Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Neurosci ; 20(14): 5544-51, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10884338

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a hematopoietic cytokine that may affect various functions of the CNS because the molecule and its receptors are expressed in the brain. The present study examines the effects of GM-CSF on sleep using rats and the secretion of three neurotransmitters/hormones that are involved in sleep regulation. When infused intracerebroventricularly at doses as low as 10 pmol for 10 hr during the dark period, GM-CSF promoted predominantly rapid eye movement (REM) sleep and moderate amounts of non-REM sleep without eliciting fever. An injection of GM-CSF (3.0 pmol) into the arcuate nucleus increased the release of nitric oxide (NO) from the hypothalamus but did not alter plasma levels of growth hormone. The release of somatostatin (SRIF) from the medial basal hypothalamus was stimulated by 1 x 10(-)(11) M GM-CSF. These findings indicated that centrally administered GM-CSF stimulates SRIF release through activation of the NO system in the hypothalamus. Because SRIF promotes REM sleep, it may also mediate the effects of GM-CSF on REM sleep. The present study indicates a novel central effect of GM-CSF that modulates sleep, supporting the notion that hematopoietic cytokines also play roles in the CNS.


Subject(s)
Brain/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Sleep, REM/physiology , Animals , Antibodies/pharmacology , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Body Temperature/drug effects , Brain/drug effects , Circadian Rhythm/drug effects , Culture Media, Conditioned/chemistry , Dose-Response Relationship, Drug , Electroencephalography , Electromyography , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Growth Hormone/blood , In Vitro Techniques , Injections, Intraventricular , Male , Microinjections , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Sleep, REM/drug effects , Somatostatin/analysis , Somatostatin/biosynthesis
2.
Brain Res ; 842(2): 351-8, 1999 Sep 25.
Article in English | MEDLINE | ID: mdl-10526131

ABSTRACT

Ataxia-telangiectasia (A-T) is a human disorder with pleiotropic manifestations that include neoplasms, immune dysfunction and neurodegeneration. The disorder is due to mutations in the gene known as ATM (A-T, mutated), which causes a deficiency in its protein product (Atm in mice) that is necessary for DNA damage surveillance. This nuclear function of Atm explains in principle the propensity to cancer and immunodeficiency in A-T, but not the neurodegeneration which results in the earliest clinical manifestations and causes progressive disability. Here we report ultrastructural evidence of cytoplasmic localization of Atm-like immunoreactivity (ALI) within endosomes in murine cerebellocortical neurons, one of the principal targets of A-T. The ALI was obtained with two separate monoclonal antibodies that recognize Atm specifically. By contrast, electron-dense endosomes that could be confused with ALI occur in negligible amounts in both wild-type mice and in mice deficient in Atm ("knockout" mice). Furthermore, there was a marked preferential distribution of Atm-immunopositive endosomes in the granule cell layer - where they are present in granule neurons - with a much lower density in the Purkinje and molecular layers. These observations suggest that endosome-bound Atm may be more important for the function of certain neurons than others - or that it is processed differently among them - and that this protein may be involved in molecular sorting in the cytoplasm. This is relevant to elucidating the role of Atm deficiency in the pathobiology of neurodegeneration in A-T.


Subject(s)
Ataxia Telangiectasia/genetics , Cerebellar Cortex/metabolism , Endosomes/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Endosomes/ultrastructure , Humans , Mice , Mice, Inbred Strains , Mice, Knockout , Microscopy, Immunoelectron , Protein Serine-Threonine Kinases/analysis , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Proteins
3.
Med Hypotheses ; 52(5): 377-82, 1999 May.
Article in English | MEDLINE | ID: mdl-10416943

ABSTRACT

Ataxia-telangiectasia (A-T) is a pleiotropic, multi-system disorder with manifestations that include immune deficiency, sensitivity to ionizing radiation and neoplasms. Many of these manifestations are understood in principle since the identification in A-T patients of mutations in a gene encoding a protein kinase that plays a key role in signaling and repair of DNA damage. However, the cause of the neurodegeneration that afflicts patients with A-T for at least a decade before they succumb to overwhelming infections or malignancy remains mysterious. Based on our work in a mouse model of A-T and previous evidence of extra-neural autoimmune disorders in A-T, we postulate that the neurodegenerative process in A-T is not due to a function for A-T mutated (ATM) essential for the postnatal brain, but to an autoimmune process (hence 'horror autotoxicus', Paul Ehrlich's term for autoimmune disorder). This hypothetical mechanism may be analogous to that in the so-called 'paraneoplastic' neurodegenerative syndromes in patients with various malignancies. Thus, alterations in the balance between cellular and humoral immunity in A-T probably result in autoantibodies to cerebral epitopes shared with cells of the immune system. This hypothesis has important implications for the understanding and development of effective palliative and even preventative strategies for A-T, and probably for other so far relentlessly progressive neurodegenerative disorders.


Subject(s)
Ataxia Telangiectasia/physiopathology , Autoimmune Diseases/physiopathology , Nerve Degeneration/physiopathology , Animals , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/immunology , Autoantibodies , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Brain/immunology , DNA Damage , DNA Repair , Humans , Mice , Models, Biological , Nerve Degeneration/etiology
4.
Brain Res ; 816(1): 241-4, 1999 Jan 16.
Article in English | MEDLINE | ID: mdl-9878761

ABSTRACT

Nitric oxide (NO) synthase (NOS)-containing cerebrocortical neurons degenerate in patients with amyotrophic lateral sclerosis (ALS) and dementia, and in transgenic mice expressing a mutated superoxide dismutase gene (G93A) associated with familial ALS. The cerebral cortex of transgenic mice displayed decreased NOS activity (p<0.001) and cGMP levels (p<0.01), but no changes in NOS content indicating that less NO is produced. Therefore, NOSN degeneration is not caused by elevated NO.


