Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Cell Mol Neurobiol ; 38(6): 1197-1206, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29802603

ABSTRACT

It has become apparent that the molecular and biochemical integrity of interactive families, genera, and species of human gut microflora is critically linked to maintaining complex metabolic and behavioral processes mediated by peripheral organ systems and central nervous system neuronal groupings. Relatively recent studies have established intrinsic ratios of enterotypes contained within the human microbiome across demographic subpopulations and have empirically linked significant alterations in the expression of bacterial enterotypes with the initiation and persistence of several major metabolic and psychiatric disorders. Accordingly, the goal of our review is to highlight potential thematic/functional linkages of pathophysiological alterations in gut microbiota and bidirectional gut-brain signaling pathways with special emphasis on the potential roles of gut dysbiosis on the pathophysiology of psychiatric illnesses. We provide critical discussion of putative thematic linkages of Parkinson's disease (PD) data sets to similar pathophysiological events as potential causative factors in the development and persistence of diverse psychiatric illnesses. Finally, we include a concise review of preclinical paradigms that involve immunologically-induced GI deficits and dysbiosis of maternal microflora that are functionally linked to impaired neurodevelopmental processes leading to affective behavioral syndromes in the offspring.


Subject(s)
Brain/metabolism , Dysbiosis/genetics , Gastrointestinal Microbiome/genetics , Microbiota/genetics , Parkinson Disease/genetics , Animals , Brain/physiopathology , Humans , Mental Disorders/genetics , Mental Disorders/physiopathology , Parkinson Disease/physiopathology
2.
Folia Biol (Praha) ; 58(2): 49-56, 2012.
Article in English | MEDLINE | ID: mdl-22578954

ABSTRACT

Positive evolutionary pressure has apparently preserved the ability to synthesize chemically authentic morphine, albeit in homeopathic concentrations, throughout animal phyla. Despite the establishment of a progressively rigorous and mechanistically focused historical literature extending from the mid 1970s to the mid 1980s that supported the expression of chemically authentic morphine by animal cellular and organ systems, prejudicial scepticism and early dismissal by scientists and clinicians most often obscured widespread acceptance of the biological importance and medical implications of endogenous morphine. The current critical paper presents and evaluates key recent coordinated studies in endogenous morphine research, highlighting those that have advanced our understanding of the functional roles of cognate alkaloid-selective µ(3) and µ(4) opiate receptors. We propose that the expression of endogenous morphine by animal and human cells is designed to mediate homeopathic regulation of metabolic activity via activation of cognate µ(3) and µ(4) receptors that serve as transductive conduits for shortcircuit Ca(++) fluxes. The implications of endogenous morphine coupling to nitric oxide regulation of mitochondrial function, with special reference to the cardiovascular system, are now formulated after many years of neglect.


Subject(s)
Morphine/metabolism , Receptors, Opioid, mu/metabolism , Animals , Cardiovascular System , Dopamine/metabolism , Gene Expression Regulation , Humans , Mitochondria/metabolism , Models, Biological , Models, Chemical , Nitric Oxide/metabolism , Signal Transduction
3.
J Hum Hypertens ; 24(11): 713-20, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20613782

ABSTRACT

It has been shown that vascular endothelial cells functionally express a local circuit autocrine-paracrine regulatory pathway driven by endogenously expressed chemically authentic morphine, its cognate opiate alkaloid-selective mu3 and mu4 receptors, and constitutive nitric oxide (NO). Accordingly, the aim of the study was to examine morphine-mediated changes in hypertension-associated gene expression in two independent cell models: primary cultures of human white blood cells (WBCs) and human multilineage progenitor cells (MLPCs). In separate incubations, primary cultures of human WBC and MLPC were treated with morphine at a final concentration of 1 µM morphine for 2-4 h. After RNA extraction and reverse transcription, Human Genome Survey Arrays were used to construct and differentially analyze by strict statistical criteria transcriptional/gene expression profiles of WBC and undifferentiated human MLPC in three independent experiments. The Applied Biosystems Human Genome Survey Array contains 31,700 60-mer oligonucleotide probes representing a set of 27,868 individual human genes and >1000 control probes. After DNA microarray analyses, a variety of hypertension-associated genes from both cell types were observed to be significantly downregulated. The only genes expressed in both cell types were ß-adrenergic receptor kinase 2 (ADRBK2) and coding protein kinase WNK1 (PRKWNK1); however, only PRKWNK1 showed downregulation of its expression after morphine exposure. Only two genes were observed to be significantly upregulated after morphine treatment: ADRBK2 in stem cells and ß3-adrenergic receptor in WBC. Morphine administration to primary cultures of human WBC and MLPC altered the expression profile of 16 candidate hypertension-associated genes. The majority of relevant genes was observed to be downregulated, suggesting ongoing homeostatic regulation by endogenous morphine coupled to NO production and release. In sum, these data suggest a predominantly antihypertensive role for endogenous morphine/NO signaling events.


