Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Exp Mol Pathol ; 84(2): 178-88, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18262521

ABSTRACT

The development of an effective pharmacological countermeasure is needed to reduce the morbidity and mortality in military and civilian populations associated with possible exposure to ionizing radiation. Previous studies in mice have shown that a single subcutaneous (sc) injection of the natural steroid androst-5-ene-3beta,17beta-diol (5-androstenediol, 5-AED), 24-48 h prior to a lethal dose of whole-body (60)Co gamma radiation, stimulated hematopoiesis and enhanced survival. These effects are consistent with our previous observation of 5-AED-induced elevations in circulating G-CSF in normal and irradiated mice. The purpose of this study was to obtain data on the pharmacokinetics of 5-AED after sc and buccal administration to mice, and to determine whether cytokine genes are induced by sc 5-AED in hematopoietic tissues (bone marrow, spleen). We studied effects on serum cytokines and chemokines, and also analyzed the pharmacokinetics of 5-AED after sc administration and compared it with buccal delivery. 5-AED was administered 24 h before irradiation or sham-irradiation. Cytokine mRNAs were quantified by quantitative real-time PCR (QRT-PCR), and cytokine levels in serum by multiplex Luminex. 5-AED administration was associated with elevation of message for GM-CSF, IL-2, IL-3, IL-6, and IL-10 in spleen, and GM-CSF and IL-2 in bone marrow. Irradiation enhanced G-CSF, GM-CSF, IFN-gamma, TPO, IL-2, IL-3, IL-6, IL-10, and IL-12 in spleen, and GM-CSF, IFN-gamma, TPO, IL-3, and IL-10 in bone marrow. Serum levels of G-CSF were significantly elevated in 5-AED-treated mice 4 h after irradiation or sham-irradiation. Serum macrophage inflammatory protein-1gamma (MIP-1gamma) was significantly elevated 4 h after irradiation in 5-AED-treated mice. Plasma 5-AED peaked 2 h after sc injection (30 mg/kg), and remained significantly above control after 4 days, but not 8 days. The time course of plasma 5-AED after buccal delivery (60 mg/kg) was similar, but levels were significantly lower compared to sc delivery. Plasma 5-AED 24 h after administration was not significantly different between sc and buccal delivery. However, in contrast to many studies showing enhanced survival after sc administration of 5-AED, we found no effect on survival of buccal 5-AED. The results suggest that radioprotection is not dependent on the 5-AED concentration at the time of irradiation, but rather on events triggered during the first few hours after administration. The current results suggest that further studies are warranted to directly test the roles of cytokines in the radioprotective effects of 5-AED.


Subject(s)
Anabolic Agents/pharmacokinetics , Androstenediol/pharmacokinetics , Cytokines/genetics , Gene Expression/physiology , Radiation-Protective Agents/pharmacokinetics , Spleen/metabolism , Administration, Oral , Animals , Bone Marrow/metabolism , Bone Marrow/radiation effects , Cytokines/metabolism , Gamma Rays , Injections, Subcutaneous , Male , Mice , Mice, Inbred C3H , RNA, Messenger/metabolism , Spleen/radiation effects
2.
Radiat Res ; 156(3): 283-93, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11500137

ABSTRACT

We showed previously that 5-androstenediol stimulates myelopoiesis, increases the numbers of circulating neutrophils and platelets, and enhances resistance to infection in gamma-irradiated mice. We have extended those studies to include monocytes, natural killer (NK) cells, eosinophils and basophils, and we have measured the activation marker CD11b using flow cytometry. Androstenediol (160 mg/kg) was administered subcutaneously to female B6D2F1 mice 24 h before whole-body gamma irradiation. Androstenediol treatments increased the blood levels of neutrophils, monocytes and NK cells in unirradiated animals; decreased the numbers of circulating eosinophils; and ameliorated radiation-induced decreases in neutrophils, monocytes, NK cells, erythrocytes and platelets. The androstenediol treatments had no significant effect on the numbers of circulating B cells or T cells. CD11b labeling intensity on monocytes was decreased slightly after androstenediol treatment. In contrast, radiation or androstenediol alone caused increases in CD11b labeling intensity on NK cells. Androstenediol and radiation combined caused a marked increase in NK cell CD11b. The results indicate that androstenediol increases the numbers of the three major cell types of the innate immune system (neutrophils, monocytes and NK cells), that androstenediol-induced changes in blood elements in irradiated animals persist for at least several weeks, and that there is a significant positive interaction between radiation and administration of androstenediol in the activation of NK cells.