Subject(s)
Cerebral Cortex/enzymology , Nerve Degeneration/metabolism , Neurons/enzymology , Nitric Oxide/biosynthesis , Superoxide Dismutase/genetics , Amino Acid Substitution , Animals , Blotting, Western , Cerebellum/enzymology , Cerebellum/metabolism , Cerebellum/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cyclic GMP/metabolism , Disease Models, Animal , Genotype , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Nerve Degeneration/genetics , Neurons/metabolism , Neurons/pathology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Polymerase Chain Reaction , Superoxide Dismutase/metabolism
5.
Peptides ; 19(9): 1573-9, 1998.
Article in English | MEDLINE | ID: mdl-9864065

ABSTRACT

Growth hormone (GH) secretion from the pituitary is known to be under the dual control of GH-releasing factor (GRF) and somatostatin (SRIF). Hypothalamic SRIF, the major inhibitor of pituitary growth hormone secretion, inhibits its own release by a negative ultrashort-loop feedback mechanism. However, it is not known whether this negative regulation is mediated by inhibition of SRIF mRNA production. GRF may also inhibit its own release, thereby modifying pituitary GH secretion, possibly through an ultrashort-loop feedback mechanism. Thus, SRIF production and GRF release are both regulated by SRIF. Periventricular nucleus (PeN) and mediobasal hypothalamus (MBH) from adult male rats were incubated for 6 h in Waymouth's medium with either SRIF or the SRIF agonist analog RC 160 (10(-9) to 10(-6) M). Levels of SRIF mRNA were determined by an S1 nuclease protection assay using a 32[P]-labeled rat SRIF riboprobe. SRIF (10(-7) M) and RC 160 (10(-8), 10(-7) M) significantly (p< or =0.01) decreased SRIF mRNA levels in the PeN. The levels of SRIF mRNA in the MBH were not modified by either SRIF or RC 160. SRIF (10(-7) and 10(-6) M) significantly (p < or = 0.01 and p < or = 0.001, respectively) inhibited the release of GRF at 30 min in the MBH. Likewise, the release of GRF was slightly decreased by 10(-7) M RC 160, and significantly inhibited by 10(-6) M (p < or = 0.001) at 30 min. At 6 h, the levels of GRF were significantly reduced by 10(-7) M SRIF (p < or = 0.05) and by RC 160 (10(-7), 10(-6) M; p < or = 0.001 and p < or = 0.05, respectively). In contrast with these results, the SRIF analog was unable to alter SRIF release at 30 min. At 6 h incubation, RC 160 (10(-7) M) significantly (p < or = 0.001) reduced SRIF release from MBH fragments. These results demonstrate that SRIF and a SRIF analog decrease SRIF mRNA levels in the PeN and inhibit the release of SRIF from the nerve terminals of the MBH. Thus, SRIF appears to regulate its own gene expression by negative ultrashort-loop feedback. Therefore, when SRIF is secreted from these neurons in response to GRF, it down-regulates the preceding stimulatory input as well as its own secretion.


Subject(s)
Cerebral Ventricles/drug effects , Hypothalamus/drug effects , RNA, Messenger/analysis , Somatostatin/metabolism , Somatostatin/pharmacology , Animals , Feedback , Gene Expression , Growth Hormone-Releasing Hormone/metabolism , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley , Somatostatin/analogs & derivatives , Somatostatin/genetics
6.
Proc Natl Acad Sci U S A ; 94(23): 12688-93, 1997 Nov 11.
Article in English | MEDLINE | ID: mdl-9356511

ABSTRACT

Neural degeneration is one of the clinical manifestations of ataxia-telangiectasia, a disorder caused by mutations in the Atm protein kinase gene. However, neural degeneration was not detected with general purpose light microscopic methods in previous studies using several different lines of mice with disrupted Atm genes. Here, we show electron microscopic evidence of degeneration of several different types of neurons in the cerebellar cortex of 2-month-old Atm knockout mice, which is accompanied by glial activation, deterioration of neuropil structure, and both pre- and postsynaptic degeneration. These findings are similar to those in patients with ataxia-telangiectasia, indicating that Atm knockout mice are a useful model to elucidate the mechanisms underlying neurodegeneration in this condition and to develop and test strategies to palliate and prevent the disease.