Subject(s)
Cell Lineage , Gene Expression Regulation/drug effects , Hypertension/genetics , Leukocytes/drug effects , Morphine/pharmacology , Stem Cells/drug effects , Cells, Cultured , G-Protein-Coupled Receptor Kinase 3/genetics , Gene Expression Profiling/methods , Humans , Hypertension/blood , Hypertension/physiopathology , Hypertension/prevention & control , Intracellular Signaling Peptides and Proteins , Leukocytes/metabolism , Minor Histocompatibility Antigens , Nitric Oxide/metabolism , Oligonucleotide Array Sequence Analysis , Opioid Peptides/metabolism , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/blood , Receptors, Adrenergic, beta-3/genetics , Stem Cells/metabolism , Time Factors , WNK Lysine-Deficient Protein Kinase 1
4.
Folia Biol (Praha) ; 56(6): 231-41, 2010.
Article in English | MEDLINE | ID: mdl-21324264

ABSTRACT

For over 30 years empirical studies have repeatedly demonstrated that the biosynthesis of morphine by diverse animal and human tissues occurs. Recently, the blue mussel's neural tissues and human white blood cells were used to demonstrate the de novo biosynthesis of morphine for small precursor molecules derived from the aromatic amino acid L-tyrosine. Because catecholamine precursors, i.e., L-3,4-dihydroxyphenylalanine (L-DOPA), were also found to be utilized as morphine precursors, a novel reciprocally interactive mechanism is apparent that links catecholamine and opioid pathways in the activation and inhibition of diverse tissue responses. Additionally, these observations provide new insights into morphinergic signalling that transcend analgesia and addiction. We have also linked the biological effects of nitric oxide into a common effect in endogenous morphine signalling. Given the singular importance of dopamine and morphine's interaction in the CNS, the presence and association of this signalling with nitric oxide all promises to provide novel answers for mental health phenomena, which have been lacking because of the inability in accepting the empirical endogenous morphine studies.


Subject(s)
Morphine Dependence/psychology , Morphine/biosynthesis , Mytilus edulis/metabolism , Animals , Catecholamines/metabolism , Dopamine/pharmacology , Humans , Leukocytes/metabolism , Levodopa/metabolism , Morphine/pharmacology , Nitric Oxide/pharmacology , Opioid Peptides/metabolism , Signal Transduction , Tyrosine/metabolism
5.
Int J Immunopathol Pharmacol ; 20(2): 229-37, 2007.
Article in English | MEDLINE | ID: mdl-17624236

ABSTRACT

Recent work from our laboratory has demonstrated that human white blood cells make morphine and that substances of abuse, i.e. nicotine, alcohol and cocaine have the ability to release this endogenous substance, suggesting a common mechanism of action. We now demonstrate that the nicotinic process is more complex than formerly envisioned. The incorporation rate of 125I-labeled morphine into PMN and MN are 7.85+/-0.36%, 1.42+/-0.19%, respectfully, suggesting in MN this process is of low activity. Separate incubations of PMN with varying concentrations of nicotine or the nicotine agonist epibatidine resulted in a statistically significant enhancement of 125I-trace labeled morphine released into the extracellular medium. In order to ascertain the specificity of the nicotine stimulated morphine release the following experiments were performed. Co-incubation of hexamethonium dichloride (5 microg/ml and at 10 microg/ml), which preferentially blocks nicotinic receptors at autonomic ganglia, with nicotine, exerted a very weak inhibitory effect. Co-incubation of alpha-BuTx or atropine or chlorisondamine diiodide or dihydro-Beta-erythroidine hydrobromide, an alpha4Beta2 receptor antagonist, did not block nicotine induced morphine release alone or in combination, suggesting either the response was not specific or it was mediated by a novel nicotinic receptor. Human leukocyte total RNA isolated from whole blood were analyzed, using the Human Genome Survey microarray (Applied Biosystems), for cholinergic receptor expression. PMN nicotinic receptor gene expression was present and contained numerous variants (eight). The number of variants suggests that indeed a novel nicotinic receptor may be mediating this effect, while simultaneously demonstrating the significance of the cholinergic receptor expression in these immune cells.


