased morbidity and mortality if not appropriately treated. Recurrence and/or persistence of hypercortisolemia after surgical treatment, especially for Cushing's disease, are high, and long-term medical treatment is used to decrease cortisol levels and risk of metabolic comorbidities. Medical treatment is also often required while waiting for radiation effects to take place. In some cases, severe or life-threatening hypercortisolism must be urgently and medically treated, via intravenous medications or with combination therapy, before patients can undergo surgery. In the last decade, medical treatment has progressed from a few steroidogenesis inhibitors to three novel drug groups: new inhibitors for steroidogenic enzymes with possibly fewer side effects, pituitary-directed drugs that aim to inhibit the pathophysiological pathways of Cushing's disease, and glucocorticoid receptor antagonists that block cortisol's action. Understanding the pathophysiology of Cushing's syndrome has also led to the identification of potential targets that may decrease adrenocorticotrophic hormone and/or cortisol excess, and/or decrease tumor cell proliferation, and induce senescence or apoptosis. We provide here a review of current and near-future medical options to treat Cushing's syndrome, and discuss updates on clinical trials and the efficacy and safety of novel or in-development drugs, as well as future potential targets.


Subject(s)
Adrenocorticotropic Hormone/antagonists & inhibitors , Cushing Syndrome/drug therapy , Enzyme Inhibitors/therapeutic use , Hydrocortisone/biosynthesis , Receptors, Glucocorticoid/antagonists & inhibitors , Clinical Trials as Topic , Cushing Syndrome/metabolism , Drug Therapy, Combination , Endocrinology/methods , Endocrinology/trends , Enzyme Inhibitors/pharmacology , Humans , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Receptors, Glucocorticoid/metabolism , Treatment Outcome
10.
Cell Biochem Funct ; 26(5): 578-81, 2008.
Article in English | MEDLINE | ID: mdl-18508386

ABSTRACT

Tetrahymena populations were treated with 10(-15) g ml(-1) or 10(-6) g ml(-1) concanavalin-A (Con-A) in tryptone-yeast medium for 1 h. Rat peritoneal immune cells (mast cells, lymphocytes, monocyte-granulocyte group) were also treated with 10(-6) g ml(-1) Con-A, for 1 h. The cells' hormone (ACTH, histamine, serotonin, endorphin, triiodothyronine (T(3))) content was measured by using immunocytochemistry and flow cytometry. The extremely low dose of Con-A universally and significantly elevated the hormone contents, while the result of higher dose was uncertain. In the immune cells, Con-A significantly decreased the ACTH level in each cell type and histamine level in mast cells. The results demonstrate the very high sensitivity of Tetrahymena receptors for a non-hormone (lectin) molecule, which can bind to the insulin receptors and mimics the effect of insulin. The results also show that Tetrahymena receptors are more sensitive to lower concentrations of molecules than to higher ones. The universal hormone-production stimulating effect of Con-A-which is observed in Tetrahymena-is specified in rat.


Subject(s)
Concanavalin A/pharmacology , Hormones/biosynthesis , Immune System/cytology , Immune System/drug effects , Mitogens/pharmacology , Tetrahymena pyriformis/drug effects , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/biosynthesis , Animals , Granulocytes/drug effects , Granulocytes/metabolism , Histamine/biosynthesis , Immune System/metabolism , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mast Cells/drug effects , Mast Cells/metabolism , Monocytes/drug effects , Monocytes/metabolism , Rats , Rats, Wistar , Tetrahymena pyriformis/metabolism
11.
J Clin Invest ; 52(9): 2272-7, 1973 Sep.
Article in English | MEDLINE | ID: mdl-4353776

ABSTRACT

Plasma aldosterone, cortisol, and renin activity were measured in nine recumbent patients with hyperaldosteronism, including seven with adenomas, one with idiopathic hyperplasia, and one with glucocorticoid suppressible hyperplasia. All had peak values of plasma aldosterone concentration from 3 a.m. to noon and lowest values at 6 p.m. or midnight. This rhythm was similar to the circadian pattern of plasma cortisol in the same patients. When these data were normalized to eliminate the wide variation in ranges of plasma aldosterone and cortisol between individuals, there was an excellent correlation (r = + 0.87, P < 0.005) between the two hormones. In contrast, plasma aldosterone concentrations did not correlate with plasma renin activity before or after normalization of data. Short term suppression of ACTH by administration of dexamethasone eliminated the circadian variation of plasma aldosterone in both patients with hyperplasia and in four of five patients with adenomas, while it markedly altered the rhythm in the fifth. Similar doses of dexamethasone were administered to four normal subjects and did not flatten the circadian rhythm of plasma aldosterone. These data suggest that patients with primary aldosteronism have a circadian rhythm of plasma aldosterone mediated by changes in ACTH.