Subject(s)
Androstenediol/pharmacology , Leukocyte Count , Radiation-Protective Agents/pharmacology , Animals , Erythrocyte Count , Female , Flow Cytometry , Macrophage-1 Antigen/blood , Mice , Neutrophils/enzymology , Peroxidases/blood , Whole-Body Irradiation
4.
Int J Immunopharmacol ; 22(1): 1-14, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10684984

ABSTRACT

The ionizing radiation-induced hemopoietic syndrome is characterized by defects in immune function and increased mortality due to infections and hemorrhage. Since the steroid 5-androstene-3beta, 17beta-diol (5-androstenediol, AED) modulates cytokine expression and increases resistance to bacterial and viral infections in rodents, we tested its ability to promote survival after whole-body ionizing radiation in mice. In unirradiated female B6D2F1 mice, sc AED elevated numbers of circulating neutrophils and platelets and induced proliferation of neutrophil progenitors in bone marrow. In mice exposed to whole-body (60)Co gamma-radiation (3 Gy), AED injected 1 h later ameliorated radiation-induced decreases in circulating neutrophils and platelets and marrow granulocyte-macrophage colony-forming cells, but had no effect on total numbers of circulating lymphocytes or erythrocytes. In mice irradiated (0, 1 or 3 Gy) and inoculated four days later with Klebsiella pneumoniae, AED injected 2 h after irradiation enhanced 30-d survival. Injecting AED 24 h before irradiation or 2 h after irradiation increased survival to approximately the same extent. In K. pneumoniae-inoculated mice (irradiated at 3-7 Gy) and uninoculated mice (irradiated at 8-12 Gy), AED (160 mg/kg) injected 24 h before irradiation significantly promoted survival with dose reduction factors (DRFs) of 1.18 and 1.26, respectively. 5-Androstene-3beta-ol-17-one (dehydroepiandrosterone, DHEA) was markedly less efficacious than AED in augmenting survival, indicating specificity. These results demonstrate for the first time that a DHEA-related steroid stimulates myelopoiesis, and ameliorates neutropenia and thrombocytopenia and enhances resistance to infection after exposure of animals to ionizing radiation.


Subject(s)
Androstenediol/pharmacology , Bacterial Infections/immunology , Hematopoiesis/drug effects , Radiation-Protective Agents/pharmacology , Animals , Blood Platelets/drug effects , Female , Gamma Rays , Mice , Neutrophils/drug effects
5.
Endocr Res ; 23(4): 365-76, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9430824

ABSTRACT

Lewis rats display hyporesponsive hypothalamo-pituitary-adrenocortical (HPA) axes, overproduction of cytokines, and susceptibility to inflammatory disease. The Lewis corticotropin-releasing hormone (CRH) neurosecretory system contains normal numbers of vasopressin (VP)-deficient axon varicosities, but abnormally sparse VP-containing varicosities in the external zone of the median eminence, compared to the normoresponsive Sprague Dawley (SD), Wistar and Fischer 344 strains. Since VP may act as a thyrotropin-releasing factor, we hypothesized that thyroid axis responsivity may be altered in Lewis rats. T3, T4 and TSH were measured by radioimmunoassay, and free T4 by equilibrium dialysis, in adult male Lewis and SD rats. One h cold (5 degrees C) induced significant increases in T3, T4 and TSH levels in Lewis rats but not in SD rats. Ninety min insulin-induced hypoglycemia (1 IU/kg, i.p.) induced a significant T3 increase in Lewis rats and a significant T4 increase in SD rats. Two h after ip LPS (0.25 or 0.75 mg/kg), T4 levels fell significantly in Lewis rats but not in SD rats. TSH decreases were significant in Lewis rats after 0.75 mg/kg and in SD rats after 0.25 mg/kg. Baseline hormone levels were generally higher in Lewis rats; the differences were significant for T3 and T4 in the insulin experiments and for T3, T4 and free T4 in the LPS experiments. The data suggest that reduced inhibition from the adrenocortical axis in Lewis rats leads to hyperresponsivity of the thyroid axis to cold, and greater LPS-induced decreases in T4 levels, probably due to an exaggerated inhibitory cytokine response.


