Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 164(8): 2015-28, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21615729

ABSTRACT

BACKGROUND AND PURPOSE: Little information exists on the mechanisms that precipitate brain stem death, the legal definition of death in many developed countries. We investigated the role of tropomyocin receptor kinase B (TrkB) and its downstream signalling pathways in the rostral ventrolateral medulla (RVLM) during experimental brain stem death. EXPERIMENTAL APPROACH: An experimental model of brain stem death that employed microinjection of the organophosphate insecticide mevinphos bilaterally into the RVLM of Sprague-Dawley rats was used, in conjunction with cardiovascular, pharmacological and biochemical evaluations. KEY RESULTS: A significant increase in TrkB protein, phosphorylation of TrkB at Tyr(516) (pTrkB(Y516) ), Shc at Tyr(317) (pShc(Y317) ) or ERK at Thr(202) /Tyr(204) , or Ras activity in RVLM occurred preferentially during the pro-life phase of experimental brain stem death. Microinjection bilaterally into RVLM of a specific TrkB inhibitor, K252a, antagonized those increases. Pretreatment with anti-pShc(Y317) antiserum, Src homology 3 binding peptide (Grb2/SOS inhibitor), farnesylthioacetic acid (Ras inhibitor), manumycin A (Ras inhibitor) or GW5074 (Raf-1 inhibitor) blunted the preferential augmentation of Ras activity or ERK phosphorylation in RVLM and blocked the up-regulated NOS I/protein kinase G (PKG) signalling, the pro-life cascade that sustains central cardiovascular regulation during experimental brain stem death. CONCLUSIONS AND IMPLICATIONS: Activation of TrkB, followed by recruitment of Shc/Grb2/SOS adaptor proteins, leading to activation of Ras/Raf-1/ERK signalling pathway plays a crucial role in ameliorating central cardiovascular regulatory dysfunction via up-regulation of NOS I/PKG signalling cascade in the RVLM in brain stem death. These findings provide novel information for developing therapeutic strategies against this fatal eventuality.


Subject(s)
Brain Death , Cardiovascular System/drug effects , Cholinesterase Inhibitors/toxicity , Mevinphos/toxicity , Receptor, trkB/metabolism , Animals , Blotting, Western , Cardiovascular System/physiopathology , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Male , Microinjections , Phosphorylation , Rats , Rats, Sprague-Dawley
2.
Neuropharmacology ; 52(5): 1263-73, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17336342

ABSTRACT

Status epilepticus results in preferential neuronal cell loss in the hippocampus. We evaluated the hypothesis that the repertoire of intracellular events in the vulnerable hippocampal CA3 subfield after induction of experimental temporal lobe status epilepticus entails upregulation of nitric oxide synthase II (NOS II), followed by the release of mitochondrial cytochrome c that triggers the cytosolic caspase-3 cascade, leading to apoptotic cell death. In Sprague-Dawley rats, significant and temporally correlated upregulation of NOS II (3-24h), but not NOS I or II expression, enhanced cytosolic translocation of cytochrome c (days 1 and 3), augmented activated caspase-3 in cytosol (days 1, 3 and 7) and DNA fragmentation (days 1, 3 and 7) was detected bilaterally in the hippocampal CA3 subfield after elicitation of sustained seizure activity by microinjection of kainic acid into the unilateral CA3 subfield. Application bilaterally into the hippocampal CA3 subfield of a selective NOS II inhibitor, S-methylisothiourea, significantly blunted these apoptotic events; a selective NOS I inhibitor, N(omega)-propyl-l-arginine or a potent NOS III inhibitor, N(5)-(1-iminoethyl)-l-ornithine was ineffective. We conclude that upregulation of NOS II contributes to apoptotic cell death in the hippocampal CA3 subfield via a cytochrome c/caspase-3 signaling cascade following the induction of experimental temporal lobe status epilepticus.


