Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Mol Psychiatry ; 23(3): 544-555, 2018 03.
Article in English | MEDLINE | ID: mdl-29038598

ABSTRACT

Neurodevelopmental disorders, including autism spectrum disorders, are highly male biased, but the underpinnings of this are unknown. Striatal dysfunction has been strongly implicated in the pathophysiology of neurodevelopmental disorders, raising the question of whether there are sex differences in how the striatum is impacted by genetic risk factors linked to neurodevelopmental disorders. Here we report male-specific deficits in striatal function important to reward learning in a mouse model of 16p11.2 hemideletion, a genetic mutation that is strongly associated with the risk of neurodevelopmental disorders, particularly autism and attention-deficit hyperactivity disorder. We find that male, but not female, 16p11.2 deletion animals show impairments in reward-directed learning and maintaining motivation to work for rewards. Male, but not female, deletion animals overexpress mRNA for dopamine receptor 2 and adenosine receptor 2a in the striatum, markers of medium spiny neurons signaling via the indirect pathway, associated with behavioral inhibition. Both sexes show a 50% reduction of mRNA levels of the genes located within the 16p11.2 region in the striatum, including the kinase extracellular-signal related kinase 1 (ERK1). However, hemideletion males show increased activation in the striatum for ERK1, both at baseline and in response to sucrose, a signaling change associated with decreased striatal plasticity. This increase in ERK1 phosphorylation is coupled with a decrease in the abundance of the ERK phosphatase striatum-enriched protein-tyrosine phosphatase in hemideletion males. In contrast, females do not show activation of ERK1 in response to sucrose, but notably hemideletion females show elevated protein levels for ERK1 as well as the related kinase ERK2 over what would be predicted by mRNA levels. These data indicate profound sex differences in the impact of a genetic lesion linked with neurodevelopmental disorders, including mechanisms of male-specific vulnerability and female-specific resilience impacting intracellular signaling in the brain.


Subject(s)
Corpus Striatum/metabolism , Learning/physiology , Neurodevelopmental Disorders/genetics , Animals , Autism Spectrum Disorder/metabolism , Autistic Disorder/genetics , Chromosome Deletion , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , MAP Kinase Signaling System/genetics , Male , Mice , Mitogen-Activated Protein Kinase 3/genetics , Motivation/genetics , Neurodevelopmental Disorders/metabolism , Phosphorylation , Reward , Sex Factors , Signal Transduction/genetics
2.
Neuroscience ; 226: 165-77, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-22999971

ABSTRACT

The development of tolerance to the antinociceptive effects of morphine has been associated with networks within ventrolateral periaqueductal gray (vlPAG) and separately, nitric oxide signaling. Furthermore, it is known that the mechanisms that underlie tolerance differ with age. In this study, we used a rat model of antinociceptive tolerance to morphine at two ages, postnatal day (PD) 7 and adult, to determine if changes in the vlPAG related to nitric oxide signaling produced by chronic morphine exposure were age-dependent. Three pharmacological groups were analyzed: control, acute morphine, and chronic morphine group. Either morphine (10mg/kg) or equal volume of normal saline was given subcutaneously twice daily for 6½ days. Animals were analyzed for morphine dose-response using Hot Plate test. The expression of several genes associated with nitric oxide metabolism was evaluated using rtPCR. In addition, the effect of morphine exposure on immunohistochemistry for Fos, and nNOS as well as nicotinamide adenine dinucleotide phosphate diaphorase (NADPH-d) reaction at the vlPAG were measured. In both age groups acute morphine activated Fos in the vlPAG, and this effect was attenuated by chronic morphine, specifically in the vlPAG at the level of the laterodorsal tegmental nucleus (LDTg). In adults, but not PD7 rats, chronic morphine administration was associated with activation of nitric oxide function. In contrast, changes in the gene expression of PD7 rats suggested superoxide and peroxide metabolisms may be engaged. These data indicate that there is supraspinal neuroplasticity following morphine administration as early as PD7. Furthermore, oxidative stress pathways associated with chronic morphine exposure appear age-specific.