Subject(s)
Adrenal Cortex/physiopathology , Corticotropin-Releasing Hormone/genetics , Neurosecretory Systems/cytology , Pituitary Gland/physiopathology , Pituitary-Adrenal System/physiopathology , Thyroid Gland/physiology , Vasopressins/genetics , Adrenocorticotropic Hormone/blood , Animals , Axons/physiology , Blood Glucose/drug effects , Blood Glucose/metabolism , Cold Temperature , Corticotropin-Releasing Hormone/metabolism , Dose-Response Relationship, Drug , Fasting , Hypoglycemia/blood , Hypoglycemia/chemically induced , Hypoglycemic Agents/pharmacology , Hypothalamo-Hypophyseal System/physiopathology , Insulin/pharmacology , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/pharmacology , Male , Mutation , Neurosecretory Systems/chemistry , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Thyroid Gland/drug effects , Thyrotropin/blood , Thyrotropin/drug effects , Thyroxine/blood , Triiodothyronine/blood , Vasopressins/deficiency
6.
Radiat Res ; 141(3): 336-41, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7871163

ABSTRACT

Recombinant human interleukin-1 alpha (rhIL-1 alpha) has significant potential as a radioprotector and/or treatment for radiation-induced hematopoietic injury. Both IL-1 and whole-body ionizing irradiation acutely stimulate the hypothalamic-pituitary-adrenal axis. We therefore assessed the interaction of whole-body irradiation and rhIL-1 alpha in altering the functioning of the axis in mice. Specifically, we determined the adrenocorticotropin (ACTH) and corticosterone responses to rhIL-1 alpha administered just before and hours to days after whole-body or sham irradiation. Our results indicate that whole-body irradiation does not potentiate the rhIL-1 alpha-induced increase in ACTH levels at the doses used. In fact, the rhIL-1 alpha-induced increase in plasma ACTH is transiently impaired when the cytokine is administered 5 h after, but not 1 h before, exposure to whole-body irradiation. The ACTH response may be inhibited by elevated corticosterone levels after whole-body irradiation, or by other radiation-induced effects on the pituitary gland and hypothalamus.


Subject(s)
Adrenocorticotropic Hormone/drug effects , Adrenocorticotropic Hormone/radiation effects , Interleukin-1/pharmacology , Radiation-Protective Agents/pharmacology , Whole-Body Irradiation , Adrenalectomy , Adrenocorticotropic Hormone/physiology , Animals , Corticosterone/blood , Female , Humans , Mice , Mice, Inbred C3H , Recombinant Proteins/pharmacology
7.
Exp Cell Res ; 216(2): 411-21, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7843286

ABSTRACT

Digitally enhanced low-light-level fluorescence video microscopy and immunochemical staining were used to examine influenza virus envelope lipid and protein redistribution during pH-induced fusion. Video microscopy was performed using viruses labeled with either the lipid analogue octadecylrhodamine B (R18) or fluorescein isothiocyanate (FITC) covalently linked to envelope proteins. Viruses were bound to human red blood cells, and the pattern and intensity of fluorescence were monitored for 30 min while cell-virus complexes were perfused with pH 7.4 or 4.8 media at temperatures either above or below 20 degrees C. R18 showed complete redistribution and dequenching by 30 min at all incubation temperatures, confirming reports that viral fusion occurs at subphysiological temperatures. FITC-labeled protein showed spatial redistribution at 28 degrees C but no change at low temperature. Electron microscopy observations of immunochemical staining of viral proteins confirmed both that protein redistribution at 37 degrees C was slower than R18 and the failure of movement within 30 min at 16 degrees C. Video microscopy monitoring of RNA staining by acridine orange of virus-cell complexes showed redistribution to the RBCs at all temperatures but only after low pH-induced fusion. The results are consistent with differential dispersion of viral components into the RBC and the existence of relatively long-lived barriers to diffusion subsequent to fusion pore formation.


Subject(s)
Erythrocyte Membrane/metabolism , Membrane Fusion , Membrane Lipids/metabolism , Orthomyxoviridae/physiology , Viral Envelope Proteins/metabolism , Animals , Erythrocyte Membrane/ultrastructure , Erythrocytes/virology , Humans , Hydrogen-Ion Concentration , Kinetics , Microscopy, Fluorescence , Microscopy, Video , RNA, Viral/metabolism , Rabbits , Temperature
8.
Neuroendocrinology ; 60(4): 418-25, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7824083