Subject(s)
Apoptosis/physiology , Caspase 3/physiology , Cytochromes c/physiology , Epilepsy, Temporal Lobe/physiopathology , Hippocampus/physiology , Nitric Oxide Synthase Type II/biosynthesis , Signal Transduction/physiology , Status Epilepticus/physiopathology , Up-Regulation/drug effects , Animals , Blotting, Western , Cytosol/enzymology , DNA Fragmentation/drug effects , Electroencephalography/drug effects , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Functional Laterality/physiology , Hippocampus/cytology , In Situ Nick-End Labeling , Male , Microscopy, Confocal , Nitric Oxide/biosynthesis , Nitric Oxide/physiology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
3.
Neuropharmacology ; 51(7-8): 1109-19, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16984802

ABSTRACT

Our current understanding of the nature of cell death that is associated with fatal organophosphate poisoning and the underlying cellular mechanisms is surprisingly limited. Taking advantage of the absence in an in vitro system of acetylcholinesterase, the pharmacological target of organophosphate compounds, the present study evaluated the hypothesis that the repertoire of cholinergic receptor-independent cellular events that underlie fatal organophosphate poisoning entails induction of mitochondrial dysfunction, followed by bioenergetic failure that leads to necrotic cell death because of ATP depletion. Pheochromocytoma PC12 cells incubated with the organophosphate pesticide mevinphos (0.4 or 4mumol) for 1 or 3h underwent a dose-related and time-dependent loss of cell viability that was not reversed by muscarinic (atropine) or nicotinic (mecamylamine) blockade. This was accompanied by depressed NADH cytochrome c reductase, succinate cytochrome c reductase or cytochrome c oxidase activity in the mitochondrial respiratory chain, reduced mitochondrial transmembrane potential, decreased ATP concentration, elevated ADP/ATP ratio, increased lactate dehydrogenase release and necrotic cell death. We conclude that Mev induces cholinergic receptor-independent necrotic cell death by depressing the activity of Complexes I to IV in the mitochondrial respiratory chain, eliciting reduction in mitochondrial transmembrane potential, depleting intracellular ATP contents and damaging cell membrane integrity.


Subject(s)
Adenosine Triphosphate/metabolism , Electron Transport/drug effects , Energy Metabolism/drug effects , Mevinphos/toxicity , Mitochondria/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , PC12 Cells/drug effects , Animals , Atropine/pharmacology , Chemical Warfare Agents/pharmacology , Chemical Warfare Agents/toxicity , Cholesterol/analogs & derivatives , Cholesterol/pharmacology , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/toxicity , Electron Transport Complex IV/antagonists & inhibitors , Insecticides/pharmacology , Insecticides/toxicity , L-Lactate Dehydrogenase/analysis , Mecamylamine/pharmacology , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mevinphos/antagonists & inhibitors , Mevinphos/pharmacology , Mitochondria/enzymology , Mitochondria/physiology , Muscarinic Antagonists/pharmacology , NADH Dehydrogenase/antagonists & inhibitors , Necrosis , Nicotinic Antagonists/pharmacology , Oxidative Phosphorylation/drug effects , PC12 Cells/physiology , Polyethylene Glycols/pharmacology , Rats , Receptors, Cholinergic/physiology , Ubiquinone/pharmacology
4.
Neuropharmacology ; 48(1): 161-72, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15617736

ABSTRACT

The organophosphate insecticide mevinphos (Mev) acts on the rostral ventrolateral medulla (RVLM), where sympathetic vasomotor tone originates, to elicit phasic cardiovascular responses via nitric oxide (NO) generated by NO synthase (NOS) I and II. We evaluated the contribution of soluble guanylyl cyclase (sGC)/cyclic guanosine monophosphate (cGMP)/protein kinase G (PKG) cascade and peroxynitrite in this process. PKG expression in ventrolateral medulla of Sprague-Dawley rats manifested an increase during the sympathoexcitatory phase (Phase I) of cardiovascular responses induced by microinjection of Mev bilaterally into the RVLM that was antagonized by co-administration of 7-nitroindazole or Nomega-propyl-L-arginine, two selective NOS I inhibitors or 1-H-[1,2,4]oxadiaolo[4,3-a]quinoxalin-1-one (ODQ), a selective sGC antagonist. Co-microinjection of ODQ or two PKG inhibitors, KT5823 or Rp-8-Br-cGMPS, also blunted the Mev-elicited sympathoexcitatory effects. However, the increase in nitrotyrosine, a marker for peroxynitrite, and the sympathoinhibitory circulatory actions during Phase II Mev intoxication were antagonized by co-administration of S-methylisothiourea, a selective NOS II inhibitor, Mn(III)-tetrakis-(4-benzoic acid) porphyrin, a superoxide dismutase mimetic, 5,10,15,20-tetrakis-N-methyl-4'-pyridyl)-porphyrinato iron (III), a peroxynitrite decomposition catalyst, or L-cysteine, a peroxynitrite scavenger. We conclude that sGC/cGMP/PKG cascade and peroxynitrite formation may participate in Mev-induced phasic cardiovascular responses as signals downstream to NO generated respectively by NOS I and II in the RVLM.