Subject(s)
Aldosterone/blood , Circadian Rhythm , Hyperaldosteronism/physiopathology , Adenoma/complications , Adrenal Gland Neoplasms/complications , Adrenocortical Hyperfunction/complications , Adrenocorticotropic Hormone/antagonists & inhibitors , Dexamethasone/pharmacology , Humans , Hydrocortisone/blood , Hyperaldosteronism/etiology , Potassium/blood , Renin/blood
12.
J Clin Invest ; 47(8): 1742-52, 1968 Aug.
Article in English | MEDLINE | ID: mdl-4299011

ABSTRACT

Patients with the "non-salt-losing" form of the adrenogenital syndrome were studied before and after suppression of adrenal cortical activity with carbohydrate-active steroids. The response of aldosterone secretion to sodium deprivation was measured; in some patients response to adrenocorticotropic hormone (ACTH) was measured as well. The aldosterone secretion was normal and responded normally to sodium deprivation in all patients studied during suppression with carbohydrate-active steroids. This finding suggests that 21-hydroxylation of progesterone is normal in this syndrome. The sole abnormality in the production of aldosterone in these patients was found to be excessive secretion of aldosterone while they were not receiving suppressive doses of carbohydrate-active steroids. This finding strongly supports the view that the biogenetic pathways through which aldosterone is produced from progesterone are intact in this syndrome. No patient showed hypertension or hypokalemic alkalosis despite very high aldosterone secretion rates. This observation suggests that the hyper-aldosteronism is secondary to a tendency to sodium loss in the patient whose ACTH production is not suppressed. These studies provide additional evidence in support of the hypothesis that the salt-losing and "non-salt-losing" forms of adrenogenital syndrome are genetically and biochemically distinct.


Subject(s)
Adrenal Cortex Hormones/biosynthesis , Adrenal Hyperplasia, Congenital/congenital , Hyperaldosteronism/etiology , Sodium/metabolism , Adolescent , Adrenal Cortex Hormones/antagonists & inhibitors , Adrenal Hyperplasia, Congenital/drug therapy , Adrenal Hyperplasia, Congenital/metabolism , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/metabolism , Adult , Aldosterone/biosynthesis , Child , Child, Preschool , Female , Glucocorticoids/therapeutic use , Humans , Hyperplasia , Hyponatremia/metabolism , Male , Progesterone/biosynthesis , Sodium Chloride/urine
13.
J Clin Invest ; 108(8): 1159-66, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11602623

ABSTRACT

Stimulation of the hypothalamic-pituitary-adrenal (HPA) axis by proinflammatory cytokines results in increased release of glucocorticoid that restrains further development of the inflammatory process. IL-6 has been suggested to stimulate the HPA axis during immune activation independent of the input of hypothalamic corticotropin-releasing hormone (CRH). We used the corticotropin-releasing hormone-deficient (Crh(-/-)) mouse to elucidate the effect of CRH deficiency on IL-6 expression and IL-6-induced HPA axis activation during turpentine-induced inflammation. We demonstrate that during inflammation CRH is required for a normal adrenocorticotropin hormone (ACTH) increase but not for adrenal corticosterone rise. The paradoxical increase of plasma IL-6 associated with CRH deficiency suggests that IL-6 release during inflammation is CRH-dependent. We also demonstrate that adrenal IL-6 expression is CRH-dependent, as its basal and inflammation-induced expression is blocked by CRH deficiency. Our findings suggest that during inflammation, IL-6 most likely compensates for the effects of CRH deficiency on food intake. Finally, we confirm that the HPA axis response is defective in Crh(-/-)/IL-6(-/-) mice. These findings, along with the regulation of IL-6 by CRH, support the importance of the interaction between the immune system and the HPA axis in the pathophysiology of inflammatory diseases.