ABSTRACT

The administration of lipopolysaccharide (LPS) results in the activation of the hypothalamic-pituitary-adrenal axis (HPAA). We recently reported that the participation and interaction of LPS-induced proinflammatory cytokines were obligatory for the stimulation of adrenocorticotropic hormone (ACTH) release by LPS. LPS and LPS-derived cytokines also stimulate the release of histamine (HA). HA is a known hypothalamic neurotransmitter and activates the HPAA. Therefore, to elucidate the role of HA in LPS- and cytokine-induced ACTH release, we evaluated the effects of several HA H1 and H2 receptor antagonists on the ACTH response to LPS, recombinant human interleukin-1 alpha (rhIL-1 alpha) and HA in mice. Although all 3 of the H1 receptor antagonists administered (mepyramine (MEP), diphenhydramine (DPH) or promethazine (PMZ) were able to block the 10-min ACTH response to HA, only PMZ (a less selective H1 receptor antagonist than MEP) was able to reduce the LPS- or rhIL-1 alpha-induced ACTH responses. Ranitidine, a powerful and selective H2 receptor antagonist, had little effect on the LPS- and rhIL-1 alpha-induced ACTH responses, while metiamide (MET), a much less potent first-generation H2 receptor antagonist, substantially diminished ACTH release. The greater effectiveness of PMZ, in contrast to MEP or DPH, probably relates to the ability of phenothiazine derivatives to inhibit non-HA-dependent pathways involved in the stimulation of the HPAA by cytokines; the same may be true of MET.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Adrenocorticotropic Hormone/metabolism , Histamine H1 Antagonists/pharmacology , Histamine H2 Antagonists/pharmacology , Interleukin-1/pharmacology , Lipopolysaccharides/pharmacology , Adrenocorticotropic Hormone/blood , Animals , Female , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Mice , Mice, Inbred C3H , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Recombinant Proteins/pharmacology
9.
Neuroreport ; 5(13): 1635-7, 1994 Aug 15.
Article in English | MEDLINE | ID: mdl-7819536

ABSTRACT

Rats possess stress-responsive, vasopressin (VP)-expressing and stress-nonresponsive, VP-deficient subpopulations of parvocellular corticotropin-releasing hormone (CRH) neurosecretory cells. Both subpopulations are activated by bacterial lipopolysaccharide (LPS) and cytokines. Lewis rats exhibit hyporesponsive hypothalamo-pituitary-adrenocortical axes (HPAAs). The Lewis CRH neurosecretory system has been reported to be defective in females and normal in males. We used post-embedding electron microscopic (EM) immunocytochemistry to study baseline levels and LPS-stimulated depletion of neurosecretory vesicles. Male Lewis rats possessed normal numbers of CRH+/VP- varicosities and low numbers of CRH+/VP+ varicosities, indicating abnormally low release of VP into portal blood. This defect contrasts with the reported increase in VP content and release in magnocellular neurosecretory cells in Lewis rats.


Subject(s)
Corticotropin-Releasing Hormone/physiology , Hypothalamo-Hypophyseal System/physiology , Vasopressins/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Female , Hypothalamo-Hypophyseal System/cytology , Hypothalamo-Hypophyseal System/drug effects , Immunohistochemistry , Lipopolysaccharides/pharmacology , Male , Microscopy, Electron , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Rats, Sprague-Dawley , Rats, Wistar
10.
Endocrinology ; 133(5): 2284-91, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8404682

ABSTRACT

This report describes for the first time the cellular and subcellular localization of estrogen sulfotransferase (EST) as well as the subcellular localization of hydroxysteroid sulfotransferase (HST) in the mammalian adrenal cortex. A 34-kilodalton EST and two HSTs with 3 alpha- and 3 beta-hydroxysteroid substrate specificities (32 and 33 kilodaltons, respectively) were previously purified from guinea pig adrenal cortex and characterized. Western blots were used to establish that two antisera generated against EST were highly specific for EST, whereas three antisera generated against the HSTs were highly specific for the HSTs, but did not distinguish between the 3 alpha- and 3 beta HSTs. Light and electron microscopic immunoperoxidase labeling with these antisera revealed that the sulfotransferases were expressed only within the ACTH-responsive layers of the guinea pig adrenal cortex, with EST localized to zona fasciculata and zona reticularis cells, and the HSTs confined to the zona reticularis. No labeling was detected in the zona glomerulosa or adrenal medulla. EST was concentrated in cell nuclei; sparse labeling was distributed throughout the cytoplasm. HST labeling was intense in smooth endoplasmic reticulum of zona reticularis cells, but was absent from nuclei. Ovoid inclusions about 1-4 microns in diameter, with no limiting membrane, were observed in zona reticularis cells; these inclusions were strongly labeled for both EST and HSTs. No gender-specific differences in distribution or labeling intensity were apparent. The high concentration of EST immunoreactivity in nuclei suggests that EST may play a role in modulating the ability of active estrogens to regulate gene expression in ACTH-responsive cells. The distribution of HST labeling suggests that sulfonation of adrenocortical 3-hydroxysteroids takes place largely within smooth endoplasmic reticulum in the zona reticularis in adult guinea pigs.