Subject(s)
Ataxia Telangiectasia , Mice, Knockout , Neuropil/pathology , Protein Serine-Threonine Kinases , Proteins/genetics , Synapses/pathology , Animals , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Disease Models, Animal , Mice , Microscopy, Electron , Neuroglia/pathology , Neurons/pathology , Tumor Suppressor Proteins
7.
Endocrinology ; 137(5): 1585-90, 1996 May.
Article in English | MEDLINE | ID: mdl-8612489

ABSTRACT

We have previously shown that somatostatin (SRIF) is synthesized in B and T lymphocytes of rat spleen and thymus and released into the medium of cultured lymphocytes. To determine the role of SRIF in the control of lymphocytes proliferation, the expression of SRIF in normal lymphocytes was inhibited using a 3'-terminal phosphorothioate-modified antisense oligonucleotide complementary to a sequence that includes the translation start site of the rat SRIF messenger RNA. Spleens were obtained from adult male rats, and their lymphocytes were cultured for 24 or 72 h to measure SRIF content and cell proliferation, respectively. For the proliferation studies, [3H]thymidine was incorporated during the final 18 h. The lymphocytes were incubated with 15-30 micrograms/ml SRIF antisense and control antisense. SRIF antisense (25 micrograms/ml) increased lymphocyte proliferation 15-fold (P < or = 0.001), reaching a plateau (25- to 30-fold increase) between 25-30 micrograms/ml SRIF antisense. SRIF was extracted from lymphocytes and measured by RIA. Levels of SRIF content were almost undetectable with 10 micrograms/ml antisense and were significantly lower (P < or = 0.01) with 25 micrograms/ml antisense. When RC 160 (10(-5) M), a SRIF agonist analog, was used in the incubation, the stimulation of cell proliferation exerted by the SRIF antisense was completely abolished. Control antisense had no effect on proliferation or SRIF content. These findings indicate that 1) lymphocytes in culture are able to incorporate SRIF antisense; and 2) SRIF antisense inhibits the expression of lymphocytic SRIF, which leads to lymphocyte proliferation. In conclusion, cell proliferation is dramatically increased by eliminating the expression of SRIF from the lymphocytes, which indicates that in vitro SRIF is acting in a paracrine and/or autocrine fashion to inhibit lymphocyte proliferation.


Subject(s)
Lymphocyte Activation/drug effects , Oligonucleotides, Antisense/pharmacology , Somatostatin/physiology , Animals , Base Sequence , Cells, Cultured , Lipopolysaccharides/pharmacology , Lymphocyte Activation/physiology , Male , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Somatostatin/antagonists & inhibitors , Somatostatin/genetics
8.
Neuroimmunomodulation ; 2(3): 166-73, 1995.
Article in English | MEDLINE | ID: mdl-8646567

ABSTRACT

Our previous studies have shown that the microinjection of interleukin (IL)-2 into the third ventricle of conscious rats evokes the release of adrenocorticotropin hormone (ACTH) and that its incubation with hemipituitaries in vitro was also effective in releasing ACTH. In the present experiments, we evaluated the effect of IL-2 on the release of corticotropin-releasing factor (CRF) from medial basal hypothalami (MBHs) incubated in vitro and studied the effect of other agents, whose release is altered in stress, on CRF release. IL-2 significantly stimulated CRF release at concentrations of 10(-13) and 10(-14) M, whereas increasing the concentration to 10(-12) to 10(-10) M did not produce significant release of CRF. A high concentration of potassium (55 mM) in the medium also significantly stimulated CRF release and this stimulation was not modified by IL-2. Since high-potassium-induced release of CRF is probably due to opening of voltage-dependent calcium channels, it is likely that IL-2 is releasing CRF by this mechanism. Since the release of luteinizing-hormone-releasing hormone (LHRH) is modified by stress, we evaluated the action of LHRH on CRF release and the release induced by IL-2. Although LHRH failed to alter basal CRF release, except for a slight decrease at 10(-7) M, it completely blocked IL-2-induced CRF release at this concentration. To examine a possible role for opioid peptides in CRF release, the opiate receptor blocker, naloxone (NAL), was tested. At concentrations of 5 x 10(-6) and 10(-5) M, it produced a marked increase in CRF release; however, the simultaneous exposure of MBHs to each of these concentrations of NAL plus IL-2 caused a dose-dependent decrease in IL-2-induced CRF release, suggesting that beta-endorphin or other opioid peptides may play a role in IL-2-induced CRF release. As has been previously shown for IL-1 and IL-6, IL-2-induced CRF release was blocked by alpha-melanocyte-stimulating hormone (alpha-MSH), which at high concentrations also reduced basal CRF release. As in the case of IL-1 and IL-2, dexamethasone (DEX), the highly active synthetic glucocorticoid, although not altering basal CRF release, completely blocked the response to IL-2. The inhibitor of cyclooxygenase, indomethacin (IND), also blocked IL-2-induced CRF release just as it has previously been shown to block IL-1- and IL-6-induced CRF release. The results are consistent with the hypothesis that IL-2 acts on its recently discovered receptors to induce an increase in intracellular calcium. In other experiments, we have shown that this activates nitric oxide (NO) synthase leading to production of NO by a NOergic neuron. NO diffuses to the CRF neuron and activates cyclo-oxygenase leading to generation of prostaglandin E2, which activates adenylate cyclase and increases cyclic AMP release, which then causes extrusion of CRF secretory granules. DEX presumably acts on its receptors on the CRF neuron to inhibit the increase in intracellular calcium and thereby blocks activation of phospholipase A2 necessary for activation of the arachidonic acid cascade. alpha-MSH and LHRH may similarly act on their receptors on these cells to, in some manner, block the pathway. On the other hand, beta-endorphin and/or other opioid peptides inhibit the pathway. Further experiments will be necessary to elucidate the exact points in the pathway at which these compounds are effective.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Dexamethasone/pharmacology , Gonadotropin-Releasing Hormone/pharmacology , Hypothalamus/drug effects , Indomethacin/pharmacology , Interleukin-2/pharmacology , Naloxone/pharmacology , alpha-MSH/pharmacology , Animals , Drug Interactions , Hypothalamus/metabolism , Male , Models, Biological , Neuroimmunomodulation/physiology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology
9.
Proc Natl Acad Sci U S A ; 91(2): 782-6, 1994 Jan 18.
Article in English | MEDLINE | ID: mdl-7904758