Subject(s)
Corticotropin-Releasing Hormone/physiology , Inflammation/etiology , Interleukin-6/genetics , Adrenalectomy , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/blood , Animals , Corticosterone/blood , Corticotropin-Releasing Hormone/deficiency , Corticotropin-Releasing Hormone/genetics , Gene Expression Regulation , Hypothalamo-Hypophyseal System/physiology , Inflammation/immunology , Inflammation/pathology , Inflammation/physiopathology , Interleukin-6/blood , Irritants/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Pituitary-Adrenal System/physiology , Turpentine/toxicity
14.
J Clin Invest ; 52(8): 2007-15, 1973 Aug.
Article in English | MEDLINE | ID: mdl-4352578

ABSTRACT

Adrenal ornithine decarboxylase activity was stimulated in a dose-related manner after administration of ACTH or dibutyryl ((6)N-2'-O-dibutyryl) cyclic AMP to hypophysectomized rats. Little effect was observed for 2 h, but striking increases in enzyme activity were observed 4 h after administration of these substances. Effects of ACTH and dibutyryl cyclic AMP were not secondary to stimulation of steroidogenesis, since hydrocortisone had no effect on adrenal ornithine decarboxylase although it did stimulate activity of the enzyme in the liver and kidney.ACTH, given subcutaneously to hypophysectomized rats, induced striking increases in adrenal cyclic AMP levels within 15-30 min with a fall towards the base line in 1 h. Increases in ornithine decarboxylase activity lag several hours after this endogenous cyclic AMP peak, in contrast to the stimulatin of steroidogenesis by the nucleotide that requires only 2-3 min. After graded doses of ACTH, increases in adrenal cyclic AMP levels at 30 min were paralleled by proportional increases in adrenal ornithine decarboxylase activity 4 h after hormone treatment. Whereas maximal levels of adrenal steroidogenesis have been observed at tissue cyclic AMP levels of 6 nmol/g. ACTH is capable of inducing increases in nucleotide levels up to 200 nmol/g or more. These high tissue levels of cyclic AMP, although unneccessary for maximal steroidogenesis, appear to stimulate adrenal ornithine decarboxylase activity. Several results in addition to the time lag in the stimulation of ornithine decarboxylase activity suggest a mechanism involving accumulation of the enzyme or some factor needed for its activity rather than direct activation of the enzyme by cyclic AMP. Thus, the addition of cyclic AMP directly to the ornithine decarboxylase assay mixture in vitro was without stimulatory effect. In addition, actinomycin D or cycloheximide in doses sufficient to block adrenal RNA and protein synthesis, respectively inhibited the stimulation of ornithine decarboxylase activity by ACTH in vivo. An adrenocortical cancer was found to maintain ornithine decarboxylase activity at very high levels, but did so at much lower cyclic AMP levels than those of ACTH-stimulated adrenals. It is concluded that ACTH stimulates adrenal ornithine decarboxylase activity and that this effect may be mediated by cyclic AMP. However, cyclic AMP be mediated by appear to be a determinant of the high level of enzyme activity found in adrenocortical cancer.


Subject(s)
Adrenal Glands/enzymology , Adrenocorticotropic Hormone/pharmacology , Carboxy-Lyases/metabolism , Cyclic AMP/pharmacology , Adrenal Gland Neoplasms/metabolism , Adrenal Glands/analysis , Adrenocorticotropic Hormone/antagonists & inhibitors , Angiotensin II/pharmacology , Animals , Carboxy-Lyases/analysis , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Dithiothreitol/pharmacology , Dose-Response Relationship, Drug , Hydrocortisone/pharmacology , Hypophysectomy , Kidney/enzymology , Liver/enzymology , Ornithine , Pituitary Gland/physiology , Rats , Stimulation, Chemical , Time Factors
15.
Endocrinology ; 158(1): 1-8, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27906551