Subject(s)
Aging/physiology , Morphine/pharmacology , Narcotics/pharmacology , Neuronal Plasticity/drug effects , Oxidative Stress/physiology , Periaqueductal Gray/drug effects , Animals , Animals, Newborn , Cell Count , Dose-Response Relationship, Drug , Female , Gene Expression/drug effects , Genes, fos/drug effects , Hot Temperature , Immunohistochemistry , NADPH Dehydrogenase/metabolism , Neurons/drug effects , Nitric Oxide Synthase Type I/metabolism , Oxidative Stress/drug effects , Pain Measurement/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley
3.
Neuroscience ; 179: 1-8, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21277949

ABSTRACT

Adolescence is a critical vulnerable period during which exposure to nicotine greatly enhances the possibility to develop drug addiction. Growing evidence suggests that serotonergic (5-HT) neurotransmission may contribute to the initiation and maintenance of addictive behavior. As the dorsal raphe (DR) and median raphe (MnR) nuclei are the primary 5-HT source to the forebrain, the current study tested the hypothesis that there are age-dependent effects of acute nicotine administration on activation of 5-HT neurons within these regions. Both adolescent (Postnatal day 30) and adult (Postnatal day 70) male Sprague-Dawley rats received subcutaneous injection of either saline or nicotine (0.2, 0.4, or 0.8 mg/kg). Subsequently, the number of 5-HT cells that were double-labeled for Fos and tryptophan hydroxylase was counted in seven subregions within the DR and the entire MnR. The results show that acute nicotine injection induces Fos expression in 5-HT neurons in a region-specific manner. In addition, adolescents show broader regional activations at either a lower (0.2 mg/kg) and a higher (0.8 mg/kg) dose of nicotine, displaying a unique U-shape response curve across doses. In contrast, 5-HT cells with activated Fos expression were restricted to fewer regions in adults, and the patterns of expression were more consistent across doses. The results reveal dose-dependent effects of nicotine during adolescence with apparent sensitization at different ends of the dosage spectrum examined compared to adults. These data indicate that initial exposure to nicotine may have unique effects in adolescence on the ascending 5-HT system, with the potential for consequences on the affective-motivational qualities of the drug and the subsequent propensity for repeated use.


Subject(s)
Behavior, Addictive/metabolism , Neurons/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Raphe Nuclei/drug effects , Age Factors , Animals , Behavior, Addictive/physiopathology , Immunohistochemistry , Male , Neurons/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/drug effects , Raphe Nuclei/metabolism , Rats , Rats, Sprague-Dawley , Serotonin/metabolism
4.
Brain Res ; 1314: 175-82, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-19913520

ABSTRACT

Accumulating evidence suggests that the neuropeptide substance P (SP) and its principal receptor neurokinin 1 (NK1) play a specific role in the behavioral response to opioids and stress that may help to initiate and maintain addictive behavior. In animal models, the NK1 receptor is required for opioids to produce their rewarding and motivational effects. SP neurotransmission is also implicated in the behavioral response to stress and in the process of drug sensitization, potentially contributing to vulnerability to addiction or relapse. However, SP neurotransmission only plays a minor role in opioid-mediated antinociception and the development of opioid tolerance. Moreover, the effects of SP on addiction-related behavior are selective for opioids and evidence supporting a role in the response to cocaine or psychostimulants is less compelling. This review will summarize the effects of SP neurotransmission on opioid-dependent behaviors and correlate them with potential contributing neural pathways. Specifically, SP neurotransmission within components of the basal forebrain particularly the nucleus accumbens and ventral pallidum as well as actions within the ascending serotonin system will be emphasized. In addition, cellular- or network-level interactions between opioids and SP signaling that may underlie the specificity of their relationship will be reviewed.