Subject(s)
Cyclic GMP/metabolism , Medulla Oblongata/drug effects , Mevinphos/pharmacology , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Peroxynitrous Acid/metabolism , Tyrosine/analogs & derivatives , Animals , Blood Pressure/drug effects , Blotting, Western/methods , Cardiovascular Physiological Phenomena/drug effects , Cholinesterase Inhibitors/pharmacology , Cyclic GMP-Dependent Protein Kinases/metabolism , Heart Rate/drug effects , Male , Microinjections/methods , Nerve Tissue Proteins/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Rats , Spectrum Analysis/methods , Superoxides/metabolism , Time Factors , Tyrosine/metabolism
5.
Neuropharmacology ; 46(8): 1184-94, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15111025

ABSTRACT

The organophosphate poison mevinphos (Mev) elicits cardiovascular responses via nitric oxide (NO) produced on activation of M2 muscarinic receptors (M2R) in the rostral ventrolateral medulla (RVLM), where sympathetic vasomotor tone originates. This study further evaluated the contribution of nitric oxide synthase (NOS) isoforms at the RVLM to this process, using adult Sprague-Dawley rats. Bilateral co-microinjection into the RVLM of the selective NOS I inhibitor (250 pmol), 7-nitroindazole or N(omega)-propyl-L-arginine antagonized the initial sympathoexcitatory cardiovascular responses to Mev (10 nmol). Co-administration of a selective NOS II inhibitor, N6-(1-iminoethyl)-L-lysine (250 or 500 pmol) further enhanced these cardiovascular responses and reversed the secondary sympathoinhibitory actions of Mev. A potent NOS III inhibitor, N5-(1-iminoethyl)-L-ornithine (46 or 92 nmol) was ineffective. We also found that M2R co-localized only with NOS I- or NOS II-immunoreactive RVLM neurons. Furthermore, only NOS I or II in the ventrolateral medulla exhibited an elevation in mRNA or protein levels during the sympathoexcitatory phase, with further up-regulated synthesis of NOS II during the sympathoinhibitory phase of Mev intoxication. We conclude that whereas NOS III is not engaged, NO produced by NOS I and II in the RVLM plays, respectively, a sympathoexcitatory and sympathoinhibitory role in the cardiovascular responses during Mev intoxication.


Subject(s)
Blood Pressure/drug effects , Heart Rate/drug effects , Medulla Oblongata/drug effects , Mevinphos/toxicity , Nitric Oxide Synthase/physiology , Animals , Blood Pressure/physiology , Heart Rate/physiology , Isoenzymes/physiology , Male , Medulla Oblongata/enzymology , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Rats , Rats, Sprague-Dawley
6.
Neuropharmacology ; 46(1): 126-32, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14654104