ABSTRACT

Previous work has shown that growth hormone-releasing factor (GRF) stimulates cGMP production and somatostatin [somatotropin (growth hormone)-release-inhibiting factor, SRIF] release without altering cAMP accumulation by fragments of median eminence incubated in vitro. Therefore, this study was undertaken to evaluate the effect of GRF and cGMP on SRIF mRNA and SRIF release in the periventricular nuclei of male rats in vitro. SRIF mRNA levels were determined in explants of periventricular nuclei incubated for 6 hr in Waymouth's medium in the presence of various substances. Steady-state levels of SRIF mRNA were measured by an S1 nuclease protection assay using a 32P-labeled rat SRIF RNA probe. SRIF release and cGMP formation were measured at 30 min and 6 hr by RIA. SRIF mRNA levels and SRIF release were significantly (P < 0.025) increased (approximately 2-fold) by 1 microM dibutyryl cGMP, whereas sodium butyrate had no effect. This augmentation was not influenced by cycloheximide, an inhibitor of protein synthesis. Sodium nitroprusside (10 microM), an activator of the guanylate cyclase pathway via its release of nitric oxide, augmented (P < 0.001) SRIF mRNA levels and significantly increased (P < 0.05) SRIF release. GRF (1 nM) increased SRIF mRNA (P < 0.001) and stimulated the release of SRIF at 30 min (P < 0.05) and 6 hr (P < 0.01). This stimulation was abolished by 10 microM NG-monomethyl-L-arginine (L-NMMA), a specific inhibitor of nitric oxide synthase, but not by NG-monomethyl-D-arginine (D-NMMA, the inactive isomer). GRF also increased cGMP formation. This effect was completely blocked by incubation with L-NMMA but not D-NMMA. These results indicate that GRF releases nitric oxide. The nitric oxide diffuses to the adjacent SRIF neurons, where it activates guanylate cyclase, leading to increased formation of cGMP. This cGMP increases SRIF mRNA and SRIF release in the periventricular nuclei of male rats.


Subject(s)
Gonadotropin-Releasing Hormone/pharmacology , RNA, Messenger/metabolism , Somatostatin/metabolism , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Cyclic GMP/biosynthesis , Cycloheximide/pharmacology , Dibutyryl Cyclic GMP/pharmacology , Enzyme Activation/drug effects , Guanylate Cyclase/metabolism , In Vitro Techniques , Male , Models, Biological , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Nitroprusside/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Somatostatin/genetics , omega-N-Methylarginine
10.
Brain Res ; 625(2): 213-8, 1993 Oct 22.
Article in English | MEDLINE | ID: mdl-7903898

ABSTRACT

Insulin-like growth factor I (IGF-I) has been shown to participate in feedback inhibition of growth hormone (GH) secretion at the level of both the pituitary and hypothalamus. Therefore, we tested the possible involvement of IGF-I on somatostatin (SRIF) and GH-releasing factor (GRF) release in median eminence (ME) fragments and periventricular nucleus (PeN) of male rats. The levels of SRIF messenger ribonucleic acid (mRNA) were also determined in PeN incubated in vitro with IGF-I. The ME's were incubated in Krebs-Ringer bicarbonate glucose buffer in the presence of various concentrations of IGF-I (10(-7) to 10(-11) M) for 30 min. SRIF and GRF released into the medium were quantitated by RIA. The release of SRIF and GRF from the ME's was stimulated significantly (P < 0.025 and P < 0.05, respectively) by 10(-9) M IGF-I. To determine whether the effect of IGF-I on SRIF release is mediated by GRF release in the ME, a specific GRF antibody (ab) (1:500) was used concomitantly with IGF-I (10(-9) M). The release of SRIF induced by IGF-I was blocked by the GRF ab (P < 0.001), but not by normal rabbit serum used at the same dilution. To determine the effect of IGF-I on the regulation of SRIF mRNA levels, SRIF mRNA was determined in PeN explants incubated in the presence of IGF-I (10(-8) to 10(-10) M) for 2 to 6 h. Levels of SRIF mRNA were determined by a S1 nuclease protection assay using a 32P-labelled rat SRIF riboprobe.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Growth Hormone-Releasing Hormone/physiology , Hypothalamus/metabolism , Insulin-Like Growth Factor I/physiology , RNA, Messenger/metabolism , Somatostatin/metabolism , Animals , Culture Techniques , Feedback , Male , Median Eminence/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Sprague-Dawley
11.
Brain Res ; 623(1): 89-94, 1993 Sep 24.
Article in English | MEDLINE | ID: mdl-8106120