Subject(s)
Brain/physiopathology , Stress, Psychological/physiopathology , Substance P/physiology , Substance-Related Disorders/physiopathology , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Comorbidity , Humans , Neural Pathways/metabolism , Neural Pathways/physiopathology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiopathology , Opioid-Related Disorders/metabolism , Opioid-Related Disorders/physiopathology , Raphe Nuclei/drug effects , Raphe Nuclei/metabolism , Raphe Nuclei/physiopathology , Stress, Psychological/metabolism , Substance-Related Disorders/metabolism
5.
Neuroscience ; 166(2): 720-9, 2010 Mar 17.
Article in English | MEDLINE | ID: mdl-20026253

ABSTRACT

It is poorly understood if and how pain may modify the effect of opioids on neural systems that contribute to reward and addictive behavior. We hypothesized that the activation of ascending dopaminergic and serotonergic nuclei by morphine is modified by the presence of noxious stimulation. Immunohistochemical double-labeling technique with Fos was used to examine if an intraplantar formalin injection, an acute noxious input, changed the effect of morphine on dopaminergic neurons of the ventral tegmental area (VTA), and serotonergic neurons of the dorsal raphe nucleus (DR). Four groups of rats were analyzed: (1) control injected with normal saline s.c., (2) rats treated with formalin into the hind paw 30 min after normal saline injection, (3) rats injected with morphine sulfate s.c., and (4) rats treated with formalin into the hind paw 30 min after morphine injection (morphine/formalin). Following morphine injection, there was an increase in the number of dopaminergic neurons in the VTA with Fos immunolabeling. However, noxious stimulation did not detectably change morphine's effect on Fos expression in VTA dopamine neurons. In contrast, the number of serotonergic neurons containing Fos was increased in the morphine/formalin group compared to all other groups and this effect was topographically selective for the dorsal area of the DR at mid rostro-caudal levels. Therefore, morphine's activation of the VTA, which is associated with motivated behavior and reward seeking, appears similar in the context of pain. However, activation of the ascending serotonin system, which influences mood and has the capacity to modify reward pathways, appears different. In addition, these findings reveal interactions between nociceptive signaling and opioids that contrasts with the notion that opioids simply block access of nociceptive signaling to supraspinal structures.


Subject(s)
Dopamine/metabolism , Mesencephalon/drug effects , Mesencephalon/metabolism , Morphine/pharmacology , Pain/metabolism , Serotonin/metabolism , Analysis of Variance , Animals , Cell Count , Immunohistochemistry , Male , Microscopy, Fluorescence , Neurons/drug effects , Neurons/metabolism , Pain Measurement , Physical Stimulation , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley
6.
Neurosci Lett ; 464(3): 188-92, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19699779

ABSTRACT

The third vesicular glutamate transporter (VGLUT3) is expressed in a subset of cholinergic and GABAergic neurons in the forebrain. In this study the distribution of VGLUT3 was mapped in relation to the receptor for substance P, neurokinin 1 (NK1), which has been independently reported within cholinergic and GABAergic neurons in a similar distribution. Dual immunofluorescence labeling techniques were used, sometimes in combination with triple labeling for the vesicular acetylcholine transporter (VAChT), to identify cholinergic cells. Virtually all cells immunolabeled for VGLUT3 in the nucleus accumbens core and shell regions, ventral pallidum, olfactory tubercle and caudate putamen were cholinergic and also contained immunolabeling for the NK1 receptor. In the hippocampal formation where VGLUT3 has been described in GABAergic neurons, colocalization between NK1 and VGLUT3 was also common but less complete. Cells double labeled for NK1 and VGLUT3 were most prevalent in stratum radiatum in the CA1 subfield. In the habenula VGLUT3 was also found within NK1 receptor immunolabeled neurons. However, there were some areas where neurons containing these two proteins were separate populations including the cerebral cortex and median raphe nucleus. These results reveal a trend for VGLUT3 to localize within neurons containing the NK1 receptor in several areas of the forebrain.