Subject(s)
Adrenal Cortex/enzymology , Sulfotransferases , Sulfurtransferases/analysis , Adrenal Cortex/ultrastructure , Adrenal Medulla/enzymology , Animals , Blotting, Western , Cell Nucleus/enzymology , Cytoplasm/enzymology , Female , Guinea Pigs , Immunoenzyme Techniques , Male , Microscopy, Electron , Zona Fasciculata/enzymology , Zona Glomerulosa/enzymology , Zona Reticularis/enzymology
11.
Neuroendocrinology ; 58(1): 42-8, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8264854

ABSTRACT

Stimulation of the rat hypothalamopituitary-adrenal axis during stress involves activation of central alpha 1-adrenergic receptors. The subpopulation of corticotropin-releasing hormone (CRH) neurosecretory cells that contains vasopressin (VP) is selectively activated by several types of stress (immobilization, hypoglycemia, and intracerebroventricular, i.c.v., colchicine), and is located in a catecholamine-rich area of the hypothalamic paraventricular nucleus. Therefore, we tested the hypothesis that the CRH+/VP+ subpopulation is selectively activated by central alpha 1-adrenergic receptors. The alpha 1-agonist methoxamine or vehicle alone was injected i.c.v. after habituation of rats to daily injections of vehicle through a chronic i.c.v. cannula. Activation of the CRH+/VP+ and CRH+/VP- subpopulations was measured by quantifying depletion of neurosecretory vesicles from immunocytochemically identified axons in the external zone of the median eminence. The habituated, vehicle-injected sham control group had normal levels of plasma ACTH and corticosterone, but possessed a significantly higher proportion of VP-containing CRH axons than naive animals. This change is similar to what was observed previously in rats subjected to repeated daily stress. I.c.v. methoxamine caused elevations of plasma ACTH and corticosterone and significant depletions of vesicles from the CRH+/VP+ axons at 1 and 2 h after injection, compared to the sham control group. The CRH+/VP- axons, however, displayed significant accumulations of neurosecretory vesicles at the same times after 300 micrograms methoxamine, compared to the sham control group. After 100 micrograms methoxamine, there was no change in the CRH+/VP- axons, compared to the sham control group.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Corticotropin-Releasing Hormone/analysis , Methoxamine/pharmacology , Neurosecretory Systems/chemistry , Receptors, Adrenergic, alpha-1/drug effects , Stress, Physiological/physiopathology , Adrenocorticotropic Hormone/blood , Animals , Axons/drug effects , Catheterization , Corticosterone/blood , Habituation, Psychophysiologic/physiology , Injections, Intraventricular , Male , Neurosecretory Systems/cytology , Rats , Rats, Sprague-Dawley
13.
Endocrinology ; 132(3): 946-52, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8382602

ABSTRACT

Administration of lipopolysaccharide (LPS) results in activation of the hypothalamic-pituitary-adrenal axis. LPS induces the release of a number of proinflammatory cytokines, i.e. interleukin-1 (IL-1), IL-6, and tumor necrosis factor (TNF), which activate the hypothalamic-pituitary-adrenal axis as well and may mediate the effects of LPS. Variations in the kinetics of appearance of IL-1, TNF, and IL-6 after LPS challenge suggested that these cytokines may play different roles at different times. To elucidate the mutual dependence and contribution of individual cytokines in the course of LPS-induced ACTH release, we used blocking antibodies to IL-6, TNF, and the IL-1 receptor. Our results demonstrate that anti-IL-6 antibody abrogated ACTH induction throughout the course of the response both 2 and 4 h after LPS challenge. In contrast, anti-IL-1 receptor and anti-TNF antibody, given individually, blocked ACTH production at 4 h, but not at 2 h. Only combined administration of these two antibodies diminished, but did not eliminate, ACTH release at 2 h. This is the first demonstration that all three inflammatory cytokines are obligatory for LPS-induced elevation of plasma ACTH. In addition, these results suggest that IL-1, IL-6, and TNF play different roles in LPS-induced ACTH release.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Interleukin-1/pharmacology , Interleukin-6/pharmacology , Lipopolysaccharides/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Adrenocorticotropic Hormone/blood , Animals , Drug Interactions , Escherichia coli , Female , Humans , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Kinetics , Lipopolysaccharides/isolation & purification , Mice , Mice, Inbred C3H , Recombinant Proteins/pharmacology , Reference Values , Time Factors
14.
Endocrinology ; 131(1): 37-44, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1319322