ABSTRACT

Adrenocorticotropic hormone (ACTH) is the primary regulator of adrenal glucocorticoid production. Elevated levels of ACTH play a critical role in disease progression in several indications, including congenital adrenal hyperplasia and Cushing disease. We have generated a specific, high-affinity, neutralizing monoclonal antibody (ALD1613) to ACTH. In vitro, ALD1613 neutralizes ACTH-induced signaling via all 5 melanocortin receptors and inhibited ACTH-induced cyclic adenosine monophosphate accumulation in a mouse adrenal cell line (Y1). ALD1613 administration to wild-type rats significantly reduced plasma corticosterone levels in a dose-dependent manner. In rodent models with either chronic infusion of ACTH or acute restraint stress-induced ACTH, corticosterone levels were significantly reduced by ALD1613. Administration of ALD1613 to nonhuman primates on days 1 and 7 stably reduced plasma cortisol levels >50% for 57 days. ALD1613 demonstrates the potential of a monoclonal antibody to be an effective therapeutic for conditions with elevated ACTH levels.


Subject(s)
Adrenocorticotropic Hormone/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Hydrocortisone/blood , Adrenal Hyperplasia, Congenital/drug therapy , Adrenocorticotropic Hormone/metabolism , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , CHO Cells , Corticosterone/blood , Cricetinae , Cricetulus , Drug Evaluation, Preclinical , Humans , Macaca fascicularis , Male , Pituitary ACTH Hypersecretion/drug therapy , Rabbits , Rats , Rats, Inbred Lew , Receptor, Melanocortin, Type 2/metabolism , Stress, Psychological/blood
16.
Anat Embryol (Berl) ; 211(1): 61-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16374610

ABSTRACT

Elevated glucocorticoid level in the gravid female circulation affects number of endocrine functions in fetuses and offspring. In this research female rats were injected with dexamethasone (Dx) in three consecutive daily doses of 1.0, 0.5, 0.5 mg/kg body weight, starting from day 16 of pregnancy. The influence of this treatment on the pituitary adrenocorticotrophic (ACTH) cells and adrenal glands of 19-day-old fetuses was examined immunocytochemically and by morphometric analysis. Moreover, the proliferative activity of adrenocortical cells was estimated after application of the mitotic inhibitor Oncovine. Administration of Dx to pregnant rats induced a decline of fetal ACTH cell immunopositivity and significant decreases of ACTH cell volume (23%, p < 0.05), volume density (41%, p < 0.05), and its number per unit area (17%, p < 0.05) in comparison to the control 19-day-old fetuses. Reduced proliferative activity of adrenocortical cells (31%; p < 0.05) in zona glomerulosa, as well as the volume of this zone were detected. The volume and number of fetal adrenocortical cells in the inner zone and chromoblasts were not significantly reduced after Dx treatment of pregnant rats. These results show that maternal Dx administration in the period when the fetal hypothalamo-pituitary-adrenal (PA) axis begins its function inhibited the PA axis. Reduced ACTH cell function and mitotic activity led to suppression of adrenocortical cell multiplication in zona glomerulosa, the region of the adrenal cortex where most proliferating cells were found in control 19-day-old fetuses. Thus, increased glucocorticoid levels during late pregnancy caused developmental modifications involving the fetal PA axis, which could be the basis of the altered endocrine responsiveness in adult life.


Subject(s)
Dexamethasone/pharmacology , Fetus/drug effects , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/embryology , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/metabolism , Animals , Cell Count , Dexamethasone/administration & dosage , Female , Fetus/cytology , Fetus/physiology , Growth Inhibitors/administration & dosage , Growth Inhibitors/pharmacology , Injections, Subcutaneous , Male , Pituitary-Adrenal System/cytology , Pituitary-Adrenal System/physiology , Pregnancy , Rats , Rats, Wistar
17.
Life Sci ; 151: 277-280, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26979774