Subject(s)
Prosencephalon/metabolism , Receptors, Neurokinin-1/metabolism , Vesicular Glutamate Transport Proteins/metabolism , Animals , Microscopy, Fluorescence , Neurons/metabolism , Prosencephalon/anatomy & histology , Rats , Vesicular Acetylcholine Transport Proteins/metabolism
7.
J Chem Neuroanat ; 38(4): 273-81, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19467322

ABSTRACT

The serotonergic dorsal raphe nucleus (DR) is an area enriched with cell bodies and axons containing the third vesicular glutamate transporter, VGLUT3. However, the role of VGLUT3-containing axons in modulating activity of serotonin (5-HT) neurons within the DR remains poorly understood. In this study, neurochemical features and topography of VGLUT3-containing cell bodies and axons in the DR were examined. Since many 5-HT cells have been reported to express VGLUT3, the distribution of dually labeled axons was examined within the DR. Axons containing both VGLUT3 and 5-HT immunolabeling had a topographic distribution: they innervated the ependyma and ramified within the caudal DR at the base of the aqueduct, an area known to give rise to ependymal innervation. Thus VGLUT3 is only present in a specific subcomponent of recurrent 5-HT axon collaterals. Remaining VGLUT3 axons were only rarely dually immunolabeled for markers of monoamines, GABA, or acetycholine, suggesting these axons contain a predominance of glutamate-filled vesicles. Since the substance P receptor, neurokinin 1 (NK1), has previously been associated with local glutamate neurons in the DR, the relationship between NK1, 5-HT and VGLUT3 cells was examined using triple-immunolabeling. Results indicate that the majority of non-5-HT VGLUT3-containing cell bodies in the DR contain NK1 immunolabeling. Taken together, these findings indicate locally collateralizing glutamate neurons responsive to substance P contain VGLUT3.


Subject(s)
Glutamic Acid/metabolism , Mesencephalon/metabolism , Neurons/metabolism , Raphe Nuclei/metabolism , Substance P/metabolism , Vesicular Glutamate Transport Proteins/metabolism , Animals , Axons/drug effects , Axons/metabolism , Axons/ultrastructure , Brain Mapping , Ependyma/cytology , Ependyma/metabolism , Immunohistochemistry , Male , Mesencephalon/cytology , Mesencephalon/drug effects , Neural Pathways/cytology , Neural Pathways/drug effects , Neural Pathways/metabolism , Neurons/cytology , Neurons/drug effects , Raphe Nuclei/cytology , Raphe Nuclei/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/drug effects , Receptors, Neurokinin-1/metabolism , Serotonin/metabolism , Substance P/pharmacology , Synaptic Transmission/physiology
8.
Neuroscience ; 153(3): 851-9, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18403129

ABSTRACT

Nicotinic acetylcholine receptors containing the alpha4 and beta2 subunits constitute the most abundant high-affinity binding site of nicotine in the brain and are critical for the addictive qualities of nicotine. 5-HT neurotransmission is thought to be an important contributor to nicotine addiction. Therefore in this study it was examined how alpha4-containing receptors are positioned to modulate the function of 5-HT neurons using ultrastructural analysis of immunolabeling for the alpha4 receptor subunit in the dorsal raphe nucleus (DR), a primary source of forebrain 5-HT in the rat. Of 150 profiles labeled for the alpha4 subunit, 140 or 93% consisted of either soma or dendrites, these were often small-caliber (distal) dendrites <1.5 microm in diameter (63/150 or 42%). The majority (107/150 or 71%) of profiles containing labeling for alpha4 were dually labeled for the synthetic enzyme for 5-HT, tryptophan hydroxylase (TPH). Within dendrites immunogold labeling for alpha4 was present on the plasma membrane or near postsynaptic densities. However, labeling for alpha4 was commonly localized to the cytoplasmic compartment often associated with smooth endoplasmic reticulum, plausibly representing receptors in transit to or from the plasma membrane. Previous studies have suggested that nicotine presynaptically regulates activity onto 5-HT neurons, however alpha4 immunolabeling was detected in only 10 axons in the DR or 7% of profiles sampled. This finding suggest that alpha4 containing receptors are minor contributors to presynaptic regulation of synaptic activity onto 5-HT neurons, but rather alpha4 containing receptors are positioned to influence 5-HT neurons directly at postsynaptic sites.