ABSTRACT

We established previously that nitric oxide (NO) in the hippocampal formation (HF) participates actively in negative feedback regulation of penile erection. This study further evaluated whether this process engaged soluble guanylyl cyclase (sGC)/cGMP cascade or peroxynitrite in the HF. Intracavernous pressure (ICP) recorded from the penis in adult, male Sprague-Dawley rats anesthetized with chloral hydrate was employed as our experimental index for penile erection. Microinjection bilaterally of a NO-independent sGC activator, YC-1 (0.1 or 1 nmol) or a cGMP analog, 8-Bromo-cGMP (0.1 or 1 nmol), into the HF elicited a significant reduction in baseline ICP. Bilateral application into the HF of equimolar doses (0.5 or 1 nmol) of a sGC inhibitor, LY83583 or a NO-sensitive sGC inhibitor, ODQ significantly antagonized the decrease in baseline ICP induced by co-administration of the NO precursor, L-arginine (5 nmol), along with significant enhancement of the magnitude of papaverine-induced elevation in ICP. In contrast, a peroxynitrite scavenger, L-cysteine (50 or 100 pmol), or an active peroxynitrite decomposition catalyst, 5,10,15,20-tetrakis-(N-methyl-4'-pyridyl)-porphyrinato iron (III) (10 or 50 pmol), was ineffective in both events. These results suggest that NO may participate in negative feedback regulation of penile erection by activating the sGC/cGMP cascade in the HF selectively.


Subject(s)
Cyclic GMP/analogs & derivatives , Cyclic GMP/physiology , Feedback/physiology , Hippocampus/physiology , Nitric Oxide/physiology , Penile Erection/drug effects , Peroxynitrous Acid/physiology , Aminoquinolines/pharmacology , Animals , Cyclic GMP/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Hippocampus/drug effects , Indazoles/pharmacology , Male , Microinjections , Rats , Rats, Sprague-Dawley , Venous Pressure/drug effects
7.
Acta Anaesthesiol Scand ; 47(5): 532-40, 2003 May.
Article in English | MEDLINE | ID: mdl-12699509

ABSTRACT

BACKGROUND: General anaesthesia may stimulate the renin-angiotensin system. Exogenous administration of angiotensin II elevates blood pressure mainly via increased total peripheral resistance caused by direct vasoconstrictor actions. It is also well established that the hypertensive effect of angiotensin II involves a cerebrally mediated component. The hypertensive effect of an intravascular infusion of angiotensin II is substantially reduced by isoflurane anaesthesia. A likely mechanism is that isoflurane anaesthesia reduces the cerebral component of the angiotensin II effect on blood pressure, which involves influences on autonomic nervous activity. In an experimental study in sheep we used real-time spectral analysis of arterial blood pressure signals to obtain information on parasympathetic, respectively, sympathetic autonomic nervous activity in response to angiotensin II administration during isoflurane anaesthesia. METHODS: The study was performed on conscious and isoflurane-anaesthetized sheep that were subjected to an intracarotid infusion of angiotensin II (85 ng kg(-1) min(-1)) during 20 min followed by a recovery period of 30 min and thereafter an injection of the angiotensin II, AT1-receptor antagonist losartan (10 mg kg(-1)) i.v. Systemic and regional (renal and femoral) circulation was monitored in parallel to real-time spectral analysis of the arterial blood pressure signal. RESULTS: Isoflurane anaesthesia reduced both magnitude and duration of the hypertensive response to angiotensin II infusion. The power spectral density in the frequency band that represents sympathetic activation, correlated to the changes in mean arterial pressure in conscious animals, but not during isoflurane anaesthesia. CONCLUSION: We conclude that the cerebrally mediated component of the hypertensive effect of circulating angiotensin II is largely eliminated by isoflurane anaesthesia. Spectral power analysis of the blood pressure signal indicates that the cerebral angiotensin II effect involves activation of sympathetic nervous activity.


Subject(s)
Anesthesia , Angiotensin II/pharmacology , Hemodynamics/drug effects , Anesthesia, Inhalation , Anesthetics, Inhalation/pharmacology , Animals , Blood Pressure/drug effects , Cardiac Output/drug effects , Female , Femur/blood supply , Heart Rate/drug effects , Infusions, Intravenous , Isoflurane/pharmacology , Parasympathetic Nervous System/drug effects , Regional Blood Flow/drug effects , Renal Circulation/drug effects , Renin/blood , Sheep , Sympathetic Nervous System/drug effects , Vascular Resistance/drug effects , Vasopressins/blood
8.
Neuropharmacology ; 43(5): 889-98, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12384174