ABSTRACT

Administration of interleukin-1 (IL-1) induces increases in plasma ACTH and glucocorticoids. Numerous experiments have implicated the hypothalamic CRH neurosecretory system in these responses, but have failed to provide evidence for involvement of the ACTH secretagogue vasopressin (VP). The rat CRH neurosecretory system contains two types of cells: VP expressing and VP deficient. Hence, the above findings suggested that IL-1 may selectively activate the VP-deficient subtype of CRH neurosecretory cells. In this study we employed postembedding electron microscopic immunocytochemistry to directly assay IL-1-induced depletion of secretory vesicles from identified VP-expressing and VP-deficient CRH neurosecretory axons. IL-1-induced depletion of secretory vesicles from these axons was correlated with increases in plasma ACTH and decreases in plasma PRL. No dose of IL-1 was found that could selectively activate one subtype of CRH neurosecretory axons; at doses of 0.67 microgram/100 g and above for both IL-1 alpha and IL-1 beta, equal depletion of vesicles from the two subtypes was observed. Similar results were previously found after the injection of bacterial lipopolysaccharide, which induces the release of IL-1 from macrophages. The findings unequivocally establish for the first time that IL-1 activates hypothalamic CRH neurosecretory cells in the absence of surgical stress, anesthesia, disruption of the infundibular area, or administration of toxic drugs. In addition, these data clearly demonstrate that IL-1 induces the release of VP from neurosecretory axons in the portal capillary zone of the external zone of the median eminence. Previous studies have shown that the VP-deficient subtype of CRH neurosecretory axons is not strongly activated by several types of stress; therefore, activation of the system by inflammatory mediators involves mechanisms different from those mediating the stress response.


Subject(s)
Axons/physiology , Corticotropin-Releasing Hormone/metabolism , Interleukin-1/pharmacology , Neurosecretory Systems/physiopathology , Stress, Physiological/physiopathology , Adrenocorticotropic Hormone/blood , Animals , Blood Glucose/metabolism , Cytoplasmic Granules/physiology , Cytoplasmic Granules/ultrastructure , Immunoenzyme Techniques , Kinetics , Male , Median Eminence/physiopathology , Median Eminence/ultrastructure , Microscopy, Electron , Neurosecretory Systems/ultrastructure , Prolactin/blood , Rats , Rats, Inbred Strains , Stress, Physiological/pathology , Vasopressins/metabolism
15.
Neuroendocrinology ; 53(2): 150-9, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1849619

ABSTRACT

Stress-induced release of corticotropin-releasing factor (CRF) and vasopressin (AVP) was studied in rats by measuring the decline of CRF and AVP stores in the median eminence after blockade of fast axonal transport with colchicine (5 micrograms per rat intracisternally). Quantitative immunocytochemistry was used to detect changes in CRFi and AVPi in the external zone of the median eminence (ZEME) selectively. Immobilization stress induced a fast ACTH response to 1,000-2,000 pg/ml which was associated with a fall in both CRFi and AVPi of 34% during the first 30 min. This is followed by different time courses of further AVPi and CRFi depletion. In addition, we investigated the effect of repeated daily stress exposure on CRFi and AVPi in the ZEM 1 day after stress exposure. Repeated daily immobilization for 9 or 16 subsequent days did not affect the CRFi stores in the ZEME, but increased the AVPi stores to 161 +/- 13% and 218 +/- 11% respectively. Quantitative analysis of electron microphotographs of repeatedly handled rats showed a mean density of CRF positive profiles in the ZEME of 45.5 +/- 2.5 per 500 microns 2 of which 25% also stained for pro-AVP-derived peptides. After 9 subsequent days of immobilization the total density of CRF-positive profiles remained unchanged, but the fraction of CRF swellings that also stained for pro-AVP-derived peptides increased approximately 2-fold. We conclude that (1) the secretion of AVPi and CRFi from the ZEME are independently controlled, indicating differential activation of AVP containing and AVP deficient CRF neurons during acute immobilization, and (2) repeated stress leads to plastic changes in hypothalamic CRF neurons resulting in increased AVP stores and colocalization in CRF nerve terminals.


Subject(s)
Arginine Vasopressin/metabolism , Corticotropin-Releasing Hormone/metabolism , Median Eminence/metabolism , Neurons/physiology , Stress, Physiological/metabolism , Adrenocorticotropic Hormone/blood , Animals , Colchicine/pharmacology , Kinetics , Male , Rats , Rats, Inbred Strains , Restraint, Physical , Stress, Physiological/etiology
16.
J Comp Neurol ; 302(2): 220-35, 1990 Dec 08.
Article in English | MEDLINE | ID: mdl-1705265