Subject(s)
Leukocytes/metabolism , Morphine/pharmacokinetics , Oligonucleotide Array Sequence Analysis , Receptors, Nicotinic/metabolism , Cells, Cultured , Humans , Nicotine/pharmacokinetics
6.
J Pharmacol Exp Ther ; 295(3): 1142-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11082451

ABSTRACT

The neuropeptide substance P (SP), apart from its traditional role in spinal nociceptive processing, is an important regulatory effector of opioid-dependent analgesic processes. The present study stems from our original findings indicating that 1) pharmacologically administered SP mediates a strong inhibitory activity on the development of morphine tolerance in rats, and that 2) a novel SP-opioid peptide chimera YPFFGLM-NH(2), designated ESP7, produces opioid-dependent analgesia without tolerance development. To further examine the effects of simultaneous activation of two distinct opposing spinal systems on opioid tolerance and the mechanisms underlying chimeric peptide function, a second SP-opioid chimera was synthesized. This chimera, designated ESP6 (YPFFPLM-NH(2)), contains overlapping domains of endomorphin-2 and SP, respectively. ESP6 is distinguished from ESP7 by a glycine to proline substitution at position 5. Intrathecal administration of morphine sulfate (MS) with ESP6 leads to a prolongation of MS analgesia over a 5-day period. The analgesia produced by ESP6 and MS is opioid receptor-dependent, due to the ability of naltrexone to block the analgesic response. Furthermore, when ESP6 and MS are administered with concurrent NK-1 receptor blockade, a decay in analgesic potency similar to that seen with MS alone results. The presence of a proline in ESP6 appears to reduce its conformational flexibility, limit its potency at the micro-opioid receptor, and hinder its analgesic effectiveness alone. However, ESP6 represents a novel adjuvant for the maintenance of opioid analgesia over time and provides a means to predict the pharmacological properties of a chimera from its structure.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Oligopeptides/pharmacology , Substance P/pharmacology , Animals , Drug Tolerance , Indoles/pharmacology , Isoindoles , Male , Naltrexone/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/physiology , Structure-Activity Relationship
7.
Proc Natl Acad Sci U S A ; 97(13): 7621-6, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10852965

ABSTRACT

To elucidate mechanisms of acute and chronic pain, it is important to understand how spinal excitatory systems influence opioid analgesia. The tachykinin substance P (SP) represents the prototypic spinal excitatory peptide neurotransmitter/neuromodulator, acting in concert with endogenous opioid systems to regulate analgesic responses to nociceptive stimuli. We have synthesized and pharmacologically characterized a chimeric peptide containing overlapping NH(2)- and COOH-terminal functional domains of the endogenous opioid endomorphin-2 (EM-2) and the tachykinin SP, respectively. Repeated administration of the chimeric molecule YPFFGLM-NH(2), designated ESP7, into the rat spinal cord produces opioid-dependent analgesia without loss of potency over 5 days. In contrast, repeated administration of ESP7 with concurrent SP receptor (SPR) blockade results in a progressive loss of analgesic potency, consistent with the development of tolerance. Furthermore, tolerant animals completely regain opioid sensitivity after post hoc administration of ESP7 alone, suggesting that coactivation of SPRs is essential to maintaining opioid responsiveness. Radioligand binding and signaling assays, using recombinant receptors, confirm that ESP7 can coactivate mu-opioid receptors (MOR) and SPRs in vitro. We hypothesize that coincidental activation of the MOR- and SPR-expressing systems in the spinal cord mimics an ongoing state of reciprocal excitation and inhibition, which is normally encountered in nociceptive processing. Due to the ability of ESP7 to interact with both MOR and SPRs, it represents a unique prototypic, anti-tolerance-forming analgesic with future therapeutic potential.


Subject(s)
Analgesics/pharmacology , Oligopeptides/pharmacology , Pain/prevention & control , Recombinant Fusion Proteins/pharmacology , Substance P/pharmacology , Animals , Drug Design , Male , Oligopeptides/genetics , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Substance P/genetics
8.
Neuroreport ; 10(14): 3015-9, 1999 Sep 29.
Article in English | MEDLINE | ID: mdl-10549815

ABSTRACT

Prompted by previous studies linking social defeat stress to changes in opioid antinociception, we evaluated the expression of mu-opioid receptor (MOR)-encoding mRNA in selected rat brain areas as a function of this type of stress. Because opioids mediate significant regulatory activities of brain dopamine neurons, dopaminergic loci in the ventral tegmental area (VTA) and substantia nigra (SN) were selected for analysis. Within 30 min after social defeat stress, the level of MOR-encoding mRNA, as detected and quantified by in situ hybridization histochemistry, increased in the lateral VTA and this increase was present for at least 6 h. In contrast, defeat stress was without effect on the expression of MOR-encoding mRNA in the SN. These data suggest that stress-induced alteration of MOR-encoding mRNA expression in the VTA may be involved in the consequences of social defeat stress.