ABSTRACT

We evaluated the contribution of peroxynitrite to the fatal cardiovascular depression induced by overproduction of nitric oxide (NO) after activation of inducible NO synthase (iNOS) in the rostral ventrolateral medulla (RVLM), the origin of sympathetic vasomotor tone. In Sprague-Dawley rats maintained under propofol anesthesia, microinjection of E. coli lipopolysaccharide (LPS) bilaterally into the RVLM elicited significant hypotension, bradycardia, reduction in sympathetic vasomotor tone and mortality. There was also a discernible elevation of iNOS expression in the ventrolateral medulla, followed by a massive production of nitrotyrosine, an experimental index for peroxynitrite. Co-administration bilaterally into the RVLM of the selective iNOS inhibitor, S-methylisothiourea (50, 100 or 250 pmol), an active peroxynitrite decomposition catalyst, 5,10,15,20-tetrakis- (N-methyl-4'-pyridyl)-porphyrinato iron (III) (10 or 50 pmol), a peroxynitrite scavenger, L-cysteine (5, 50 or 100 pmol), or a superoxide dismutase mimetic, Mn(III)-tetrakis-(4-benzoic acid) porphyrin (1 or 10 pmol), significantly prevented mortality, reduced nitrotyrosine production and reversed the NO-induced cardiovascular suppression after application of LPS into the RVLM. We conclude that the formation of peroxynitrite by a reaction between superoxide anion and NO is primarily responsible for the fatal cardiovascular depression induced by overproduction of NO after activation of iNOS at the RVLM.


Subject(s)
Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/mortality , Hemodynamics/drug effects , Medulla Oblongata/metabolism , Nitric Oxide/metabolism , Nitric Oxide/toxicity , Peroxynitrous Acid/metabolism , Peroxynitrous Acid/toxicity , Animals , Blotting, Western , Cyclic GMP/metabolism , Depression, Chemical , Guanylate Cyclase/metabolism , Male , Medulla Oblongata/cytology , Medulla Oblongata/drug effects , Microinjections , Muscle Tonus/drug effects , Muscle, Smooth, Vascular/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Rats , Rats, Sprague-Dawley , Survival Analysis , Time Factors
9.
Neuroscience ; 112(1): 113-20, 2002.
Article in English | MEDLINE | ID: mdl-12044476

ABSTRACT

Antihypertensive therapy not only normalizes the elevated blood pressure but also restores the reduced baroreceptor reflex response associated with hypertension, although the underlying mechanism is not fully understood. We assessed the hypothesis that a reversal of the enhanced basal Fos expression seen during hypertension in nucleus tractus solitarii, the terminal site of baroreceptor afferents, underlies the restoration of baroreceptor reflex sensitivity after antihypertensive treatment. Male adult spontaneously hypertensive or normotensive Wistar-Kyoto rats received for 3 weeks captopril (100 mg/kg/day) added to their drinking water. Evaluated subsequently under pentobarbital anesthesia, captopril-treated spontaneously hypertensive rats exhibited significantly lowered systolic blood pressure and restoration of the sensitivity in baroreceptor reflex control of heart rate to levels comparable with Wistar-Kyoto rats. Reverse transcription-polymerase chain reaction analysis and immunohistochemical evaluation revealed concomitant down-regulation of basal expression in nucleus tractus solitarii of c-fos gene at both mRNA and protein levels. Captopril treatment, on the other hand, elicited no discernible effect on systolic blood pressure, cardiac baroreceptor reflex sensitivity or basal expression of Fos protein at the nucleus tractus solitarii of normotensive Wistar-Kyoto rats. From these findings we suggest that a down-regulation of basal Fos expression in nucleus tractus solitarii may contribute to the restoration of baroreceptor reflex sensitivity in spontaneously hypertensive rats that received antihypertensive treatment such as captopril.