ABSTRACT

Growth hormone (GH) suppresses its own secretion by stimulating somatostatin (SRIF) release. Thus, the possible regulation of GH-releasing factor (GRF) and SRIF release and SRIF messenger ribonucleic acid (mRNA) by GH was studied in the hypothalamus of male rats in vitro. The median eminences (ME's) were incubated in buffer containing 10(-7)-10(-11) M GH for 30 min. SRIF and GRF released into the medium were quantitated by RIA. The release of SRIF from ME fragments was significantly increased (P < 0.001) by 10(-9) M GH; however, 10(-9) M GH also inhibited (P < 0.01) GRF release from the ME. To determine the effect of GH on SRIF mRNA levels, periventricular nucleus (PeN) explants were cultured during 6 h in medium with 10(-7)-10(-11) M GH. Levels of SRIF mRNA (determined by an S1 nuclease protection assay) were significantly elevated in the presence of 10(-10)-10(-7) M GH. Likewise, 10(-9) M GH significantly stimulated SRIF release from PeN explants at 30 min and at 6 h. Surprisingly, 10(-9) M GH also significantly increased GRF release from the PeN explants at these times as well. This GRF was not responsible for the increased SRIF release or SRIF mRNA induced by GH since GRF antibody did not modify the GH-induced increases in SRIF release and mRNA levels. These results demonstrate a negative short-loop feedback of GH mediated at the ME by suppression of GRF and stimulation of SRIF release, whereas in the PeN GH increased both SRIF release and SRIF mRNA levels.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Growth Hormone/pharmacology , Paraventricular Hypothalamic Nucleus/metabolism , RNA, Messenger/metabolism , Somatostatin/biosynthesis , Somatostatin/metabolism , Animals , Dose-Response Relationship, Drug , Gene Expression/drug effects , Kinetics , Male , Organ Culture Techniques , Paraventricular Hypothalamic Nucleus/drug effects , Rats , Rats, Sprague-Dawley , Transcription, Genetic/drug effects
12.
Neuroendocrinology ; 58(2): 185-90, 1993 Aug.
Article in English | MEDLINE | ID: mdl-7903432

ABSTRACT

Interleukin-2 (IL-2), which plays a major role in the bidirectional intercellular communication between the neuroendocrine and immune systems, suppressed the release of GH from anterior pituitary halves at femtomolar concentrations. It is well established that the release of GH from the anterior pituitary is regulated by growth hormone releasing hormone (GRH) and growth hormone release-inhibiting hormone (somatostatin). Consequently, we studied the possible effect of IL-2 on the release of somatostatin and GRH from the mediobasal hypothalamus (MBH) in vitro. Single MBHs were incubated with fresh Krebs-Ringer bicarbonate (KRB) buffer alone or KRB containing different concentrations of IL-2 (10(-15)-10(-10) M) for 30 min. After collection of the media, the MBHs were incubated with KRB containing high potassium (high K+ = 56 mM) without IL-2 for a period of 30 min to study the effect of pretreatment with IL-2 on depolarization-induced somatostatin and GRH release. Experiments were also undertaken to study the effect of IL-2 in the presence of high K+ or IL-2 in the presence of DA (60 microM), a potent stimulator of somatostatin and GRH release. The minimal effective dose of IL-2 which significantly stimulated the release of somatostatin was 10(-14) M. Depolarization-induced release of somatostatin was reduced significantly by prior treatment with all the concentrations of IL-2 tested (10(-13)-10(-10) M).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Dopamine/pharmacology , Growth Hormone-Releasing Hormone/metabolism , Hypothalamus, Middle/drug effects , Interleukin-2/pharmacology , Pituitary Gland/drug effects , Somatostatin/metabolism , Animals , Hypothalamus, Middle/metabolism , Membrane Potentials/drug effects , Pituitary Gland/metabolism , Potassium/pharmacology
13.
Neuroendocrinology ; 56(6): 889-94, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1285434

ABSTRACT

Galanin has been reported to stimulate secretion of GH in humans and rats. Thus, to investigate whether the effect of galanin on GH release is the result of either a stimulation of GH-releasing factor (GRF) and/or an inhibition of somatostatin (SRIF) release, we have evaluated the action of galanin on the release of SRIF and GRF from median eminence (ME) fragments in vitro. The MEs from adult male rats were incubated in Krebs-Ringer bicarbonate-glucose buffer, pH 7.4, at 37 degrees C, in an atmosphere of 95% O2, 5% CO2 with constant shaking for 30 min. Medium was discarded and replaced by medium containing various concentrations of galanin (10(-10)-10(-7) M). Galanin stimulated SRIF and GRF release in a dose-related manner. This effect was significant at concentrations varying from 10(-8) to 10(-7) M. To determine the mechanism by which galanin stimulated SRIF and GRF release, MEs were incubated with pimozide (dopaminergic blocker), phentolamine (alpha-adrenergic blocker) or naloxone (opioid blocker), at concentrations of 10(-6) M, and the effect of galanin was then evaluated. Phentolamine and naloxone did not alter the stimulatory effect of galanin, but when galanin was tested with pimozide, the galanin-induced release of SRIF and GRF was blocked. To determine whether the effect of galanin is mediated through D-1 and/or D-2 dopamine receptors, selective antagonists of D-1 (SCH 23390) and D-2 receptors (domperidone) were used (10(-7) M) in the presence of galanin (10(-7) M).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Growth Hormone-Releasing Hormone/metabolism , Median Eminence/metabolism , Peptides/pharmacology , Somatostatin/metabolism , Animals , Domperidone/pharmacology , Dose-Response Relationship, Drug , Galanin , Growth Hormone-Releasing Hormone/drug effects , Male , Median Eminence/drug effects , Naloxone/pharmacology , Phentolamine/pharmacology , Pimozide/pharmacology , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Somatostatin/drug effects
14.
Neuroendocrinology ; 56(5): 674-9, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1362597