Subject(s)
Neurons/ultrastructure , Raphe Nuclei/ultrastructure , Receptors, Nicotinic/metabolism , Synaptic Membranes/ultrastructure , Animals , Fluorescent Antibody Technique , Immunohistochemistry , Microscopy, Electron, Transmission , Neurons/metabolism , Raphe Nuclei/metabolism , Rats , Serotonin/metabolism , Synaptic Membranes/metabolism
9.
Neuroscience ; 145(3): 1077-86, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17289275

ABSTRACT

Stress causes increased dynorphin (DYN) expression in limbic brain regions and antagonism of kappa-opioid receptors may offer therapeutic potential for the treatment of depression. A potential site of DYN action relevant to stress and related neuropsychiatric disorders is the locus coeruleus (LC), the primary source of forebrain norepinephrine. Therefore, using immunofluorescence and immunoelectron microscopic analyses, we characterized the cellular substrates for interactions between DYN and tyrosine hydroxylase (TH), a catecholamine synthesizing enzyme in single sections through the rat LC. Light microscopic analysis of DYN immunoreactivity indicated that DYN fibers are distributed within the core and pericoerulear subregions of the LC. Using electron microscopy, immunoperoxidase labeling for DYN was primarily found in axon terminals, although in some cases was diffusely localized to somatodendritic processes. When DYN-containing axons formed synaptic contacts, they typically (89%) exhibited an asymmetric morphology. Almost a third (28%) of the postsynaptic targets of DYN-containing axons contained immunogold labeling for TH. These findings reveal some diversity as to the localization of DYN in the LC within axons that contact both TH and non-TH containing dendrites. However, the present data provide the first ultrastructural evidence that DYN-containing axon terminals directly innervate catecholaminergic LC dendrites. Moreover, DYN axon terminals targeting catecholaminergic LC dendrites via asymmetric synapses are consistent with localization within excitatory type afferents to the LC. Therefore, direct modulation of catacholaminergic LC neurons maybe an important site of action for DYN relevant to stress and stress-related disorders.


Subject(s)
Axons/physiology , Dynorphins/physiology , Locus Coeruleus/physiology , Norepinephrine/physiology , Animals , Immunohistochemistry , Locus Coeruleus/physiopathology , Locus Coeruleus/ultrastructure , Male , Microscopy, Confocal , Microscopy, Electron , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism
10.
Neuropeptides ; 39(1): 1-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15627494

ABSTRACT

The dorsal raphe nucleus (DR) contains serotonin (5-HT) neurons that innervate the cortex and limbic system and through these projections is thought to regulate cognition and behavior. Clinical and pharmacological findings implicate dysfunctions in the DR-5-HT system in affective disorders, including anxiety, depression and suicide. Although the DR is often considered in light of its 5-HT neurons, recent studies underscore the complexity of this nucleus and its heterogeneous nature. Of particular interest, are peptides that are either present within neurons in the DR, innervate DR-5-HT neurons or act upon local circuitry within the DR to indirectly impact on this 5-HT system. These peptides are positioned to fine-tune the activity of selective groups of serotonergic neurons within the DR and thereby 5-HT release in different terminal fields. This review will focus on substance P and corticotropin-releasing factor as two peptides that act independently and interdependently to influence DR-5-HT function. The role of non-serotonergic components of the DR in translating the effect of each of these peptides is discussed. This synthesis refines our views on the regulation of the DR-5-HT system and importantly, gives insight into mechanisms of endogenous control of DR function, the dysregulation of which may contribute to pathophysiology.


Subject(s)
Brain Chemistry/physiology , Neuropeptides/physiology , Raphe Nuclei/physiology , Serotonin/physiology , Animals , Humans
11.
Brain Res ; 935(1-2): 87-102, 2002 May 10.
Article in English | MEDLINE | ID: mdl-12062477