Subject(s)
RNA, Messenger/biosynthesis , Receptors, Opioid, mu/biosynthesis , Social Behavior , Stress, Psychological/physiopathology , Ventral Tegmental Area/metabolism , Animals , Autoradiography , Dopamine/physiology , Image Processing, Computer-Assisted , In Situ Hybridization , Male , Mesencephalon/physiology , Neuronal Plasticity/physiology , Rats , Rats, Long-Evans , Stress, Psychological/metabolism
9.
Pain ; 79(2-3): 187-200, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10068164

ABSTRACT

Recent animal models of experimental nerve injury have proven useful in evaluating potential sympathetic involvement in neuropathic pain syndromes. We have employed a widely adopted unilateral L5/L6 spinal nerve ligation model to compare the development of mechanical allodynia with neurochemical changes both at the site of peripheral nerve injury and in the dorsal root ganglia (DRG). We have focused on the expression of neuropeptide Y (NPY), a well-studied regulatory peptide and phenotypic marker of sympathetic neurons, and functionally related Y-receptor binding sites following nerve injury. In sympathetic neurons, NPY is colocalized and coreleased with norepinephrine (NE) at peripheral sites of action. Furthermore, NPY gene expression is induced within the population of medium- and large-diameter DRG neurons of the A beta-fiber class after experimental nerve injury. We therefore hypothesized that concurrent alterations in NPY and NE expression by sympathetic and sensory neurons may be a contributing factor to sympathetically-maintained neuropathic conditions. Animals with unilateral L5/L6 spinal nerve ligation developed mechanical allodynia of the hind paw ipsilateral to the site of injury that persisted until sacrifice at postoperative day 10. A significant induction of preproneuropeptide Y-encoding (PPNPY) mRNA, as detected by in situ hybridization histochemistry (ISHH), occurred in populations of medium- and large-diameter DRG neurons ipsilateral to the site of injury. Immunohistochemical analysis indicated a marked decline in the number of labeled sympathetic axons positive for tyrosine hydroxylase-like and NPY-like immunoreactivities (TH-LI and NPY-LI, respectively) proximal to the site of nerve injury and almost complete elimination of immunopositive fibers distal to the site of ligation. Whereas, the extent of colocalization of NPY-LI to TH-LI-positive sympathetic axons in unaffected L4 or L5 nerve segments exceeded 80%, this figure declined to approximately 50% in regenerating axons of ligated spinal nerve L5. The portion of NPY-LI that was not colocalized to sympathetic TH-LI-positive fibers was most likely contributed by regenerating sensory axons, consistent with marked de novo synthesis of NPY by DRG neurons. In end bulb axon terminals, i.e. morphological profiles characteristic of neuromas, NPY-LI-positive elements that were not colocalized to TH-LI-positive sympathetic elements appeared to be spatially segregated from those of sympathetic origin with colocalized TH-LI and NPY-LI. Receptor autoradiography indicated that small- and medium-diameter DRG somata of the C-fiber class normally express both Y1 and Y2 receptor subtypes. The pattern of the distribution of Y-receptor binding sites appeared to be relatively unaffected by spinal nerve ligation. In contrast, there was a marked increase in the density of Y2 receptor binding sites in the proximal segment of ligated spinal nerve L5, consistent with previously published data indicating differential transport of the Y2 autoregulatory receptor subtype to nerve terminals. Induction of NPY gene expression in injured DRG neurons is consistent with appearance of NPY-LI-positive end bulbs derived from regenerating sensory axons that are found in developing neuromas containing a relatively high density of transported prejunctional Y2 receptors. Newly established functional interactions of spatially segregated sensory- and sympathetically-derived end bulbs in developing neuromas may enhance neuronal hyperexcitability engendered by aberrant electrical activity at the site of injury. Injury-related alterations in the regulatory activities of NPY released within the DRG at somally-distributed Y-receptors may also contribute to the development and/or persistence of symptoms characteristic of sympathetically-maintained pain. Finally, at later times NPY-mediated modulation of NE release from invading sympathetic axon terminals within the DRG may affect the extent of alpha2 rece


Subject(s)
Neuropeptide Y/metabolism , Pain/physiopathology , Receptors, Neuropeptide Y/metabolism , Spinal Nerves/physiopathology , Tyrosine 3-Monooxygenase/metabolism , Animals , Autoradiography , Axons/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Immunohistochemistry , In Situ Hybridization , Ligation , Male , Neurons/metabolism , Neuropeptide Y/biosynthesis , Pain/metabolism , Physical Stimulation , Protein Precursors/biosynthesis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Spinal Nerves/metabolism , Spinal Nerves/ultrastructure
10.
Psychopharmacology (Berl) ; 141(3): 225-34, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10027503

ABSTRACT

The experiments explored the nature and time course of changes in behavior and Fos expression in the periaqueductal grey area (PAG) in response to an injection of cocaine that was given following a single episode of social defeat stress. Social defeat stress was defined as an intruder mouse's response to an aggressive resident mouse. First, the intruder was briefly attacked, and secondly, it was threatened while protected by a perforated cage for 20 min. Plasma corticosterone levels rose after the beginning of the confrontation and remained elevated during the protected phase. In a first experiment, separate groups of intruder and control mice were challenged once with cocaine (20, 30, or 40 mg/kg) or saline. During tests for motor activity, behavioral measurements were obtained via (1) photobeam interruptions, (2) tracking of movements via image analysis, and (3) quantitative ethological analysis of postures and acts via videorecords. Several indices of ambulatory or horizontal forward locomotion confirmed the stimulant effects of cocaine. In a further experiment, separate groups of mice were challenged with 40 mg/kg cocaine at one time point, either during the social stress or 3, 5, 7 or 9 days thereafter. A cocaine challenge significantly increased locomotion 5 and 7 days after a brief social defeat stress, in excess of the level that is seen in non-stressed animals. Further experiments used immunohistochemical assays of sections through the caudal ventrolateral PAG and showed a significant increase in Fos-like immunoreactivity (Fos-LI) 1 h after the social stress experience or after cocaine. Importantly, concurrent administration of cocaine with social defeat stress produced inhibition of Fos expression throughout the PAG. A partial to complete recovery of cocaine-induced Fos expression was observed 5-7 days after social defeat stress. The results suggest that a single social stress episode is sufficient to engender a delayed sensitization of stimulant hyperactivity. The initial inhibition of Fos expression by concurrent social stress and cocaine may point to a relevant initiating event in the process of sensitization to stimulants.


Subject(s)
Behavior, Animal/drug effects , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Genes, fos/genetics , Periaqueductal Gray/metabolism , Stress, Psychological/psychology , Aggression/physiology , Animals , Corticosterone/blood , Image Processing, Computer-Assisted , Immunohistochemistry , Interpersonal Relations , Male , Mice , Motor Activity/drug effects , Time Factors
11.
Brain Res ; 810(1-2): 200-10, 1998 Nov 09.
Article in English | MEDLINE | ID: mdl-9813326

ABSTRACT

The objective of the present study was to determine how c-fos gene expression in brainstem structures after a brief episode of social defeat stress is related to behavioral sensitization to cocaine challenge. Social stress was defined as defeat in a brief confrontation with an aggressive resident mouse and subsequent 20-min exposure to the resident's threats behind a protective screen. Mice were treated with cocaine (40 mg/kg, i.p.) immediately or 1 week after social defeat stress. Fos-like immunoreactive (Fos-LI) cell nuclei were analyzed in the ventral tegmental area (VTA), dorsal raphe nucleus (DR), periaqueductal grey area (PAG) and locus coeruleus (LC). One episode of social stress induced behavioral sensitization to cocaine as indicated by an augmented locomotor response to a challenge injection 7 days after a single defeat. In naive mice, social stress markedly increased the number of Fos-LI nuclei in the DR, PAG and LC, but not in the VTA. Similarly, cocaine administration resulted in a significantly increased number of Fos-LI nuclei in the same areas. Administration of cocaine immediately following social defeat significantly reduced the number of Fos-LI nuclei in the DR, PAG and LC. Cocaine-induced Fos expression returned in the PAG and DR, but not in the LC, 1 week after social stress. In conclusion, the present results suggest that the presence of brainstem Fos be related to the ability to express stress-induced behavioral sensitization to cocaine.


Subject(s)
Anesthetics, Local/pharmacology , Behavior, Animal/drug effects , Brain Stem/metabolism , Cocaine/pharmacology , Proto-Oncogene Proteins c-fos/biosynthesis , Social Behavior , Stress, Psychological/psychology , Animals , Brain Stem/drug effects , Image Interpretation, Computer-Assisted , Immunohistochemistry , Male , Mice , Mice, Inbred Strains , Motor Activity/drug effects , Motor Activity/physiology , Stress, Psychological/metabolism
12.
Anesthesiology ; 89(4): 961-8, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9778014

ABSTRACT

BACKGROUND: This study determined the induction profiles of immediate-early genes in the ovine brain after cardiopulmonary bypass (CPB) and hypothermic circulatory arrest (HCA), and the effects of the noncompetitive N-methyl-D-aspartate antagonist, aptiganel, on immediate-early gene expression, neuronal necrosis, and functional outcome. METHODS: Cannulas were inserted into isoflurane-anesthetized neonatal lambs undergoing CPB. One group received 2.5 mg/kg intravenous aptiganel. Animals underwent 90 or 120 min of HCA at 16 degrees C, were rewarmed to 38 degrees C, and were weaned from CPB. One hour after CPB was discontinued, brain perfusion was fixed and removed for immunohistochemical analysis in one half of the animals. The other half survived 2 or 3 days before their brains were evaluated for neuronal degeneration. Data were analyzed using analysis of variance; P < 0.05 was considered significant. RESULTS: Cardiopulmonary bypass and HCA differentially induced c-Jun and Fos proteins in the hippocampal formation, with c-Jun expression increasing with the duration of HCA, whereas Fos protein expressions were greatest after 90 min of HCA. The c-Jun protein was expressed in all neurons except the dentate gyrus. The Fos proteins were expressed in all neurons, including the dentate gyrus. Neuronal necrosis was observed in CA1 (73%) and CA3 (29%) neurons but not in the dentate gyrus after 120 min of HCA. Aptiganel completely inhibited c-Jun expression (P < 0.001) but not Fos, improved functional outcome, and attenuated neuronal necrosis (P < 0.05). CONCLUSIONS: The c-Jun and c-Fos proteins are expressed differentially in hippocampal neurons after CPB and HCA. Expression of c-Jun is associated with neuronal necrosis, whereas Fos protein expression is associated with survival. Aptiganel inhibits c-Jun expression, attenuates neuronal necrosis, and improves outcome.


Subject(s)
Brain/metabolism , Brain/pathology , Cardiopulmonary Bypass , Genes, Immediate-Early , Heart Arrest, Induced , Hypothermia, Induced , Nerve Degeneration/metabolism , Anesthesia, General , Anesthetics, Inhalation , Animals , Brain/physiology , Female , Gene Expression Regulation , Guanidines/pharmacology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Isoflurane , Necrosis , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-jun/biosynthesis , Sheep
13.
Peptides ; 19(6): 1115-7, 1998.
Article in English | MEDLINE | ID: mdl-9700764

ABSTRACT

This study examines the role of carboxypeptidase E (CPE) in processing pro tachykinin to form the final bioactive amidated undecapeptide, substance P (SP) in various rat brain regions. Cpe(fat)/Cpe(fat) mice brain tissue was analyzed for total SP forms (including intermediates), and final amidated SP was compared to Cpe+/Cpe+ and Cpe+/Cpe- controls. In all brain regions tested by radioimmunoassay, amidated fully processed SP was more than fivefold lower in Cpe(fat)/Cpe(fat) mice than in controls whereas total SP species levels were unchanged. This demonstrates that CPE is required for normal SP proteolytic processing. Substance P has numerous functions in the brain; therefore, SP deficiency due to the CPE mutation may contribute to the obese phenotype or even to other phenotypes not yet described in Cpe(fat)/Cpe(fat) mice.


Subject(s)
Brain/metabolism , Carboxypeptidases/genetics , Substance P/metabolism , Animals , Carboxypeptidase H , Carboxypeptidases/metabolism , Mice , Mutation , Radioimmunoassay , Rats
14.
Br J Urol ; 81(2): 224-8, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9488063

ABSTRACT

OBJECTIVES: To determine whether substance P (SP, also known as neurokinin 1, NK1) receptors are differentially expressed in bladder biopsies from patients with interstitial cystitis (IC) compared with matched controls. MATERIALS AND METHODS: Cold-cup biopsies were taken during routine diagnostic cystoscopy. NK1-receptor expression was assessed using a quantitative analysis of NK1-receptor-encoding mRNA in bladder biopsies from patients and controls using in situ hybridization histochemistry (ISHH) combined with autoradiographic image analysis. RESULTS: Autoradiographic signal indicating the presence of NK1-encoding mRNA was localized to detrusor muscle, urothelium and vascular structures. In the bladder vasculature, the signal was predominantly associated with endothelial cells. NK1 receptor-encoding mRNA within the vascular endothelium was increased in the biopsies obtained from patients with IC. CONCLUSION: Increased levels of NK1 receptor-encoding mRNA within the bladder vascular endothelium suggests the up-regulation of NK1 receptor as a putative factor in the pathogenesis of pain related to IC. Increased responsiveness to SP released from the perivascular sensory terminals may result in a local cascade of neurogenic inflammatory responses which trigger the pathophysiological changes, including pain, characteristic of IC.


Subject(s)
Cystitis, Interstitial/metabolism , Receptors, Neurokinin-1/metabolism , Adult , Biopsy , Cystitis, Interstitial/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Histocytochemistry , Humans , Middle Aged , RNA, Messenger/metabolism
15.
Ann Thorac Surg ; 64(4): 1082-7; discussion 1088, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9354532

ABSTRACT

BACKGROUND: Altered gene expression occurs in the brain after global ischemia. We have developed a model to examine the effects of cardiopulmonary bypass and hypothermic circulatory arrest (HCA) on the induction of the immediate-early gene c-fos in the brains of neonatal lambs. We then tested the effects of the noncompetitive N-methyl-D-aspartate antagonist, aptiganel hydrochloride (Cerestat), on c-fos expression and neuronal injury. METHODS: Neonatal lambs (weight, 4 to 6 kg) anesthetized with isoflurane were supported by cardiopulmonary bypass, subjected to 90 or 120 minutes of HCA at 15 degrees C, and rewarmed on bypass to 38 degrees C. One hour after cardiopulmonary bypass was terminated, the brains were perfusion fixed and removed for in situ hybridization and immunohistochemical analysis. Some animals survived 3 days before their brains were removed to examine for neuronal necrosis. One group of lambs (n = 20) received aptiganel (2.5 mg/kg). A second group (n = 25) received saline vehicle only. RESULTS: Increasing duration of HCA induced a corresponding increase in c-fos messenger RNA expression throughout the hippocampal formation and cortex. However, Fos protein synthesis peaked after 90 minutes of HCA and decreased significantly (p < 0.01) after 120 minutes of HCA. Aptiganel administration caused a significant decrease in (p < 0.001) c-fos messenger RNA expression and Fos protein synthesis after 90 minutes of HCA and preserved Fos protein synthesis after 120 minutes of HCA. Neuronal necrosis was observed in the brains of vehicle-treated lambs after 120 minutes of HCA but was significantly decreased (p < 0.05) in the lambs given aptiganel. CONCLUSIONS: These experiments indicate that the transcriptional processes of immediate-early genes remain intact, whereas translational processes are impaired after prolonged HCA. The inability to synthesize Fos proteins after 120 minutes of HCA was associated with neuronal degeneration. Aptiganel preserved translational processes and caused a significant improvement in the neurologic outcome.


Subject(s)
Gene Expression/drug effects , Genes, fos , Guanidines/pharmacology , Heart Arrest, Induced , Hippocampus/pathology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Animals, Newborn , Cardiopulmonary Bypass , Cell Death/drug effects , Hippocampus/metabolism , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/genetics , Sheep
16.
Oral Dis ; 3 Suppl 1: S58-63, 1997 May.
Article in English | MEDLINE | ID: mdl-9456659

ABSTRACT

OBJECTIVE: Human saliva is known to decrease HIV infectivity in vitro. The purpose of this study was to extend these findings and to focus on the mechanism of action of these salivary factor(s). DESIGN: A number of viruses and several assay systems have been utilized to determine if the effect of submandibular saliva is directly on the virus, on the host cell, or on the virus-cell interaction. MATERIALS AND METHODS: Submandibular saliva from seronegative donors was incubated with HIV-1, other retroviruses, or unrelated viruses. Viral infectivity was monitored either by determining p24 antigen levels in peripheral blood mononuclear cells or Sup T1 cells, or using HeLa cells expressing CD4 and an HIV derived long terminal repeat linked to the beta-galactosidase gene. RESULTS: The inhibition of viral infectivity by submandibular saliva is specific for HIV-1. While inhibition increases with time of incubation of saliva with virus, pretreatment of cells with saliva does not inhibit HIV production, and saliva has only modest inhibitory effects when added to HIV-infected cells. CONCLUSIONS: It appears that the effect of submandibular saliva on decreasing the infectivity of HIV-1 is directly on the virus, rather than on the host cell.


Subject(s)
Antiviral Agents , HIV-1/pathogenicity , Saliva/physiology , HeLa Cells/immunology , HeLa Cells/virology , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Saliva/virology , Submandibular Gland/metabolism , Virulence , Virus Cultivation
17.
Pharmacology ; 54(2): 92-7, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9088042

ABSTRACT

Prior studies in rodents have shown significant depletion of reduced glutathione (GSH) in peripheral organs following acute systemic or central administration of opioids. However, little information exists on whether opioid administration affects concentrations of brain GSH. Recently, clinical observations have indicated acute declines of GSH concentrations in the cerebrospinal fluid of cancer patients after acute intracerebroventricular (ICV) morphine which may contribute to the development of organic behavioral brain syndromes associated with central opioid analgesia. Collectively these data led us to investigate the affect of acute systemic and central morphine on regional concentrations of GSH in rat brain. Systemic morphine had no effect on GSH concentrations in selected brain areas. In contrast, ICV morphine resulted in selective GSH depletion in the caudate nucleus, consistent with concurrent excitatory locomotive behavior. This change may have reflected morphine-induced oxidative stress together with increased metabolic activity within the extrapyramidal system.


Subject(s)
Analgesics, Opioid/pharmacology , Brain Chemistry/drug effects , Glutathione/metabolism , Morphine/pharmacology , Analgesics, Opioid/administration & dosage , Animals , Chromatography, High Pressure Liquid , Infusions, Parenteral , Injections, Intraventricular , Male , Morphine/administration & dosage , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley
18.
Anesthesiology ; 85(6): 1439-46, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8968192