ABSTRACT

Thymosin alpha 1 (T alpha 1) is a well-characterized immunopotentiating polypeptide originally isolated from calf thymus. We have recently shown in vivo, probable hypothalamic effects of T alpha 1 to decrease the release of the pituitary hormones, TSH, PRL and ACTH from the pituitary gland. Therefore, in the present study we evaluated the effect of the peptide on the release of hypothalamic regulatory hormones: thyrotropin-releasing hormone (TRH) and corticotropin-releasing hormone (CRH), as well as somatostatin (SRIH), from medial basal hypothalamic (MBH) fragments incubated in vitro. After a preliminary time-course study indicated that a 30-min incubation period was optimal, it was used for all the other experiments. At the end of the incubation the tissue was still able to respond to a depolarizing K+ concentration for 15 min by a 4-fold increase of TRH concentration compared to control basal release during the preceding 30 min. T alpha 1 was shown to inhibit the release of TRH and CRH from MBH fragments incubated in vitro with a minimal effective dose (MED) of 10(-11) M. SRIH and CRH release was also inhibited but the MED for these peptides was 10(-9) M. The relative responsiveness to the action of T alpha 1 was TRH greater than CRH, which was greater than SRIH. This correlated with our previous in vivo results for pituitary hormone release, except in the case of SRIH since we previously did not detect any significant effect of the peptide on growth hormone release. Finally, we evaluated the possible involvement of other neurotransmitters in the effect of T alpha 1 on TRH release.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Hypothalamus, Middle/drug effects , Hypothalamus, Middle/metabolism , Somatostatin/metabolism , Thymosin/analogs & derivatives , Thyrotropin-Releasing Hormone/metabolism , Animals , Dopamine/pharmacology , In Vitro Techniques , Male , Metergoline/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin/pharmacology , Thymalfasin , Thymosin/pharmacology
15.
Proc Natl Acad Sci U S A ; 88(24): 11485-9, 1991 Dec 15.
Article in English | MEDLINE | ID: mdl-1684864

ABSTRACT

Because several peptides originally found in the pituitary as within the central nervous system have been localized in lymphoid tissues and because somatostatin (somatotropin-release-inhibiting hormone, SRIH) can act on cells of the immune system, we searched for this peptide in lymphoid organs. We demonstrated that SRIH mRNA exists in lymphoid tissue, albeit in smaller levels than in the periventricular region of the hypothalamus, the brain region that contains the highest level of this mRNA. SRIH mRNA was found in the spleen and thymus of male rats and in the spleen, thymus, and bursa of Fabricius of the chicken. Its localization in the bursa indicates that the peptide must be present in B lymphocytes since this is the site of origin of B lymphocytes in birds. The SRIH concentration in these lymphoid organs as determined by radioimmunoassay was greater in the thymus than in the spleen of the rat. These concentrations were 50 times less than those found in the periventricular region of the hypothalamus, the site of the perikarya of SRIH-containing neurons. In the chicken, as in the rat, the concentration of SRIH was greater in the thymus than in the spleen; it was present in the bursa of Fabricius, also in higher concentration than in the spleen. Fluorescence immunocytochemistry revealed the presence of SRIH-positive cells in clusters inside the white pulp and more dispersed within the red pulp of the spleen of both the rat and the chicken. The thymus from these species also contained SRIH-positive cells within the medulla and around the corticomedullary junction. In the chicken, there were large clusters of SRIH-positive cells in the medullary portion of each nodule of the bursa of Fabricius. Preabsorption of the primary antiserum or replacing this antiserum with normal rabbit serum verified the specificity of staining. Sequential immunostaining of the same sections from rat spleen using first SRIH antibody and subsequently a monoclonal antibody against a rat B-cell surface antigen revealed the presence of SRIH immunoreactivity in some, but not all, B cells. Other cell types in spleen not yet identified also stained positively with the SRIH antibody but were not reactive to monoclonal antibodies to rat Thy-1.1, a marker for all the thymic T lymphocytes. The possibility that SRIH is present in other populations of cells in the spleen cannot be ruled out. Sequential immunostaining of the same sections of rat thymus revealed the presence of SRIH immunoreactivity in a small population of T lymphocytes in the medulla, as revealed by the Thy-1.1 marker. The SRIH-positive cells were nonimmunoreactive when exposed to the B-cell marker; however, the possibility that SRIH is present in other cells was not investigated. Thus, our results indicate that SRIH is synthesized and stored in cells of the immune system. SRIH may be secreted from these cells to exert paracrine actions that alter the function of immune cells in spleen and thymus.


Subject(s)
Bursa of Fabricius/physiology , Somatostatin/genetics , Spleen/physiology , Thymus Gland/physiology , Transcription, Genetic , Animals , Chickens , DNA Probes , Male , Organ Specificity , Protein Precursors/genetics , RNA Probes , RNA, Messenger/analysis , RNA, Messenger/genetics , Radioimmunoassay , Rats , Rats, Inbred Strains , Somatostatin/biosynthesis , Somatostatin/metabolism , T-Lymphocytes/physiology
16.
Neuroendocrinology ; 54(5): 515-20, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1684229

ABSTRACT

In order to evaluate the release of somatostatin (SRIF) and growth hormone-releasing factor (GRF) into the pituitary gland in response to ether stress, a push-pull perifusion (PPP) technique has been used in freely moving rats. Push-pull cannulae (PPC) were implanted into the anterior pituitary (AP) glands of male rats. After a 7-day recovery period the rats were fitted with indwelling jugular catheters. The next day the animals were subjected to PPP of the AP during 1 h followed by ether stress (2 min) and another hour of perifusion. The perifusion flow was 20 microliters/min and 10-min fractions were collected and assayed for SRIF and GRF by RIA. Plasma growth hormone (GH) levels were assayed every 10 min. At the end of the experiments, the accuracy of PPC tip placements was ascertained with a dissecting microscope. Under basal conditions there were 2.9 pulses/h of SRIF with an amplitude of 12.76 +/- 0.46 pg. The output of SRIF and GRF in the 10-min period beginning with application of ether was increased 2-fold (p less than 0.005 and p less than 0.01, respectively). Interestingly, the increased release of SRIF continued for an additional 10 min, whereas GRF output decreased and was almost undetectable. The release of both GRF and SRIF had returned to basal values 20-30 min after stress. Mean plasma GH levels were significantly lowered 10 min after stress. Each of the 9 animals showed a restoration of pulsatile GH release to basal levels within 20-30 min after stress. Our findings provide compelling evidence that SRIF plays a prominent role in stress-induced inhibition of GH release in the rat by blocking the response to the transient elevation of GRF and continuing to suppress GH release for 20 min.


Subject(s)
Ether , Growth Hormone-Releasing Hormone/physiology , Growth Hormone/metabolism , Somatostatin/physiology , Stress, Physiological/physiopathology , Animals , Growth Hormone-Releasing Hormone/metabolism , Kinetics , Male , Periodicity , Rats , Rats, Inbred Strains , Somatostatin/metabolism , Stress, Physiological/chemically induced
17.
Endocrinology ; 129(2): 931-8, 1991 Aug.
Article in English | MEDLINE | ID: mdl-1677332

ABSTRACT

Neuropeptide-Y (NPY) and glucocorticoid receptors are coexpressed in many neurons in the brain. We addressed the question: Do glucocorticoids regulate the accumulation and/or secretion of immunoreactive (IR) NPY by fetal rat brain cells in culture, and if so, is the effect developmental stage dependent? Aggregates, formed from dissociated cells obtained from the hypothalamus-olfactory tubercle of 17-day-old fetuses, were cultured in serum-free medium for 23 days. On day 23, the aggregate NPY content was 6 ng/flask, and secretion (last 2 days) was approximately 12 ng/24 h. Exposure to dexamethasone (Dex; 20 nM) between days 0-23 led to a 1.9-fold increase in the aggregate content of NPY, whereas NPY secretion was not altered. When Dex exposure was limited to days 12-23, 16-23, 19-23, or 21-23, only a 12- to 23-day exposure induced NPY accumulation, and it was as effective as a 0- to 23-day exposure. The Dex-induced increase in NPY content was evident after a lag period of 4 days or more. When Dex exposure occurred on days 0-12, the aggregate NPY content on day 12 or 23 was not altered. None of these treatments altered the aggregate/medium content of immunoreactive somatostatin (SRIF) or the response to a 48-h exposure to forskolin (10 microM). Dex induction of NPY accumulation was a saturable function of the Dex concentration (maximal at 20 nM), and it was completely inhibited by RU486, a glucocorticoid/progesterone receptor antagonist; neither progesterone, 17 beta-estradiol, nor testosterone altered aggregate/medium NPY contents. Protein/DNA contents of the aggregates were either unaffected or slightly reduced by Dex. Thus, 1) Dex stimulates the accumulation of immunoreactive NPY, but not SRIF, by cultured fetal brain cells; 2) this effect requires a continuous 8-12 days of exposure to Dex during a late developmental stage in culture; 3) Dex does not potentiate or attenuate forskolin action on the NPY neuron; and 4) Dex action appears to be mediated by the glucocorticoid receptor. These results are consistent with glucocorticoid induction of production and/or decreased intracellular degradation of NPY, and with glucocorticoids regulating the NPY neuron in the perinatal brain in a developmental age-dependent manner.


Subject(s)
Brain/embryology , Cell Aggregation/drug effects , Dexamethasone/pharmacology , Neurons/cytology , Neuropeptide Y/metabolism , Animals , Brain/cytology , Cells, Cultured , Colforsin/pharmacology , DNA/metabolism , Female , Kinetics , Mifepristone/pharmacology , Neurons/drug effects , Proteins/metabolism , Rats , Rats, Inbred Strains , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/physiology , Somatostatin/metabolism
18.
Life Sci ; 49(1): 67-74, 1991.
Article in English | MEDLINE | ID: mdl-1675756

ABSTRACT

The effects of rat growth hormone releasing factor (rGRF) on somatostatin (SRIF) secretion, cyclic nucleotide production and phosphatidylinositol metabolism were investigated in the median eminence (ME), using an in vitro system. Medium was discarded and replaced by medium containing various concentrations of rGRF or rGRF plus epinephrine (E, 6 x 10(-7) M). rGRF had no effect on basal or E-stimulated release of cAMP. In the same experiments rGRF markedly stimulated SRIF release. These results suggested that cAMP is not involved in the stimulatory effect of GRF on SRIF release. However, GRF significantly stimulated release of both SRIF and cGMP in a dose-related manner. Maximal stimulation was observed at 10(-10) M GRF (p less than 0.005) which also produces maximal SRIF release. 2'0-monobutyrylguanosine 3'5' cyclic phosphate (mbcGMP, 10(-11) to 10(-10) M) stimulated SRIF release from ME fragments (p less than 0.001 at 10(-10) M) whereas the control, sodium butyrate (10(-6) M), had no effect. GRF caused significant elevation of 30.6% in the concentration of labelled inositol phosphates [( 3H]-IPs) in the ME. These data indicate that GRF stimulation of SRIF release is accompanied by increased cGMP production and phosphatidyl-inositol (PI) metabolism but does not alter cAMP production. Because mbcGMP can directly stimulate SRIF release, we suggest that GRF causes a receptor-mediated increase in the metabolism of phosphatidylinositol and cGMP formation. These actions therefore may be among the early metabolic events in the mechanism of GRF-stimulated SRIF release from the ME.


Subject(s)
Cyclic GMP/biosynthesis , Growth Hormone-Releasing Hormone/metabolism , Median Eminence/metabolism , Phosphatidylinositols/metabolism , Animals , Cyclic AMP/biosynthesis , Epinephrine/metabolism , Inositol Phosphates/metabolism , Male , Radioimmunoassay , Rats , Rats, Inbred Strains , Second Messenger Systems , Somatostatin/metabolism
19.
Proc Natl Acad Sci U S A ; 87(24): 10063-6, 1990 Dec.
Article in English | MEDLINE | ID: mdl-1979873

ABSTRACT

Previous in vivo studies have shown that delta 9-tetrahydrocannabinol (THC), the principal active ingredient in marijuana, can suppress both luteinizing hormone (LH) and growth hormone (GH) secretion after its injection into the third ventricle of conscious male rats. The present studies were designed to determine the mechanism of these effects. Various doses of THC were incubated with either stalk median eminence fragments (MEs) or mediobasal hypothalamic (MBH) fragments in vitro. Although THC (10 nM) did not alter basal release of LH-releasing hormone (LHRH) from MEs in vitro, it completely blocked the stimulatory action of dopamine or norepinephrine on LHRH release. The effective doses to block LHRH release were associated with a blockade of synthesis and release of prostaglandin E2 (PGE2) from MBH in vitro. In contrast to the suppressive effect of THC on LHRH release, somatostatin release from MEs was enhanced in a dose-related manner with a minimal effective dose of 1 nM. Since PGE2 suppresses somatostatin release, this enhancement may also be related to the suppressive effect of THC on PGE2 synthesis and release. We speculate that these actions are mediated by the recently discovered THC receptors in the tissue. The results indicate that the suppressive effect of THC on LH release is mediated by a blockade of LHRH release, whereas the suppressive effect of the compound on growth hormone release is mediated, at least in part, by a stimulation of somatostatin release.


Subject(s)
Dinoprostone/metabolism , Dronabinol/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Median Eminence/metabolism , Somatostatin/metabolism , Animals , Arachidonic Acid , Arachidonic Acids/metabolism , Dinoprost/metabolism , Dinoprostone/antagonists & inhibitors , Dopamine/pharmacology , Dose-Response Relationship, Drug , In Vitro Techniques , Kinetics , Male , Median Eminence/drug effects , Norepinephrine/pharmacology , Rats , Rats, Inbred Strains
20.
Neuroendocrinology ; 52(3): 238-42, 1990 Sep.
Article in English | MEDLINE | ID: mdl-1977095

ABSTRACT

The possible involvement of arachidonic acid (AA) release in growth-hormone-releasing factor (GRF)-induced somatostatin (SRIF) release from the median eminence (ME) of the hypothalamus was evaluated in adult male rats using an in vitro incubation system. The MEs were preincubated with [14C]-AA, then washed and incubated with vehicle or test agents, and the release of SRIF and [14C]-AA into the medium was measured. In the experiments designed only to determine SRIF release, the MEs were first preincubated for 30 min. The medium was then discarded and replaced with fresh buffer or test substances and incubated for 10, 20 and/or 30 min. GRF (10(-10) M) stimulated both AA and SRIF release significantly within 20 min, with maximum release occurring at 30 min. The stimulatory effect of GRF on AA release was coincident with the release of SRIF. A phospholipase A2 inhibitor (10(-6) M, quinacrine) completely abolished the stimulatory effect of GRF on both AA and SRIF release. The release of SRIF induced by GRF was also inhibited by both indomethacin (10(-6) M, a cyclooxygenase inhibitor) and metyrapone (10(-6) M, a cytochrome P-450 inhibitor). On the other hand, nordihydroguaiaretic acid (10(-6) M, a lipoxygenase inhibitor) had no effect on GRF-evoked SRIF release. The data presented here suggest that an important GRF-mediated event leading to SRIF secretion is an elevated release of AA from ME fragments in vitro. In conclusion, our data are suggestive that the stimulatory effect of GRF on SRIF release is due, in part, to the release and subsequent metabolism of AA to one or more metabolites.


Subject(s)
Arachidonic Acids/metabolism , Growth Hormone-Releasing Hormone/pharmacology , Median Eminence/metabolism , Somatostatin/metabolism , Animals , Arachidonic Acid , Cyclooxygenase Inhibitors , Cytochrome P-450 Enzyme Inhibitors , Indomethacin/pharmacology , Kinetics , Lipoxygenase Inhibitors , Male , Masoprocol/pharmacology , Median Eminence/drug effects , Metyrapone/pharmacology , Phospholipases A/antagonists & inhibitors , Phospholipases A2 , Quinacrine/pharmacology , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...