Subject(s)
Antihypertensive Agents/pharmacology , Baroreflex/drug effects , Captopril/pharmacology , Hypertension/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats, Inbred SHR/metabolism , Solitary Nucleus/metabolism , Animals , Blood Pressure/drug effects , Down-Regulation , Hypertension/physiopathology , Immunohistochemistry , Male , RNA, Messenger/metabolism , Rats , Rats, Inbred WKY , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Systole
10.
J Biomed Sci ; 4(2-3): 91-97, 1997.
Article in English | MEDLINE | ID: mdl-11725139

ABSTRACT

We evaluated the potential participation of galanin (GAL) at the paraventricular nucleus of hypothalamus (PVN) in the suppression of baroreceptor reflex (BRR) response by locus ceruleus (LC), using adult male Sprague-Dawley rats anesthetized with pentobarbital sodium. Microinjection of GAL (100 pmol) bilaterally into the PVN significantly depressed the BRR response. This suppressive effect was appreciably antagonized when GAL (100 pmol) and GAL antiserum (1:20) were coadministered into the bilateral PVN. Whereas bilateral microinjection of GAL antiserum into the PVN by itself elicited minimal effect, it nevertheless significantly attenuated the suppressive effect of either electrical or chemical activation of LC on the BRR response. Pretreatment with the same amount of normal rabbit serum (1:20), on the other hand, was ineffective. These results suggest that a galaninergic projection from the LC to PVN may participate in the suppression of BRR response by this dorsal pontine nucleus. Copyright 1997 S. Karger AG, Basel

11.
J Biomed Sci ; 3(5): 338-347, 1996.
Article in English | MEDLINE | ID: mdl-11725116

ABSTRACT

The present study was undertaken to further characterize the nucleus reticularis gigantocellularis (NRGC) of the medulla oblongata in the central processing of nociceptive and cardiovascular signals, and its modulation by met-enkephalin. In Sprague-Dawley rats anesthetized with pentobarbital sodium, we found that all 125 spontaneously active NRGC neurons that responded to noxious stimuli (tail clamp) also exhibited arterial pressure-relatedness. Forty neurons additionally manifested cardiac periodicity that persisted even during nociceptive responses. While maintaining their cardiovascular responsive characteristics, the nociception-related NRGC neuronal activity was blocked, naloxone-reversibly (0.5 mg/kg, i.v.), by morphine (5 mg/kg, i.v.). Microiontophoretically applied met-enkephalin suppressed the responsiveness of NRGC neurons to individually delivered tail clamp or transient hypertension induced by phenylephrine (5 &mgr;g/kg, i.v.). Interestingly, in NRGC neurons that manifested both nociception and arterial pressure relatedness, the preferential reduction in the response to noxious stimuli upon simultaneous elevation in systemic arterial pressure was reversed to one that favored nociception in the presence of met-enkephalin. All actions of met-enkephalin were discernibly blocked by the opioid receptor antagonist, naloxone. Our results suggest that individual NRGC neurons may participate in the processing of both nociceptive and cardiovascular information, or in the coordination of the necessary circulatory supports during nociception. In addition, neuropeptides such as met-enkephalin may exert differential modulation on neuronal responsiveness according to the prevailing physiologic status of the animal. They also showed that NRGC may be a central integrator for pain and cardiovascular-related functions. Copyright 1996 S. Karger AG, Basel

12.
J Biomed Sci ; 3(3): 203-210, 1996.
Article in English | MEDLINE | ID: mdl-11725101

ABSTRACT

We examined the physiologic role of endogenous brain angiotensin III (AIII), an active degradative product of angiotensin II, in drinking behavior. Adult, male spontaneously hypertensive (SH) and Wistar-Kyoto normotensive (WKY) rats that were instrumented with an intracerebroventricular (i.c.v.) cannula connected to an osmotic minipump for chronic infusion were used. 7-day i.c.v. infusion of the specific AIII antagonist, Ile(7)-AIII (10 or 100 pmol/min), resulted in no significant alteration in daily (24 h), diurnal (8:00 a.m.-8.00 p.m.) or nocturnal (8:00 p.m.-8:00 a.m.) basal water intake in both SH and WKY rats. Similar results were obtained with i.c.v. infusion of the aminopeptidase inhibitor, bestatin (150 or 300 pmol/min), given alone or simultaneously with Ile(7)-AIII (10 pmol/min). Rats that were water-deprived for the first 3 days of 7-day infusion of Ile(7)-AIII consumed significantly less water during the first 2 h after water became available. Furthermore, the accumulated water intake during the first 24 h was appreciably greater in SH than WKY rats. We interpret these results to suggest that the endogenous brain AIII may not be tonically involved in fluid homeostasis. Instead, it must be activated under conditions of dehydration, such as water deprivation, particularly in the SHRs, to initiate drinking behavior. Copyright 1996 S. Karger AG, Basel