ABSTRACT

The highest molecular weight neurofilament protein (NF-H) is multiply phosphorylated at epitopes which can be distinguished by specific monoclonal antibodies on Western blots. Eight characterized antibodies were used in immunocytochemistry to examine the tissue distributions of phosphorylated variants of NF-H in axons of the adult rat spinal cord. The most striking difference in staining was found between axons in the cuneate tract and those in the neighboring dorsal corticospinal tract. Axons in the cuneate tract reacted intensely with antibodies to phosphorylated epitopes of NF-H and poorly with antibodies to dephosphorylated epitopes of NF-H, whereas the reverse was the case for the axons of the dorsal corticospinal tract. These differences showed that systematic variations in the phosphorylation of NF-H in long-tract axons in the central nervous system occur as a function of cell type. When the cytoskeletons of these axons were compared by electron microscopy, the neurofilaments of the cuneate fibers were seen to be more abundant and formed a latticework, more compactly organized than the neurofilaments of the dorsal corticospinal axons. By comparison, the dorsal corticospinal axons were relatively richer in microtubules than the cuneate axons. Although the cuneate fiber tract contained many more large (greater than 2.0 microns 2 in cross section) axons than did the dorsal corticospinal tract, these differences in cytoskeletal organization were apparent even when myelinated axons of similar sizes (0.4 micron 2 to 2.0 microns 2) were compared. In addition, the number of neurofilaments in cuneate axons in the 0.4 to 2.0 microns 2 size range was significantly better correlated with axon size than was the case for this size range of dorsal corticospinal axons. Thus, the differences seen in the organization of the neurofilament latticework and the phosphorylation of NF-H between axons found in these two tracts both appeared to be correlated with cell type, and were independent of length or caliber of the axons.


Subject(s)
Axons/chemistry , Epitopes/analysis , Intermediate Filament Proteins/immunology , Intermediate Filaments/chemistry , Pyramidal Tracts/chemistry , Spinal Cord/chemistry , Animals , Antibodies, Monoclonal , Axons/ultrastructure , Blotting, Western , Immunohistochemistry , Male , Microscopy, Electron , Neural Pathways/chemistry , Neurons, Afferent/chemistry , Neurons, Afferent/ultrastructure , Phosphorylation , Rats , Rats, Inbred Strains , Spinal Cord/cytology
17.
J Histochem Cytochem ; 38(11): 1607-14, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2170503

ABSTRACT

Two proteins were isolated and purified from guinea pig adrenal cortex: a 34 KD protein that specifically binds pregnenolone (product of the rate-limiting step in steroidogenesis), and a novel co-purifying 32 KD protein that has not been characterized. Specific antisera were generated and used for immunocytochemical analysis. The 34 KD and 32 KD proteins were specific for the adrenal cortex and were absent from other tissues, including the testis. The 34 KD pregnenolone binding protein (PBP) was localized to zona fasciculata and zona reticularis cells and absent from zona glomerulosa cells. Thus, the PBP was absolutely correlated with ACTH-regulated steroidogenic cells, whereas steroidogenic cells regulated by other peptide hormones did not contain the PBP. This finding suggests a functional relationship between the PBP and ACTH. A second interesting finding was that a novel 32 KD co-purifying protein localized to the zona reticularis and was absent from the zona glomerulosa and the zona fasciculata. The 32 KD protein can therefore serve as an excellent marker for the reticularis cell of the adrenal cortex.


Subject(s)
Adrenal Cortex Hormones/metabolism , Adrenal Cortex/cytology , Carrier Proteins/metabolism , Immunohistochemistry/methods , Pregnenolone/metabolism , Zona Fasciculata/cytology , Zona Reticularis/cytology , Adrenal Cortex/metabolism , Adrenocorticotropic Hormone/physiology , Animals , Gonadotropins/physiology , Guinea Pigs , Leydig Cells/cytology , Leydig Cells/metabolism , Male , Renin-Angiotensin System/physiology , Zona Fasciculata/metabolism , Zona Reticularis/metabolism
18.
Neuroscience ; 36(1): 201-5, 1990.
Article in English | MEDLINE | ID: mdl-2215918

ABSTRACT

Parvocellular corticotropin-releasing hormone neurosecretory cells in the hypothalamic paraventricular nucleus project axons to the portal capillary plexus in the external zone of the median eminence. Immunocytochemical studies have identified two approximately equal subpopulations of these corticotropin-releasing hormone neurons in normal rats, distinguished by the presence or absence of co-existent vasopressin, and different responses to stress. However, it was recently proposed that the vasopressin deficient cells do not contain corticotropin-releasing hormone, but have been misidentified due to cross-reactivity of the corticotropin-releasing hormone antiserum to peptide histidine-isoleucineamide. It is shown here that the same set of corticotropin-releasing hormone neurons (including both vasopressin expressing and vasopressin deficient subtypes) was labeled with multiple corticotropin-releasing hormone antisera. These included two antisera that did not cross-react with peptide histidine-isoleucineamide: one against ovine corticotropin-releasing hormone, and one rat corticotropin-releasing hormone antiserum absorbed with peptide histidine-isoleucineamide. The results provide further support for the hypothesis of functionally distinct compartments of the corticotropin-releasing hormone neurosecretory system that can modulate the ratio of vasopressin to corticotropin-releasing hormone in portal blood.