ABSTRACT

Several lines of evidence have suggested that mu-opioids, generally regarded as inhibitory, also have effects that stimulate neural activity. To look for possible excitatory opioid action in the rat periaqueductal gray (PAG), we first re-examined data from a previous study and found that met-enkephalin could evoke a delayed, sluggish excitation, suggestive of modulation by the opioid on the action of certain excitants. This observation, coupled with other studies that show mu-opioids can modulate NMDA receptor activation, prompted us to perform extracellular recording of the responses of single ventrolateral PAG (vlPAG) neurons in brain slices to DAMGO, a mu-opioid, and to NMDA. When applied alone, DAMGO at nM concentrations, like met-enkephalin, often evoked the delayed excitation and occasionally an inhibition. When applied after a brief exposure to NMDA, DAMGO at doses as low as 0.1 nM potentiated the excitation produced by a subsequent pulse of NMDA. This occurred, depending on cell type, in 23-100% of vlPAG neurons. The potentiating action of DAMGO was blocked by naloxone, suggesting it was mediated by mu-opioid receptors. Characterization of these mu-opioid actions revealed that the potentiation and the delayed excitation, unlike the inhibition, was not blocked by another opioid antagonist, nalmefene, nor by an inhibitor of the G protein of the G(i) class, N-ethylmaleimide. Moreover, the potentiating action was distinct from the inhibition in that it was: (a) enhanced by repeated opioid applications, (b) exhibited low effective doses, (c) had a long time course (minutes to develop and last tens of minutes) and (d) was present in distinct though overlapping cell populations. These data reveal an unconventional action of opioids in PAG neurons, that is, a potentiation of excitation produced by NMDA. This effect appeared mechanistically distinct from opioid inhibition or disinhibition and may be related to established examples of direct opioid excitation. These observations may help understanding behaviorally important mechanisms linked to acute and chronic opioid functions in the vlPAG.


Subject(s)
Action Potentials/physiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , N-Methylaspartate/pharmacology , Naltrexone/analogs & derivatives , Periaqueductal Gray/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Opioid, mu/metabolism , Synaptic Transmission/physiology , Action Potentials/drug effects , Analgesics, Opioid/pharmacology , Animals , Dose-Response Relationship, Drug , Drug Interactions/physiology , Enkephalin, Methionine/pharmacology , Enzyme Inhibitors/pharmacology , Ethylmaleimide/pharmacology , Female , Male , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neural Pathways/cytology , Neural Pathways/drug effects , Neural Pathways/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Organ Culture Techniques , Periaqueductal Gray/cytology , Periaqueductal Gray/drug effects , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, Opioid, mu/drug effects , Synaptic Transmission/drug effects
12.
Neuroscience ; 108(3): 467-77, 2001.
Article in English | MEDLINE | ID: mdl-11738260

ABSTRACT

Chronic administration of mu-opioid receptor agonists is known to produce adaptive changes within noradrenergic neurons of the locus coeruleus. Although mu-opioid receptors are densely expressed by locus coeruleus neurons, the effects of acute and chronic administration of agonists on the subcellular distribution of mu-opioid receptors remain poorly understood. Therefore, we examined the ultrastructural distribution of mu-opioid receptor immunoreactivity in the locus coeruleus of rats subjected to either acute morphine, or etorphine, or chronic morphine treatment. In the locus coeruleus of control rats receiving acute saline injections or placebo pellet implants, immunogold-silver labeling for mu-opioid receptors was localized to parasynaptic and extrasynaptic portions of the plasma membranes of perikarya and dendrites. Only 8% of the gold-silver particles analyzed were distributed within the cytoplasm of dendrites and perikarya in vehicle-treated rats. Immunolabeling for mu-opioid receptors was distributed along portions of the plasma membrane that were often apposed by astroglial sheaths. After acute injections of etorphine, there was a dramatic internalization of mu-opioid receptors to intracellular compartments. Quantitative analysis of gold-silver particles indicative of mu-opioid receptors showed that a substantial number of gold particles shifted from the plasma membrane to early endosomes in dendrites from etorphine-treated rats. In dendrites sampled from etorphine-treated rats, 85% of the gold-silver grains indicative of mu-opioid receptor labeling were located in intracellular compartments as compared to 15% that were distributed along the plasma membrane. In animals that received either acute morphine injections or chronic morphine via pellet implantation, no change in the subcellular distribution of immunogold particles indicative of mu-opioid receptors was detected when compared to matched control animals. These results provide the first ultrastructural evidence that mu-opioid receptors are internalized by agonists such as etorphine, but not the partial agonist morphine, in the locus coeruleus.


Subject(s)
Etorphine/pharmacology , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Narcotics/pharmacology , Receptors, Opioid, mu/metabolism , Animals , Biological Transport/drug effects , Etorphine/administration & dosage , Immunohistochemistry , Locus Coeruleus/ultrastructure , Male , Morphine/administration & dosage , Morphine/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Time Factors
13.
J Chem Neuroanat ; 21(1): 53-62, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11173220

ABSTRACT

The ventromedial nucleus of the hypothalamus (VMN) regulates the estrogen-dependent appearance of female mating behavior, lordosis. Accumulating evidence suggests that estrogen might exert its control over lordosis by acting, in part, on neurons that contain enkephalin in the VMN. The expression of the enkephalin precursor gene is robustly stimulated by estrogen and is correlated with the later appearance of lordosis. GABA has also been implicated as an important neurotransmitter for the appearance of lordosis. Because enkephalin is thought to act in several brain areas to modulate the activity of GABAergic neurons, we studied the ultrastructural morphology and relationship between neurons containing these neurochemicals using dual-labeling immunocytochemistry in ovariectornized rats, half of which received estrogen replacement. Immunolabeling for enkephalin was almost always detected within axon terminals (695 axonal profiles sampled), while GABA immunoreactivity was more often localized to cell bodies and dendrites (191 profiles), than to axons (63 profiles). Axon terminals containing enkephalin immunolabeling provided a major innervation to soma or dendrites containing GABA. That is, over one third (94/245) of the axon terminals in contact with GABA-immunoreactive dendrites contained enkephalin. Furthermore, these GABA-immunoreactive dendrites accounted for a fifth of the somatodendritic processes associated with enkephalin-containing axon terminals. These findings support the hypothesis that enkephalin may act in the VMN by inhibiting GABAergic neurons, which could result in the disinhibition of neural circuits relevant for lordosis.


Subject(s)
Enkephalins/physiology , Ventromedial Hypothalamic Nucleus/physiology , Ventromedial Hypothalamic Nucleus/ultrastructure , Animals , Estrogens/physiology , Female , Immunohistochemistry , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/metabolism
14.
J Comp Neurol ; 430(2): 200-8, 2001 Feb 05.
Article in English | MEDLINE | ID: mdl-11135256

ABSTRACT

The delta opioid receptor (DOR) modulates nociception and blood pressure in the periaqueductal gray (PAG). To examine the cellular basis for DOR effects, the ultrastructural distribution of DOR immunoreactivity was examined in the caudal ventrolateral PAG. DOR immunoreactivity was located predominantly in axon terminals that formed asymmetric (excitatory-type) synaptic contacts. However, rather then localized to the plasma membrane of synaptic boutons, immunolabeling for the DOR was intracellular, often associated with large dense-core vesicles. This finding suggests that dense-core vesicles may play a role in targeting the DOR, as vesicle fusion would shift the distribution of the DOR to the plasma membrane. To investigate the neural circuits in which DOR may function, dual-immunolabeling was used to determine the relationship of the DOR to an endogenous ligand, enkephalin, and to a potential target, GABAergic neurons. Approximately a third (38 of 127) of DOR containing axons had enkephalin immunoreactivity, indicating DOR may act in part as a presynaptic autoreceptor. Although single axon terminals containing immunoreactivity for both DOR and GABA were not detected, some DOR-immunolabeled axon terminals (26 of 86) contacted soma or dendrites containing GABA. These data suggest that the DOR may act in part as an autoreceptor to regulate synaptic input to GABAergic as well as non-GABAergic PAG neurons. Furthermore, the exposure of the DOR to the extracellular space may be contingent upon dense-core vesicle fusion with the plasma membrane.