ABSTRACT

AIMS: Retinoic acid has recently yielded promising results in the treatment of Cushing's disease, i.e., excess cortisol secretion due to a pituitary corticotropin (ACTH)-secreting adenoma. In addition to its effect on the tumoral corticotrope cell, clinical results suggest an additional adrenal site of action. Aim of this study was to evaluate whether retinoic acid modulates cortisol synthesis and secretion by human adrenals in vitro. MAIN METHODS: Primary cultures from 10 human adrenals specimens were incubated with 10nM, 100nM and 1µM retinoic acid with and without 10nM ACTH for 24h. Cortisol levels were measured by radioimmunoassay and CYP11A1, STAR and MC2R gene expression analyzed by real-time PCR. KEY FINDINGS: Retinoic acid increased cortisol secretion (149.5±33.01%, 151.3±49.45% and 129.3±8.32% control secretion for 10nM, 100nM and 1µM respectively, p<0.05) and potentiated STAR expression (1.51±0.22, 1.56±0.15 and 1.59±0.14 fold change over baseline, for 10nM, 100nM and 1µM respectively, p<0.05). Concurrently, retinoic acid markedly blunted constitutional and ACTH-induced MC2R expression (0.66±0.11, 0.62±0.08 and 0.53±0.07 fold change over baseline, for 10nM, 100nM and 1µM respectively, p<0.05; 0.71±0.10, 0.51±0.07 and 0.51±0.08 fold change over ACTH alone, for 10nM, 100nM and 1µM respectively, p<0.05). No effect on CYP11A1 was observed. SIGNIFICANCE: Retinoic acid stimulates cortisol synthesis and secretion in human adrenals and at the same time markedly blunts ACTH receptor transcription. These results reveal a novel, adrenal effect of retinoic acid which may contribute to its efficacy in patients with Cushing's disease.


Subject(s)
Adrenal Glands/drug effects , Adrenal Glands/metabolism , Hydrocortisone/biosynthesis , Hydrocortisone/metabolism , Tretinoin/pharmacology , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/pharmacology , Cholesterol Side-Chain Cleavage Enzyme/biosynthesis , Dose-Response Relationship, Drug , Gene Expression/drug effects , Humans , Phosphoproteins/biosynthesis , Primary Cell Culture , Receptor, Melanocortin, Type 1/biosynthesis
18.
Sci Transl Med ; 8(352): 352ra109, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27535620

ABSTRACT

The aim of treatment in congenital adrenal hyperplasia is to suppress excess adrenal androgens while achieving physiological glucocorticoid replacement. However, current glucocorticoid replacement regimes are inadequate because doses sufficient to suppress excess androgens almost invariably induce adverse metabolic effects. Although both cortisol and corticosterone are glucocorticoids that circulate in human plasma, any physiological role for corticosterone has been neglected. In the brain, the adenosine 5'-triphosphate-binding cassette transporter ABCB1 exports cortisol but not corticosterone. Conversely, ABCC1 exports corticosterone but not cortisol. We show that ABCC1, but not ABCB1, is expressed in human adipose and that ABCC1 inhibition increases intracellular corticosterone, but not cortisol, and induces glucocorticoid-responsive gene transcription in human adipocytes. Both C57Bl/6 mice treated with the ABCC1 inhibitor probenecid and FVB mice with deletion of Abcc1 accumulated more corticosterone than cortisol in adipose after adrenalectomy and corticosteroid infusion. This accumulation was sufficient to increase glucocorticoid-responsive adipose transcript expression. In human adipose tissue, tissue corticosterone concentrations were consistently low, and ABCC1 mRNA was up-regulated in obesity. To test the hypothesis that corticosterone effectively suppresses adrenocorticotropic hormone (ACTH) without the metabolic adverse effects of cortisol, we infused cortisol or corticosterone in patients with Addison's disease. ACTH suppression was similar, but subcutaneous adipose transcripts of glucocorticoid-responsive genes were higher after infusion with cortisol rather than with corticosterone. These data indicate that corticosterone may be a metabolically favorable alternative to cortisol for glucocorticoid replacement therapy when ACTH suppression is desirable, as in congenital adrenal hyperplasia, and justify development of a pharmaceutical preparation.