Subject(s)
Corticotropin-Releasing Hormone/biosynthesis , Neurosecretory Systems/metabolism , Vasopressins/metabolism , Absorption , Animals , Antibody Specificity , Corticotropin-Releasing Hormone/immunology , Cross Reactions , Male , Median Eminence/cytology , Median Eminence/metabolism , Microscopy, Electron , Neurosecretory Systems/cytology , Rats , Rats, Inbred Strains , Sheep/immunology
19.
Neuroendocrinology ; 50(6): 702-7, 1989 Dec.
Article in English | MEDLINE | ID: mdl-2515467

ABSTRACT

A functional ultrastructural assay was used to determine the response of corticotropin releasing hormone (CRH) neurosecretory cells to short-term stress. Depletion of neurosecretory vesicles from axonal swellings in the external zone of the rat median eminence was used as a measure of functional activity. One hour of immobilization or 5 h of insulin-induced hypoglycemia caused marked depletion of vesicles from the vasopressin (VP)-containing CRH axons, but had no effect on the VP-deficient subpopulation of CRH axons. Injection of colchicine (100 micrograms) into the lateral ventricle also resulted in selective depletion of vesicles from the VP-containing subpopulation over the course of 5 h, with no depletion from the VP-deficient axons. By 24 h after injection of 100 micrograms colchicine, however, both the VP-containing and the VP-deficient axons were severely depleted of neurosecretory vesicles. These data demonstrate for the first time that the CRH neurosecretory system contains functionally distinct components, and that the VP secreting component may specifically mediate the ACTH response to short-term stress.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Median Eminence/ultrastructure , Neurons/physiology , Stress, Physiological/physiopathology , Vasopressins/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Colchicine/pharmacology , Cytoplasmic Granules/metabolism , Hypoglycemia/chemically induced , Hypoglycemia/physiopathology , Insulin , Male , Neurons/drug effects , Neurons/ultrastructure , Rats , Restraint, Physical
20.
Cell Mol Neurobiol ; 9(4): 427-46, 1989 Dec.
Article in English | MEDLINE | ID: mdl-2575930

ABSTRACT

1. Coexisting with oxytocin or vasopressin in the cell bodies and nerve terminals of the hypothalamic-neurohypophysial system are smaller amounts of other peptides. For a number of these "copeptides" there is strong evidence of corelease with the major magnocellular hormones. Guided by the location of their specific receptors we have studied the effects of three copeptides, dynorphin, cholecystokinin (CCK), and corticotropin releasing hormone (CRH), on the secretion of oxytocin and vasopressin from isolated rat neural lobe or neurointermediate lobe preparations in vitro. 2. Dynorphin is coreleased with vasopressin from neural lobe nerve terminals and acts on neural lobe kappa-opiate receptors to inhibit the electrically stimulated secretion of oxytocin. Naloxone augments oxytocin release from the neural lobe in a manner directly proportional to the amount of vasopressin (and presumably dynorphin) released. 3. Cholecystokinin, coreleased with oxytocin by neural lobe terminals, has been shown to have high-affinity receptors located in the NL and to stimulate secretion of both oxytocin and vasopressin. CCK's secretagogue effect was independent of electrical stimulation and extracellular Ca2+ and was blocked by an inhibitor of protein kinase C. 4. CRH, coreleased with OT from the neural lobe, has receptors in the intermediate lobe of the pituitary, but not in the neural lobe itself. CRH stimulates the secretion of oxytocin and vasopressin from combined neurointermediate lobes but not from isolated neural lobes. Intermediate lobe peptides, alpha and gamma melanocyte stimulating hormone, induced secretion of oxytocin and vasopressin from isolated neural lobes. Their effect was, like that of CCK, independent of electrical stimulation and extracellular Ca2+ and blocked by an inhibitor of protein kinase C. 5. Among the CRH-producing parvocellular neurons of the paraventricular nucleus, in the normal rat, approximately half also produce and store vasopressin. After removal of glucocorticoid influence by adrenalectomy, virtually all of the CRH neurons contain vasopressin. 6. The two subtypes of CRH neurosecretory cells found in the normal rat possess different topographical distributions in the paraventricular nucleus, suggesting the possibility of differential innervation. Stress selectively activates the vasopressin containing subpopulation of CRH neurons, indicating that there are separate channels of regulatory input controlling the two components of the parvocellular CRH neurosecretory system.


Subject(s)
Cholecystokinin/pharmacology , Corticotropin-Releasing Hormone/pharmacology , Dynorphins/pharmacology , Hypothalamus/metabolism , Animals , Oxytocin/metabolism , Rats , Vasopressins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...