13.
J Biomed Sci ; 2(3): 263-271, 1995 Aug.
Article in English | MEDLINE | ID: mdl-11725063

ABSTRACT

Previous results from our laboratory have established that the G(o) subtype of guanine nucleotide (GTP)-binding regulatory protein at the locus coeruleus (LC) may participate in the elicitation of muscular rigidity by fentanyl. The present study further examined the involvement of other subtypes of GTP-binding regulatory proteins at the LC in this process, using Sprague-Dawley rats anesthetized with ketamine (120 mg/kg, i.p., with 30 mg/kg/h i.v. infusion supplements) and under mechanical ventilation. Intravenous administration of fentanyl (100 &mgr;g/kg) induced a significant increase in electromyographic signals recorded from the sacrococcygeus dorsi lateralis muscle. Power spectral analysis revealed that this was accomplished by a decrease in the mean power frequency and an increase in the root mean square values of the signals. The above responses were appreciably antagonized by pretreating animals with bilateral microinjection into the LC of pertussis toxin (80 or 160 fmol), N-ethylmaleimide (16 pmol) or 1-(5-isoquinolinesulfonyl)-2-methylpiperazine (100 or 200 fmol); but not by cholera toxin (120 or 240 fmol), forskolin (240 or 480 pmol) or N-ethylmaleimide at a higher dose (32 pmol). These results suggest that, in addition to G(o) protein, fentanyl-induced muscular rigidity may also involve other pertussis toxin-sensitive GTP-binding regulatory proteins, possibly G(i) and G(p) subtypes, in the signal transduction processes following activation of &mgr;-opioid receptors at the LC. Copyright 1995 S. Karger AG, Basel

14.
J Biomed Sci ; 1(1): 13-18, 1994 Dec.
Article in English | MEDLINE | ID: mdl-11725002

ABSTRACT

In adult male Sprague-Dawley rats anesthetized with pentobarbital sodium, we elucidated the molecular consequence of central alpha(2)-adrenoceptor activation. The hypotensive and negative chronotropic and inotropic actions of the alpha(2)-adrenoceptor agonist guanabenz were used as our experimental index. Intracerebroventricular administration of pertussis toxin (2.5 &mgr;g) significantly attenuated the cardiovascular suppressant effects of the aminoguanidine compound (100 &mgr;g/kg i.v.). However, application of N-ethylmaleimide (0.125 or 0.250 &mgr;g), phorbol 12-myristate 13-acetate (1.25 or 2.50 &mgr;g), cholera toxin (1.25 or 2.50 &mgr;g) or forskolin (12.5 or 25.0 &mgr;g) into the lateral cerebral ventricle elicited no appreciable blunting effect on the circulatory depression produced by guanabenz. These results were essentially duplicated when pertussis toxin (0.125 or 0.250 &mgr;g), N-ethylmaleimide (0.0125 or 0.05 &mgr;g), phorbol 12-myristate 13-acetate (0.125 or 0.25 &mgr;g), cholera toxin (0.125 or 0.25 &mgr;g) or forskolin (1.25 or 2.50 &mgr;g) was microinjected bilaterally to the nucleus reticularis gigantocellularis, a medullary site believed to be intimately related to the antihypertensive action of guanabenz. These findings suggest that stimulation of the alpha(2)-adrenoceptors in the medulla oblongata may result in the activation of a pertussis toxin-sensitive GTP-binding regulatory protein. They further suggest that the biologic signals subsequent to this action may not be linked to Gs, Gi or Gp but possibly Go. Copyright 1994 S. Karger AG, Basel

SELECTION OF CITATIONS
SEARCH DETAIL
...