Subject(s)
Corticosterone/pharmacology , Hydrocortisone/pharmacology , Multidrug Resistance-Associated Proteins/metabolism , Addison Disease/drug therapy , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adrenal Hyperplasia, Congenital/drug therapy , Adrenal Hyperplasia, Congenital/genetics , Adrenal Hyperplasia, Congenital/metabolism , Adrenocorticotropic Hormone/antagonists & inhibitors , Animals , Biological Transport, Active , Brain/drug effects , Brain/metabolism , Cells, Cultured , Corticosterone/metabolism , Glucocorticoids/metabolism , Humans , Hydrocortisone/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multidrug Resistance-Associated Proteins/deficiency , Multidrug Resistance-Associated Proteins/genetics , Obesity/metabolism , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Translational Research, Biomedical
19.
Biochim Biophys Acta ; 542(2): 330-9, 1978 Aug 17.
Article in English | MEDLINE | ID: mdl-210839

ABSTRACT

Addition of the ionophore A23187 to Y-1 mouse adrenal tumor cells in monolayer culture inhibits steroidogenesis and the steroidogenic response to corticotropin (50% inhibition at 1 . 10(-7)M). Inhibition is rapid in onset and is not overcome by addition of external Ca2+. The ionophore also inhibits stimulation of steroid synthesis by cyclic AMP. A23187 inhibits incorporation of the amino acid lysine into protein by Y-1 cells and the dose dependence of this inhibition closely resembles that of the inhibition of the steroidogenic response to corticotropin. Addition of A23187 to a subcellular system for protein synthesis prepared from Y-1 cells, inhibits incorporation of the amino acid phenylalanine into protein and this effect is not overcome by high concentrations of Ca2+. The inhibitory effect of A23187 on the response to corticotropin, like that response itself, takes place at some part of steroid synthesis after entry of cholesterol into the cells and before the side-chain cleavage of cholesterol. These studies confirm the importance of protein synthesis in the response to corticotropin and demonstrate that the effect of protein synthesized under the influence of corticotropin is exerted at some point in the events which bring substrate (cholesterol) to the mitochondrial side-chain cleavage enzyme system. It is also shown that A23187 inhibits protein synthesis, and hence the response to corticotropin, by a mechanism which is independent of the concentration of available Ca2+.


Subject(s)
20-alpha-Dihydroprogesterone/biosynthesis , Adrenal Cortex/metabolism , Adrenocorticotropic Hormone/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Calcimycin/pharmacology , Progesterone/analogs & derivatives , Protein Biosynthesis , Aminoglutethimide/pharmacology , Bucladesine/antagonists & inhibitors , Calcium/metabolism , Cations, Divalent/pharmacology , Cells, Cultured , Cholesterol/metabolism , RNA/biosynthesis
20.
Biochim Biophys Acta ; 1310(3): 260-8, 1996 Feb 29.
Article in English | MEDLINE | ID: mdl-8599603

ABSTRACT

We have investigated the effect of the proteinase inhibitors 1,10-phenantroline (OP) and phenylmethylsulfonyl fluoride (PMSF) on steroidogenesis in rat adrenal cortex. Both PMSF and OP inhibited adrenocorticotropin (ACTH)- and 8-Br cAMP-induced stimulation of corticosterone synthesis. On the contrary, arachidonic acid-induced stimulation of corticosterone synthesis was only slightly inhibited by PMSF and unchanged by OP. Intra- and extracellular cAMP levels were determined by radioimmunoassay. While PMSF did not affect neither the intra- nor the extracellular cAMP levels, OP decreased the intra- and extracellular levels of unstimulated as well as ACTH-stimulated cells. The site of action of the proteinase inhibitors was also studied by recombination of mitochondria with the different subcellular fractions in vitro. Addition of PMSF abolished the stimulation achieved by in vitro activation of cytosol by cAMP and PKA. On the other hand, OP completely inhibited the activation of mitochondria. Our results provide evidence for the involvement of proteinases in ACTH-induced stimulation of steroidogenesis in adrenal cortex both prior to the release of arachidonic acid and at the level of cholesterol transport from the outer to the inner mitochondrial membrane.


Subject(s)
Corticosterone/biosynthesis , Cyclic AMP/metabolism , Endopeptidases/metabolism , Protease Inhibitors/pharmacology , Zona Fasciculata/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/pharmacology , Animals , Arachidonic Acid/pharmacology , Hydroxycholesterols/metabolism , In Vitro Techniques , Kinetics , Male , Phenanthrolines/pharmacology , Phenylmethylsulfonyl Fluoride/pharmacology , Pregnenolone/metabolism , Progesterone/metabolism , Rats , Rats, Wistar , Zona Fasciculata/cytology , Zona Fasciculata/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL