Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Neuroimmunomodulation ; 14(3-4): 188-92, 2007.
Article in English | MEDLINE | ID: mdl-18073513

ABSTRACT

During marijuana and alcohol consumption as well as during inflammation the reproductive axis is inhibited, mainly through the inhibition of luteinizing hormone-releasing hormone release. In male rats, this inhibitory effect is mediated, at least in part, by the activation of hypothalamic cannabinoid type 1 receptors (CB1). During inflammation, this activation of the endocannabinoid system seems to be mediated by an increase in TNF-alpha production followed by anandamide augmentations, similarly the effect of intragastric administration of ethanol (3 g/kg) seems to be due to an increase in anandamide. On the other hand, a number of different actions mediated by the endocannabinoid system in various organs and tissues have been described. Both cannabinoid receptors, CB1 and CB2, are localized in the submandibular gland where they mediate the inhibitory effect of intrasubmandibular injections of the endocannabinoid anandamide (6 x 10(-5)M) on salivary secretion. Lipopolysaccharide (5 mg/kg/3 h) injected intraperitoneally and ethanol (3 g/kg/1 h) injected intragastrically inhibited the salivary secretion induced by the sialogogue metacholine; this inhibitory effect was blocked by CB1 and/or CB2 receptor antagonists. Similar to the hypothalamus, these effects seem to be mediated by increased anandamide. In summary, similar mechanisms mediate the inhibitory actions of endocannabinoids and cannabinoids in both hypothalamus and submandibular gland during drug consumption and inflammation.


Subject(s)
Cannabinoid Receptor Modulators/physiology , Endocannabinoids , Hypothalamo-Hypophyseal System/drug effects , Inflammation/drug therapy , Salivary Glands/drug effects , Tumor Necrosis Factor-alpha/physiology , Animals , Arachidonic Acids/metabolism , Cannabinoids/pharmacology , Ethanol/pharmacology , Humans , Hypothalamo-Hypophyseal System/immunology , Hypothalamo-Hypophyseal System/metabolism , Inflammation/immunology , Polyunsaturated Alkamides/metabolism , Receptors, Cannabinoid/drug effects , Receptors, Cannabinoid/immunology , Salivary Glands/immunology , Salivary Glands/metabolism
2.
Proc Natl Acad Sci U S A ; 104(17): 7205-10, 2007 Apr 24.
Article in English | MEDLINE | ID: mdl-17409187

ABSTRACT

The systemic inflammatory response syndrome (SIRS) is a life-threatening medical condition characterized by a severe and generalized inflammatory state that can lead to multiple organ failure and shock. The CNS regulates many features of SIRS such as fever, cardiovascular, and neuroendocrine responses. Central and systemic manifestations of SIRS can be induced by LPS or IL-1beta administration. The crucial role of IL-1beta in inflammation has been further highlighted by studies of mice lacking caspase 1 (casp1, also known as IL-1beta convertase), a protease that cleaves pro-IL-1beta into mature IL-1beta. Indeed, casp1 knockout (casp1(-/-)) mice survive lethal doses of LPS. The key role of IL-1beta in sickness behavior and its de novo expression in the CNS during inflammation led us to test the hypothesis that IL-1beta plays a major role modulating the brain transcriptome during SIRS. We show a gene-environment effect caused by LPS administration in casp1(-/-) mice. During SIRS, the expression of several genes, such as chemokines, GTPases, the metalloprotease ADAMTS1, IL-1RA, the inducible nitric oxide synthase, and cyclooxygenase-2, was differentially increased in casp1(-/-) mice. Our findings may contribute to the understanding of the molecular changes that take place within the CNS during sepsis and SIRS and the development of new therapies for these serious conditions. Our results indicate that those genes may also play a role in several neuropsychiatric conditions in which inflammation has been implicated and indicate that casp1 might be a potential therapeutic target for such disorders.


Subject(s)
Brain/metabolism , Brain/pathology , Caspase 1/deficiency , Transcription, Genetic , Animals , Brain/drug effects , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation/drug effects , Inflammation , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Models, Biological , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Spleen/drug effects , Spleen/metabolism , Systemic Inflammatory Response Syndrome , Transcription, Genetic/drug effects
3.
Proc Natl Acad Sci U S A ; 104(6): 2007-12, 2007 Feb 06.
Article in English | MEDLINE | ID: mdl-17264205

ABSTRACT

The adrenal gland comprises two endocrine tissues of distinct origin, the catecholamine-producing medulla and the steroid-producing cortex. The inner adrenocortical zone, which is in direct contact with the adrenomedullary chromaffin cells, produces dehydroepiandrostendione (DHEA) and DHEA sulfate (DHEAS). These two androgens exhibit potential effects on neurogenesis, neuronal survival, and neuronal stem cell proliferation. Unlike the closely related sympathetic neurons, chromaffin cells are able to proliferate throughout life. The aim of this study was to investigate the effect of DHEA and DHEAS on proliferation of bovine chromaffin cells from young and adult animals. We demonstrated that graded concentrations of leukemia inhibitory factor induced proliferation of chromaffin cells from young animals, whereas EGF had no effect. On the contrary, EGF increased the cell proliferation in cells from adult animals, whereas leukemia inhibitory factor was inactive. In both cases, DHEA decreased the proliferative effect induced by the growth factors. Surprisingly, DHEAS enhanced, in a dose-dependent-manner, the effect of growth factors on proliferation in cells from adult animals but not from young animals. Flutamide, ICI 182,780, and RU 486 had no effect on the action of DHEA or DHEAS on chromaffin cell proliferation. These data show that DHEA and its sulfated form, DHEAS, differentially regulate growth-factor-induced proliferation of bovine chromaffin cells. In addition, the sensitivity of chromaffin cells to different growth factors is age-dependent. Furthermore, these two androgens may act through a receptor other than the classical steroid receptors.


Subject(s)
Aging/physiology , Cell Proliferation/drug effects , Chromaffin Cells/cytology , Chromaffin Cells/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Dehydroepiandrosterone/physiology , Growth Substances/physiology , Animals , Cattle , Cells, Cultured , Chromaffin Cells/metabolism , Female , Male
4.
Toxicol Sci ; 97(1): 75-80, 2007 May.
Article in English | MEDLINE | ID: mdl-17290048

ABSTRACT

Recently studies have demonstrated that low doses of (Mn(+2)) in the form of manganese chloride can stimulate specific puberty-related hormones and advance signs of pubertal development in immature female and male rats. In the present study, we used an in vitro system to evaluate the ability of 0, 50, 250, and 500 microM doses of Mn(+2) to stimulate luteinizing hormone-releasing hormone (LHRH) secretion and to assess the hypothalamic mechanism of this action in adult male Sprague-Dawley rats. We demonstrated that Mn(+2) at 500 microM, but not the lower doses, increased LHRH release, nitric oxide (NO) synthase (NOS) activity, and the content of cyclic cGMP in the medial basal hypothalamus. Inhibition of NOS with a competitive inhibitor (Nomega-nitro-L-arginine methyl ester hydrochloride) prevented the Mn-induced increase in LHRH release. Additionally, methylene blue and KT5823, specific inhibitors of guanylyl cyclase and protein kinase G (PKG), respectively, also blocked the stimulatory effect of Mn(+2) on LHRH release. These in vitro studies demonstrated that the hypothalamic mechanism of Mn(+2) action in adult males is by activation of the NOS/NO system, resulting in increases in cGMP and PKG and thus the secretion of LHRH from the nerve terminals. These results indicate Mn(+2) can cause LHRH release in adult males, and this action is discussed in relation to age, gender, as well as mechanistic and functional differences between adult and immature animals.


Subject(s)
Chlorides/toxicity , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/drug effects , Signal Transduction/drug effects , Age Factors , Animals , Carbazoles/pharmacology , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Female , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Hemoglobins/metabolism , Hypothalamus/metabolism , In Vitro Techniques , Indoles/pharmacology , Male , Manganese Compounds , Methylene Blue/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Sex Factors
5.
Ann N Y Acad Sci ; 1088: 1-11, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17192552

ABSTRACT

This review documents the remarkable progress over the last 50 years of our knowledge of the control of anterior pituitary hormone release and synthesis by a family of peptidic releasing and inhibiting hormones, synthesized in hypothalamic neurons and released into the hypophysial portal vessels. These vessels transport them to the anterior pituitary, where they stimulate release and synthesis of pituitary hormones or inhibit these processes. In general, there are at least two hypothalamic hormones for each pituitary hormone-vasopressin and corticotrophin-releasing hormone (CRH) for adrenocorticotropin hormone (ACTH) and growth hormone-releasing hormone (GHRH) and growth hormone-inhibiting hormone (GIH) for growth hormone (GH). Some of these hormones have extrapituitary action: for example, luteinizing hormone-releasing hormone (LHRH) stimulates mating behavior. High doses of LHRH have an inhibitory action on the growth of prostate cancer. Proinflammatory and anti-inflammatory cytokines act not only in the brain, but also on the pituitary and peripheral tissues. All of these transmitters are controlled by neuronal transmitters. We anticipate further rapid progress and clinical application of these transmitters and the discovery of new ones.


Subject(s)
Endocrinology/trends , Neuroimmunomodulation/physiology , Pituitary Hormone-Releasing Hormones/metabolism , Pituitary Hormones, Anterior/metabolism , Animals , Humans , Pituitary Hormone-Releasing Hormones/immunology , Pituitary Hormone-Releasing Hormones/pharmacology , Pituitary Hormones, Anterior/immunology
6.
Ann N Y Acad Sci ; 1088: 238-50, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17192570

ABSTRACT

It is known that Delta(9)-tetrahydrocannabinol (THC), the major active ingredient of marijuana, can suppress reproductive function. Also, we reported previously that the endocannabinoid, anandamide (AEA), inhibited gonadotropin-releasing hormone (LHRH) release from medial basal hypothalamus (MBH) of male rats incubated in vitro as well as reduced plasma LH levels after i.c.v. AEA injections into the cerebral lateral ventricle. On the other hand, it is known that during endotoxemia the hypothalamic gonadotropin axis is inhibited. Therefore, the aim of the present study was to determine whether the effect of TNF-alpha, a proinflammatory cytokine induced by lipopolysaccharide (LPS) that inhibits LHRH release, is mediated by the activation of the endocannabinoid system. The intraperitoneal injection of LPS (5 mg/kg) as well as the i.c.v. injection of tumor necrosis factor-alpha (TNF-alpha) (100 ng/rat) increased significantly the AEA synthesis measured ex vivo in MBHs removed 3 h after the treatments. To examine the possibility that TNF-alpha also acted by increasing the synthesis of AEA that was released and activated the CB1-r followed by inhibition of LHRH release, we measured the effect of TNF-alpha on the AEA synthase activity in MBHs incubated in vitro. As expected, we found that TNF-alpha (2.9 x 10(-9) M) increased the AEA synthesis. Second, we showed that TNF-alpha reduced significantly the forskolin-stimulated LHRH release and that the CB1-r antagonist AM251 (10(-5) M) blocked that inhibition, supporting the hypothesis that TNF-alpha inhibits LHRH release, acting at least in part by activating the endocannabinoid system. Therefore, our data demonstrate a key role for the endocannabinoid system in the response of the reproductive system to inflammatory signals.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Amidohydrolases/metabolism , Animals , Colforsin/pharmacology , Cyclic AMP/metabolism , Enzyme Activation/drug effects , Hypothalamus/drug effects , Hypothalamus/immunology , Injections, Intraperitoneal , Injections, Intraventricular , Lipopolysaccharides/pharmacology , Male , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
Neuroimmunomodulation ; 13(3): 152-9, 2006.
Article in English | MEDLINE | ID: mdl-17119344

ABSTRACT

OBJECTIVES: Since very little is known about neuroendocrine changes that occur in portal-systemic hepatic encephalopathy, we studied plasma prolactin (PRL) levels and the involvement of hyperammonemia, nitric oxide (NO) and dopaminergic and adrenergic systems in the control of this hormone secretion in a male rat model of prehepatic portal hypertension (PH). METHODS: We conducted in vivo studies to determine plasma ammonia and PRL levels. Dopamine (DA), dihydroxyphenylacetic acid (DOPAC), epinephrine and norepinephrine content in medial basal hypothalamus (MBH) and anterior pituitary (AP) were measured. In addition, NO synthase (NOS) activity and protein expression were evaluated in APs. In in vitro studies, the APs from intact rats were incubated with different doses of ammonia and PRL secretion was determined. In ex vivo studies, the APs from normal and PH rats were incubated in the presence of ammonia and/or a NOS inhibitor, NG-nitro-L-arginine-methyl ester (L-NAME) and PRL secretion was determined. RESULTS: PH rats had a significant increase in plasma ammonia levels (p < 0.001) and a decrease in plasma PRL levels (p < 0.05). Neither DA nor DOPAC content or DOPAC/DA ratios were modified in both MBH and APs; however, we observed a significant increase in norepinephrine content in both MBH and AP (p < 0.001 and p < 0.05, respectively) and a significant increase in epinephrine in APs (p < 0.001). Moreover, PH produced an increase in NOS activity (p < 0.01) and NOS protein expression (p < 0.0001) in APs. The ammonia (100 microM) significantly reduced PRL secretion from APs in vitro (p < 0.05). The presence of L-NAME, an inhibitor of NOS, abrogated the inhibitory effect of ammonia on PRL secretion from APs from control and PH rats. CONCLUSIONS: We found that plasma PRL levels were decreased in PH rats probably due to the high ammonia levels. The central noradrenergic system could also mediate this decrease. Also, the increase in NOS activity and/or content in AP induced NO production that directly inhibited PRL secretion from the AP, without the participation of the dopaminergic system.


Subject(s)
Ammonia/blood , Hypertension, Portal/blood , Nitric Oxide/metabolism , Prolactin/blood , Animals , Blotting, Western , Brain/metabolism , Catecholamines/metabolism , Hypertension, Portal/metabolism , Male , Nitric Oxide Synthase/metabolism , Portacaval Shunt, Surgical , Rats , Rats, Wistar
8.
Exp Biol Med (Maywood) ; 231(8): 1421-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16946411

ABSTRACT

It is known that marijuana use decreases saliva secretion. Therefore, we hypothesized that cannabinoid receptors (CBs) are located in salivary glands to mediate that effect. In these experiments, we used the submandibular gland (SMG) of male rats, which is one of the major salivary glands. Mammalian tissues contain at least two types of CBs, CB1 and CB2, mainly located in the nervous system and peripheral tissues, respectively. Both receptors are coupled to Gi protein and respond by inhibiting the activity of adenylyl cyclase. We demonstrated that both CB1 and CB2 are present in the SMG, each showing specific localizations. The best-known endocannabinoid is anandamide (AEA), which binds with high affinity to CB1 and CB2. We showed that AEA markedly reduced forskolin-induced increase of cAMP content in vitro. This effect was blocked by AM251 and AM630 (CB1 and CB2 antagonists, respectively), indicating that both receptors are implicated in SMG physiology. In addition, we showed that AEA injected intraglandularly to anesthetized rats inhibited norepinephrine (NE)- and methacholine (MC)-stimulated saliva secretion in vivo and that both AM251 or AM630 prevented the inhibitory action of AEA. Also, the intraglandular injection of AM251 increased saliva secretion induced by lower doses of NE or MC. This increase was synergized after coinjection with AM630. Therefore, we concluded that AEA decreases saliva secretion in the SMG acting through CB1 and CB2 receptors.


Subject(s)
Arachidonic Acids/administration & dosage , Cannabinoid Receptor Modulators/administration & dosage , Receptors, Cannabinoid/drug effects , Receptors, Cannabinoid/metabolism , Saliva/metabolism , Submandibular Gland/metabolism , Animals , Colforsin/pharmacology , Cyclic AMP/metabolism , Endocannabinoids , Immunohistochemistry , Indoles/pharmacology , Male , Methacholine Chloride/pharmacology , Norepinephrine/pharmacology , Parasympathomimetics/pharmacology , Piperidines/pharmacology , Polyunsaturated Alkamides , Pyrazoles/pharmacology , Rats , Rats, Wistar , Saliva/drug effects , Sympathomimetics/pharmacology
9.
Proc Natl Acad Sci U S A ; 103(41): 15124-9, 2006 Oct 10.
Article in English | MEDLINE | ID: mdl-17008408

ABSTRACT

Cyclic nucleotide phosphodiesterases (PDEs) constitute a family of enzymes that degrade cAMP and cGMP. Intracellular cyclic nucleotide levels increase in response to extracellular stimulation by hormones, neurotransmitters, or growth factors and are down-regulated through hydrolysis catalyzed by PDEs, which are therefore candidate therapeutic targets. cAMP is a second messenger implicated in learning, memory, and mood, and cGMP modulates nervous system processes that are controlled by the nitric oxide (NO)/cGMP pathway. To investigate an association between genes encoding PDEs and susceptibility to major depressive disorder (MDD), we genotyped SNPs in 21 genes of this superfamily in 284 depressed Mexican Americans who participated in a prospective, double-blind, pharmacogenetic study of antidepressant response, and 331 matched controls. Polymorphisms in PDE9A and PDE11A were found to be associated with the diagnosis of MDD. Our data are also suggestive of the association between SNPs in other PDE genes and MDD. Remission on antidepressants was significantly associated with polymorphisms in PDE1A and PDE11A. Thus, we found significant associations with both the diagnosis of MDD and remission in response to antidepressants with SNPs in the PDE11A gene. We show here that PDE11A haplotype GAACC is significantly associated with MDD. We conclude that PDE11A has a role in the pathophysiology of MDD. This study identifies a potential CNS role for the PDE11 family. The hypothesis that drugs affecting PDE function, particularly cGMP-related PDEs, represent a treatment strategy for major depression should therefore be tested.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , Genetic Predisposition to Disease , Phosphoric Diester Hydrolases/genetics , 3',5'-Cyclic-GMP Phosphodiesterases , Adult , Aged , Cyclic Nucleotide Phosphodiesterases, Type 1 , Depressive Disorder, Major/diagnosis , Depressive Disorder, Major/physiopathology , Desipramine/therapeutic use , Double-Blind Method , Female , Fluoxetine/therapeutic use , Humans , Male , Mexican Americans , Middle Aged , Polymorphism, Single Nucleotide , Prospective Studies , Single-Blind Method
10.
Neuroimmunomodulation ; 13(1): 19-27, 2006.
Article in English | MEDLINE | ID: mdl-16691037

ABSTRACT

OBJECTIVE: In the present work, we evaluated the effect of exposing the submandibular glands (SMG) to radiation, studying different functional parameters such as salivary secretion, nitric oxide (NO) production, reactive oxygen species formation, prostaglandin (PGE) content and apoptosis. METHODS: We irradiated rats in the head and neck region with a single dose of gamma-ray radiation of 15 Gy. Two hours after radiation, we measured norepinephrine-induced salivary secretion. After that, the SMG were dissected, and in this tissue, we measured the activity of NO synthase (NOS), the PGE content, the amount of reactive oxygen species, apoptotic cells and mitochondrial inducible NOS (iNOS) expression. RESULTS: We found that radiation decreased salivary secretion when 10 and 30 microg/kg of norepinephrine was administered via the right femoral vein. We observed that iNOS activity was reduced and PGE content increased after radiation in SMG, indicating that NO and PGEs may participate in salivary secretion. The expression of mitochondrial NOS was increased after radiation leading to the formation of large amounts of NO that acts as a proapoptotic signal. In fact, we observed an augmentation in apoptotic cells. In this study, we also observed an increase in lipid peroxidation induced by radiation that may contribute to tissue damage. CONCLUSIONS: Our results indicate that radiation induced a decrease in salivary secretion and SMG iNOS activity, meanwhile the PGE content, the lipid peroxidation and apoptosis increased in the tissue. These modifications decrease salivary secretion.


Subject(s)
Nitric Oxide/radiation effects , Prostaglandins/radiation effects , Radiotherapy/adverse effects , Submandibular Gland/metabolism , Submandibular Gland/radiation effects , Xerostomia/physiopathology , Animals , Apoptosis/physiology , Apoptosis/radiation effects , Disease Models, Animal , Down-Regulation/physiology , Down-Regulation/radiation effects , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Female , Head and Neck Neoplasms/radiotherapy , Lipid Peroxidation/physiology , Lipid Peroxidation/radiation effects , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type II/radiation effects , Oxidative Stress/physiology , Oxidative Stress/radiation effects , Prostaglandins/metabolism , Rats , Saliva/metabolism , Submandibular Gland/physiopathology , Xerostomia/etiology , Xerostomia/metabolism
11.
Proc Natl Acad Sci U S A ; 103(16): 6392-7, 2006 Apr 18.
Article in English | MEDLINE | ID: mdl-16606831

ABSTRACT

Sepsis and septic shock are leading killers in the noncoronary intensive care unit, and they remain worldwide health concerns. The initial host defense against bacterial infections involves Toll-like receptors (TLRs), which detect and respond to microbial ligands. In addition, a coordinated response of the adrenal and immune systems is crucial for survival during severe inflammation. Previously, we demonstrated a link between the innate immune system and the endocrine stress response involving TLR-2. Like TLR-2, TLR-4 is also expressed in human and mouse adrenals. In the present work, by using a low dose of LPS to mimic systemic inflammatory response syndrome, we have revealed marked cellular alterations in adrenocortical tissue and an impaired adrenal corticosterone response in TLR-4-/- mice. Our findings demonstrate that TLR-4 is a key mediator in the crosstalks between the innate immune system and the endocrine stress response. Furthermore, TLR polymorphisms could contribute to the underlying mechanisms of impaired adrenal stress response in patients with bacterial sepsis.


Subject(s)
Adrenal Glands/physiology , Adrenocorticotropic Hormone/blood , Corticosterone/blood , Cytokines/blood , Systemic Inflammatory Response Syndrome/immunology , Toll-Like Receptor 4/physiology , Adrenal Glands/chemistry , Adrenal Glands/cytology , Animals , Immunity, Innate/genetics , Lipopolysaccharides/immunology , Mice , Mice, Mutant Strains , NF-kappa B/metabolism , Systemic Inflammatory Response Syndrome/genetics , Toll-Like Receptor 4/analysis , Toll-Like Receptor 4/genetics
12.
Proc Natl Acad Sci U S A ; 102(23): 8303-8, 2005 Jun 07.
Article in English | MEDLINE | ID: mdl-15919819

ABSTRACT

The mortality of chronic heart failure (CHF) doubles either when CHF patients are depressed or when their plasma norepinephrine (NE) level exceeds those of controls by approximately 40%. We hypothesized that patients with major depression had centrally driven, sustained, stress-related, and treatment-reversible increases in plasma NE capable of increasing mortality in CHF patients with depression. We studied 23 controls and 22 medication-free patients with melancholic depression. In severely depressed patients before and after electroconvulsive therapy (ECT), we measured cerebrospinal fluid (CSF) NE, plasma NE, plasma epinephrine (EPI), and plasma cortisol hourly for 30 h. In mildly-to-moderately depressed melancholic patients, we assessed basal and stress-mediated arterial NE appearance. Severely depressed patients had significant increases in mean around-the-clock levels of CSF NE (P < 0.02), plasma NE (P < 0.02), plasma EPI (P < 0.02), and plasma cortisol (P < 0.02). CSF NE, plasma NE, and cortisol all rose together throughout the night and peaked in the morning. Each fell to control values after ECT. Mildly-to-moderately melancholic patients also had increased basal (P < 0.05) and stress-related (P < 0.03) arterial NE-appearance rates. Severely melancholic depressed, medication-free patients had around-the-clock increases in plasma NE levels capable of increasing mortality in CHF. Twenty-four-hour indices of central noradrenergic, adrenomedullary, and adrenocortical secretion were also elevated. Concurrent diurnal rhythms of these secretions could potentiate their cardiotoxicity. Even mildly-to-moderately depressed melancholic patients had clinically relevant increases in the arterial NE-appearance rate. These findings will not apply to all clinical subtypes of major depression.


Subject(s)
Arteries/physiology , Depression/blood , Depression/complications , Heart Diseases/blood , Heart Diseases/complications , Norepinephrine/blood , Adult , Blood Pressure , Chronic Disease , Depression/classification , Depression/physiopathology , Electroconvulsive Therapy , Female , Heart Diseases/physiopathology , Heart Rate , Humans , Hydrocortisone/blood , Male , Middle Aged , Norepinephrine/cerebrospinal fluid , Stress, Physiological/blood , Stress, Physiological/complications , Stress, Physiological/physiopathology , Time Factors
13.
Proc Natl Acad Sci U S A ; 102(17): 6213-8, 2005 Apr 26.
Article in English | MEDLINE | ID: mdl-15837925

ABSTRACT

The adrenal cortex is a major stress organ in mammals that reacts rapidly to a multitude of external and internal stressors. Adrenocorticotropin (ACTH) is the main stimulator of the adrenal cortex, activating corticosteroid synthesis and secretion. We evaluated the mechanism of action of ACTH on adrenals of male rats, preserving the architecture of the gland in vitro. We demonstrated that both sodium nitroprusside (NP), a nitric oxide (NO) donor, and ACTH stimulate corticosterone release. NO mediated the acute response to ACTH because Nomega-nitro-l-arginine methyl ester, a NO synthase inhibitor, and hemoglobin, a NO scavenger, blocked the stimulation of corticosterone release induced by ACTH. NP stimulated prostaglandin E release, which in turn stimulated corticosterone release from the adrenal. Additionally, indomethacin, which inhibits cyclooxygenase, and thereby, prostaglandin release, prevented corticosterone release from the adrenal induced by both NP and ACTH, demonstrating that prostaglandins mediate acute corticosterone release. Corticosterone content in adrenals after incubation with ACTH or NP was lower than in control glands, indicating that any de novo synthesis of corticosterone during this period was not sufficient to keep up with the release of the stored hormone. The release induced by ACTH or NP depleted the corticosterone content in the adrenal by approximately 40% compared with the content of glands incubated in buffer. The mechanism of rapid release is as follows: NO produced by NO synthase activation by ACTH activates cyclooxygenase, which generates PGE(2), which in turn releases corticosterone stored in microvesicles and other organelles.


Subject(s)
Adrenal Cortex/metabolism , Adrenocorticotropic Hormone/physiology , Corticosterone/metabolism , Dinoprostone/pharmacology , Nitric Oxide/physiology , Adrenal Cortex/drug effects , Adrenocorticotropic Hormone/metabolism , Animals , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitroprusside/pharmacology , Rats , Rats, Wistar
14.
Proc Natl Acad Sci U S A ; 101(47): 16695-700, 2004 Nov 23.
Article in English | MEDLINE | ID: mdl-15546996

ABSTRACT

Septicemia is one of the major health concerns worldwide, and rapid activation of adrenal steroid release is a key event in the organism's first line of defense during this form of severe illness. The family of Toll-like receptors (TLRs) is critical in the early immune response upon bacterial infection, and TLR polymorphisms are frequent in humans. Here, we demonstrate that TLR-2 deficiency in mice is associated with reduced plasma corticosterone levels and marked cellular alterations in adrenocortical tissue. TLR-2-deficient mice have an impaired adrenal corticosterone release after inflammatory stress induced by bacterial cell wall compounds. This defect appears to be mediated by a decrease in systemic and intraadrenal cytokine expression, including IL-1, tumor necrosis factor alpha, and IL-6. Our data demonstrate a link between the innate immune system and the endocrine stress response. The critical role of TLR-2 in adrenal glucocorticoid regulation needs to be considered in patients with inflammatory disease.


Subject(s)
Adrenal Cortex/physiopathology , Receptors, Cell Surface/deficiency , Adrenal Cortex/immunology , Adrenal Cortex/pathology , Adrenocorticotropic Hormone/metabolism , Animals , Corticosterone/blood , Corticosterone/metabolism , Cytokines/biosynthesis , Endotoxemia/immunology , Endotoxemia/pathology , Endotoxemia/physiopathology , Humans , Immunity, Innate , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , NF-kappa B/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Sepsis/immunology , Sepsis/pathology , Sepsis/physiopathology , Teichoic Acids/toxicity , Toll-Like Receptor 2
15.
Exp Biol Med (Maywood) ; 229(9): 926-34, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15388888

ABSTRACT

In the present investigation, 17beta-estradiol (E(2)) and tamoxifen, an antiestrogen, were evaluated for their effects on the release of ascorbic acid (AA) and luteinizing hormone-releasing hormone (LHRH). Medial basal hypothalami (MBH) from adult male rats were incubated with graded concentrations of E(2) (10 (-9) to 10(-6) M) or a combination of E(2) (10(-7) M) and tamoxifen (10(-7) and 10(-6) M ) in 0.5 ml of Krebs Ringer bicarbonate buffer for 1 hr. AA and LHRH in the incubation medium were measured by high-performance liquid chromatography and radioimmunoassay, respectively. E(2) significantly elevated both AA and LHRH release and the minimal effective dose was 10(-7) M. A combination of E(2) (10(-7) M) and tamoxifen (10(-6) M) totally blocked E(2)-induced AA and LHRH release. The stimulatory effect of E(2) was also suppressed in the presence of N(G)-monomethyl-L-arginine, a competitive inhibitor of nitric oxide synthase (NOS), illustrating that the release is mediated by nitric oxide (NO). To further characterize the role of NO, the tissues were incubated with E(2) or a combination of E(2) + (6 anilino-5, 8-quinolinedione) LY 83583 (10(-6) and 10(-5) M), an inhibitor of NOS. LY 83583 was effective in suppressing E(2)-induced AA and LHRH release, demonstrating that the effect was mediated by cyclic GMP. Incubation of the tissues with E(2) or a combination of E(2) + 1H-[1,2,4] oxadiazolo [4,3-a] quinoxalin-1-one (O.D.Q.) (10(-5) and 10(-4) M), a specific inhibitor of soluble guanylyl cyclase failed to alter AA release but significantly suppressed LHRH release. The role of a prostaglandin synthesis blocker in E(2)-induced AA and LHRH release was tested by incubating the tissues with E(2) or a combination of E(2) + indomethacin (1.8 x 10 (-7) or 1.8 x 10(-6) M). Indomethacin produced a significant decrease in E(2)-induced AA and LHRH release, suggesting that the release process required prostaglandins as an intracellular mediator. In conclusion, E(2) stimulated both AA and LHRH release and the effect was mediated by NO and prostaglandins.


Subject(s)
Ascorbic Acid/metabolism , Estradiol/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus, Middle/drug effects , Aminoquinolines/pharmacology , Animals , Cyclic GMP/physiology , Hypothalamus, Middle/metabolism , Indomethacin/pharmacology , Male , Nitric Oxide/physiology , Prostaglandins/physiology , Rats , Rats, Sprague-Dawley , Tamoxifen/pharmacology
16.
Proc Natl Acad Sci U S A ; 101(35): 13074-9, 2004 Aug 31.
Article in English | MEDLINE | ID: mdl-15316117

ABSTRACT

Previous studies demonstrated the presence of oxytocin (OT) and oxytocin receptors (OTRs) in the heart. The present work provides results supporting a potential role of OT in cardiomyogenesis. Here, we show a maximal OT and OTR protein level in the developing rat heart at day 21 of gestation and postnatal days 1-4, when cardiac myocytes are at a stage of intense hyperplasia. Between postnatal days 1 and 66, OT decreased linearly in all heart chambers (4.1- to 6.6-fold). Correspondingly, immunocytochemistry demonstrated that OTRs, which were eminent in postnatal cardiomyocytes, declined with age to low levels in adults. Interestingly, in coronary vasculature, OTRs developed in endothelial cells at postnatal days 12 and 22 and achieved a plateau in adult rats. These findings suggest that OT can be involved in developmental formation of the coronary vessels. In vivo, the OT/OTR system in the fetal heart was sensitive to the actions of retinoic acid (RA), recognized as a major cardiac morphogen. RA treatment produced a significant increase (2- to 3-fold) both in the OT concentration and in the OT mRNA levels. Ex vivo, an OT antagonist inhibited RA-mediated cardiomyocyte differentiation of P19 embryonic stem cells. The decline of cardiac OT expression from infancy to adulthood of the rat and changes in cell types expressing OTR indicate a dynamic regulation of the OT system in the heart rather than constitutive expression. The results support the hypothesis that RA induces cardiomyogenesis by activation of the cardiac OT system.


Subject(s)
Heart/embryology , Myocardium/metabolism , Oxytocin/metabolism , Animals , Cell Differentiation/physiology , Humans , Myocytes, Cardiac/physiology , Oxytocin/genetics , RNA, Messenger/metabolism , Rats , Tretinoin/metabolism
17.
Proc Natl Acad Sci U S A ; 101(32): 11891-6, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15280536

ABSTRACT

Because Delta-9-tetrahydrocannabinol (THC) inhibited luteinizing hormone-releasing hormone (LHRH) in male rats, we hypothesized that the endocannabinoid, anandamide (AEA), would act similarly. AEA microinjected intracerebroventricularly (i.c.v.) decreased plasma luteinizing hormone (LH) at 30 min in comparison to values in controls (P < 0.001). The cannabinoid receptor 1 (CB1-r)-specific antagonist, [N-(piperidin-1-yl)-1-(2,4-dichlorophenyl)-5-(4-chlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide] (AM251), produced a significant elevation in plasma LH (P < 0.01). AEA (10(-9) M) decreased LHRH release from medial basal hypothalami incubated in vitro. These results support the concept that endogenous AEA inhibits LHRH followed by decreased LH release in male rats. In ovariectomized (OVX) female rats, AEA i.c.v. also inhibited LH release, but in this case AM251 had an even greater inhibitory effect than AEA. In vitro, AEA had no effect on LHRH in OVX rats. It seems that endogenous AEA inhibits LHRH followed by decreased LH release in OVX rats but that AM251 has an inhibitory action in this case. In striking contrast, in OVX, estrogen-primed (OVX-E) rats, AEA i.c.v. instead of decreasing LH, increased its release. This effect was completely blocked by previous injection of AM251. When medial basal hypothalami of OVX-E rats were incubated, AEA increased LHRH release. The synthesized AEA was higher in OVX-E rats than in OVX and males, indicating that estrogen modifies endocannabinoid levels and effects. The results are interpreted to mean that sex steroids have profound effects to modify the response to AEA. It inhibits LHRH and consequently diminishes LH release in males and OVX females, but stimulates LHRH followed by increased LH release in OVX-E-primed rats.


Subject(s)
Arachidonic Acids/pharmacology , Estrogens/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Animals , Endocannabinoids , Female , Gonadal Steroid Hormones/pharmacology , Gonadotropin-Releasing Hormone/drug effects , Luteinizing Hormone/blood , Male , Ovariectomy , Polyunsaturated Alkamides , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/physiology , Sex Factors
18.
Proc Natl Acad Sci U S A ; 101(28): 10434-9, 2004 Jul 13.
Article in English | MEDLINE | ID: mdl-15231997

ABSTRACT

Ghrelin plays a key role in the regulation of growth hormone secretion and energy homeostasis. Adiponectin is exclusively secreted by adipose tissue and is abundantly present in the circulation, with important effects on metabolism. We studied five lean and five obese young men [ages: 24.2 +/- 1.0 (lean) and 21.8 +/- 1.6 (obese) years (difference not significant); body mass indexes: 35.0 +/- 1.3 and 23.0 +/- 0.3 kg/m2 (P = 0.01)], sampled blood every 7 min over 24 h, and measured ghrelin, adiponectin, and leptin in 2,070 samples for a total of 6,210 data points. Circulating 24-h ghrelin showed significant ultradian fluctuations and an orderly pattern of release in lean and obese subjects with similar pulsatility characteristics. Plasma adiponectin concentrations were significantly lower in the obese group, with lower pulse height. In contrast to leptin, which is secreted in an orderly manner, the 24-h patterns of adiponectin were not significantly different from random in both the lean and obese groups. We show here that adipocytes can simultaneously secrete certain hormones, such as leptin, in patterns that are orderly, whereas other hormones, such as adiponectin, are secreted in patterns that appear to be random. The cross-approximate entropy statistic revealed pattern synchrony among ghrelin-leptin, ghrelin-adiponectin, and leptin-adiponectin hormone time series in the lean and obese subjects. Plasma ghrelin concentrations showed a nocturnal rise that exceeded the meal-associated increases in lean subjects, and this newly identified nocturnal rise was blunted in the obese. We suggest that the blunting of the nocturnal rise of ghrelin is a biological feature of human obesity.


Subject(s)
Intercellular Signaling Peptides and Proteins , Leptin/blood , Obesity/blood , Peptide Hormones/blood , Proteins/metabolism , Adipocytes/metabolism , Adipocytes/physiology , Adiponectin , Adult , Body Weight/physiology , Circadian Rhythm/physiology , Energy Metabolism/physiology , Ghrelin , Humans , Leptin/metabolism , Male , Pulsatile Flow
19.
Exp Biol Med (Maywood) ; 229(7): 650-6, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15229359

ABSTRACT

Melatonin (MEL), the principle secretory product of the pineal gland, has been shown to function as an antioxidant and free-radical scavenger. We previously showed that the release of ascorbic acid (AA) and luteinizing hormone releasing hormone (LHRH) from medial basal hypothalamus (MBH) was mediated by nitric oxide (NO) that released cyclic guanosine 3'5'-mono-phosphate (cGMP). Therefore, it was of interest to evaluate the effect of MEL on AA and LHRH release and study the effect of a nitric oxide synthase (NOS) inhibitor, 6-anilino-5,8-quinoline-dione (LY 83583), and a guanylyl cyclase (GC) inhibitor, 1H-[1,2,4] oxadiazolo [4,3-a] quinoxalin-1-one (O.D.Q.), on the release process. Because NO has been shown to activate soluble guanylyl cyclase that elicited an elevation of cGMP in target cells, in the current investigation LY 83583, O.D.Q., or N(G)-monomethyl-l-arginine (NMMA), a competitive inhibitor of NOS, were used to evaluate their effects on MEL-induced AA and LHRH release. Medial basal hypothalami were incubated in 0.5 ml of Krebs-Ringer bicarbonate (KRB) buffer for 1 hr. Subsequently, the tissues were incubated with graded concentrations of MEL (10(-8) to 10(-4) M), MEL + NMMA (3 x 10(-4) M), MEL + LY 83583 (10(-6) M), or MEL + O.D.Q. (10(-5) M) for 1 hr. Ascorbic acid and LHRH released into the medium were measured by high-performance liquid chromatography (HPLC) and radio-immunoassay (RIA), respectively. Melatonin (10(-6) and 10(-5) M) significantly stimulated both AA and LHRH release, but the lower and the highest concentrations were ineffective. A combination of MEL + NMMA completely blocked both AA and LHRH release, supporting a role for NO in the releasing action. Both LY 83583 and O.D.Q. significantly suppressed MEL-induced AA and LHRH release, emphasizing the role of NOS, GC, and cGMP in mediating the action of MEL. The data of these in vitro experiments support a role for MEL in the hypothalamic control of AA and LHRH release.


Subject(s)
Aminoquinolines/pharmacology , Ascorbic Acid/metabolism , Cyclic GMP/physiology , Enzyme Inhibitors/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Melatonin/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Animals , Guanylate Cyclase/antagonists & inhibitors , Male , Rats , Rats, Sprague-Dawley , omega-N-Methylarginine/pharmacology
20.
Proc Natl Acad Sci U S A ; 101(13): 4531-6, 2004 Mar 30.
Article in English | MEDLINE | ID: mdl-15070752

ABSTRACT

Genetic mutations in the leptin pathway can be a cause of human obesity. It is still unknown whether leptin can be effective in the treatment of fully established morbid obesity and its endocrine and metabolic consequences in adults. To test the hypothesis that leptin has a key role in metabolic and endocrine regulation in adults, we examined the effects of human leptin replacement in the only three adults identified to date who have genetically based leptin deficiency. We treated these three morbidly obese homozygous leptin-deficient adult patients with recombinant human leptin at low, physiological replacement doses in the range of 0.01-0.04 mg/kg for 18 months. Patients were hypogonadal, and one of them also had type 2 diabetes mellitus. We chose the doses of recombinant methionyl human leptin that would achieve normal leptin concentrations and administered them daily in the evening to model the normal circadian variation in endogenous leptin. The mean body mass index dropped from 51.2 +/- 2.5 (mean +/- SEM) at baseline to 26.9 +/- 2.1 kg/m2 after 18 months of treatment, mainly because of loss of fat mass. We document here that leptin replacement therapy in leptin-deficient adults with established morbid obesity results in profound weight loss, increased physical activity, changes in endocrine function and metabolism, including resolution of type 2 diabetes mellitus and hypogonadism, and beneficial effects on ingestive and noningestive behavior. These results highlight the role of the leptin pathway in adults with key effects on the regulation of body weight, gonadal function, and behavior.


Subject(s)
Behavior/physiology , Diabetes Mellitus, Type 2/genetics , Hypogonadism/genetics , Leptin/deficiency , Leptin/therapeutic use , Adult , Body Composition , Body Mass Index , Circadian Rhythm , Female , Humans , Leptin/genetics , Phenotype , Weight Loss
SELECTION OF CITATIONS
SEARCH DETAIL
...