ABSTRACT

BACKGROUND: Cardiopulmonary bypass (CPB) and hypothermic circulatory arrest (HCA) are associated with neurological injury. Altered immediate-early gene expression occurs rapidly in the brain in response to ischemia, hypoxia, and severe metabolic stress, which results in long-term changes in the molecular phenotype of neurons. This study determined the effects of CPB and HCA on the expression of the immediate-early gene c-fos. METHODS: Neonatal lambs were subjected to 2 h of CPB at 38 degrees C (n = 4) or 60 min (n = 6), 90 min (n = 7), and 120 min (n = 6) of HCA at 15 degrees C. One hour after terminating CPB at 38 degrees C, the brains were analyzed for FOS-encoding mRNA and FOS-like immunoreactivity in the hippocampal formation. Other animals (n = 15), subjected to the same CPB and HCA protocol, were allowed to survive 3-5 days before their brains were examined for dead neurons. RESULTS: Minimal c-fos mRNA and FOS proteins were observed in neurons of animals subjected to normothermic bypass and of those that served as controls. Non-neuronal FOS proteins were observed in the choroid plexus, ependyma, and blood vessels at all times, including normothermic CPB, but not in the control animals without CPB. The magnitude of c-fos mRNA expression in hippocampal neurons increased directly with the duration of HCA. In contrast, expression of FOS proteins peaked after 90 min of HCA and declined significantly thereafter. Dead neurons were seen in surviving animals after 2 h of HCA only. CONCLUSIONS: Cardiopulmonary bypass and HCA alter immediate-early gene expression in the brain. Translational processes are impaired after 120 min of HCA and correlate with neuron death in the hippocampus.


Subject(s)
Cardiopulmonary Bypass/adverse effects , Genes, fos , Hippocampus/pathology , Hypothermia, Induced/adverse effects , Animals , Cell Death , Gene Expression , Hippocampus/metabolism , In Situ Hybridization , Interneurons/pathology , Models, Biological , Protein Biosynthesis , Sheep
19.
Proc Natl Acad Sci U S A ; 93(18): 9858-63, 1996 Sep 03.
Article in English | MEDLINE | ID: mdl-8790421

ABSTRACT

Olfactory marker protein (OMP) is an abundant, phylogentically conserved, cytoplasmic protein of unknown function expressed almost exclusively in mature olfactory sensory neurons. To address its function, we generated OMP-deficient mice by gene targeting in embryonic stem cells. We report that these OMP-null mice are compromised in their ability to respond to odor stimull, providing insight to OMP function. The maximal electroolfactogram response of the olfactory neuroepithelium to several odorants was 20-40% smaller in the mutants compared with controls. In addition, the onset and recovery kinetics following isoamyl acetate stimulation are prolonged in the null mice. Furthermore, the ability of the mutants to respond to the second odor pulse of a pair is impaired, over a range of concentrations, compared with controls. These results imply that neural activity directed toward the olfactory bulb is also reduced. The bulbar phenotype observed in the OMP-null mouse is consistent with this hypothesis. Bulbar activity of tyrosine hydroxylase, the rate limiting enzyme of catecholamine biosynthesis, and content of the neuropeptide cholecystokinin are reduced by 65% and 50%, respectively. This similarity to postsynaptic changes in gene expression induced by peripheral olfactory deafferentation or naris blockade confirms that functional neural activity is reduced in both the olfactory neuroepithelium and the olfactory nerve projection to the bulb in the OMP-null mouse. These observations provide strong support for the conclusion that OMP is a novel modulatory component of the odor detection/signal transduction cascade.


Subject(s)
Gene Deletion , Nerve Tissue Proteins/genetics , Neurons, Afferent/physiology , Olfactory Bulb/physiology , Animals , Base Sequence , Cell Line , Electrophysiology , Female , Male , Mice , Molecular Sequence Data , Nerve Tissue Proteins/physiology , Olfactory Bulb/cytology , Olfactory Marker Protein , Restriction Mapping
20.
Neurosci Lett ; 211(2): 77-80, 1996 Jun 21.
Article in English | MEDLINE | ID: mdl-8830848

ABSTRACT

Complications arising from diabetes mellitus include hormonal dysfunctions such as impairment in the regulation of gonadatroph and corticotroph secretion. Preprotachykinin (PPT) mRNA encoding the peptide substance P (SP), has been localized in the anterior pituitary. The goal of this study was to determine if streptozotocin (STZ)-induced diabetes affects the SP content or PPT mRNA level in the pituitary of male rats. We injected STZ (55 mg/kg) to 6-week-old rats which developed hyperglycemia (blood glucose > 400 mg/dl) by 6 weeks post-injection. SP-like immunoreactivity in the pituitary dropped 54%. In situ hybridization was performed using a PPT-specific oligonucleotide with signal intensity differences semi-quantified using an image analysis system. Normal pituitary had a regional distribution of PPT mRNA, with no detectable signal in the posterior or intermediate lobes, while the anterior lobe displayed a distinctive pattern of labeled cells arranged in clusters. In diabetic rats there was a 23% decrease in the PPT-mRNA hybridization signal compared to controls (P < 0.05). The changes observed in PPT gene expression and SP content may be additional factors participating in the hormonal complications seen in diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Pituitary Gland/metabolism , Protein Precursors/biosynthesis , RNA, Messenger/biosynthesis , Substance P/metabolism , Tachykinins/biosynthesis , Animals , Gene Expression Regulation/drug effects , Hypoglycemic Agents/pharmacology , Image Processing, Computer-Assisted , In Situ Hybridization , Insulin/pharmacology , Male , Oligonucleotide Probes , Pituitary Gland/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...