Subject(s)
Periaqueductal Gray/physiology , Presynaptic Terminals/physiology , Rats/physiology , Receptors, Opioid, delta/physiology , Animals , Enkephalins/metabolism , Immunohistochemistry , Periaqueductal Gray/ultrastructure , Presynaptic Terminals/ultrastructure , gamma-Aminobutyric Acid/metabolism
15.
J Comp Neurol ; 419(4): 532-42, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10742719

ABSTRACT

The ventrolateral portion of the periaqueductal gray (PAG) is one brain region in which ligands of the mu-opioid receptor (MOR) produce analgesia. In the PAG, MOR ligands are thought to act primarily on inhibitory [e.g., gamma-aminobutyric acidergic (GABAergic)] neurons to disinhibit PAG output rather than directly on medullary-projecting PAG neurons. In this study, the ultrastructural localization of MOR immunolabeling was examined with respect to either GABAergic PAG neurons or PAG projection neurons that were labeled retrogradely from the rostral ventromedial medulla. Immunoreactivity for MOR and GABA often coexisted within dendrites. Dual-labeled profiles accounted for subpopulations of dendrites containing immunoreactivity for either MOR (65 of 145 dendrites; 45%) or GABA (65 of 183 dendrites; 35%). In addition, nearly half of PAG neuronal profiles (148 of 344 profiles) that were labeled retrogradely from the ventromedial medulla contained MOR immunoreactivity. MOR was distributed equally among retrogradely labeled neuronal profiles in the lateral and ventrolateral columns of the caudal PAG. With respect to the presynaptic distribution of MOR, approximately half of MOR-immunolabeled axon terminals (35 of 69 terminals) also contained GABA. Some MOR and GABA dual-immunolabeled axon terminals contacted unlabeled dendrites (11 of 35 terminals), whereas others contacted GABA-immunoreactive dendrites (15 of 35 terminals). Furthermore, axon terminals synapsing on medullary-projecting PAG neurons sometimes contained immunoreactivity for MOR. These data support the model that MOR ligands can act by inhibiting GABAergic neurons, but they also provide evidence that MOR ligands may act directly on PAG output neurons. In addition, MOR at presynaptic sites could affect both GABAergic neurons and output neurons. Thus, the disinhibitory model represents only partially the potential mechanisms by which MOR ligands can modulate output of the PAG.


Subject(s)
Medulla Oblongata/metabolism , Neural Pathways/metabolism , Periaqueductal Gray/metabolism , Presynaptic Terminals/metabolism , Receptors, Opioid, mu/analysis , Synapses/metabolism , gamma-Aminobutyric Acid/analysis , Animals , Male , Medulla Oblongata/ultrastructure , Neural Pathways/ultrastructure , Periaqueductal Gray/ultrastructure , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Synapses/ultrastructure
16.
J Comp Neurol ; 417(1): 103-14, 2000 Jan 31.
Article in English | MEDLINE | ID: mdl-10660891

ABSTRACT

The authors previously showed that a subset of axon terminals in the locus coeruleus (LC) contains methionine5-enkephalin (ENK) and gamma-aminobutyric acid (GABA) immunoreactivities. However, numerous ENK-labeled terminals lacked GABA and exhibited synaptic specializations that were characteristic of excitatory-type transmitters. To determine whether ENK coexists with glutamate in the LC, preembedding immunoperoxidase detection of ENK or immunogold-silver was combined with postembedding identification of glutamate using a gold marker. Indeed, 28% of the ENK-labeled axon terminals examined (n = 250 axon terminals) also contained glutamate. To define further sites for functional interactions between opiate ligands and excitatory amino acid receptors, the ultrastructural localization of the mu-opioid receptor (MOR) was examined with respect to either the kainate receptor (KAR) or the R1 subunit of the N-methyl-D-aspartate (NR1)-type glutamate receptor in the LC. Gold-silver labeling for MOR and peroxidase labeling for either KAR or NR1 indicated that the MOR often was localized to the plasma membrane of dendrites that also exhibited immunolabeling for either glutamate receptor subtype. In contrast to the KAR, which was identified primarily in somata and dendrites, NR1 immunoreactivity also was found frequently in axon terminals as well as in glial processes. Glial processes containing NR1 occasionally exhibited immunolabeling for MOR and sometimes were directly apposed to MOR-containing dendrites in the LC. Furthermore, NR1-labeled receptors in axon terminals sometimes were presynaptic to MOR-labeled dendrites. The authors concluded that ENK and glutamate may be cotransmitters in LC afferents. Moreover, ligands at the KAR may modulate directly MOR-containing neurons in the LC, whereas actions at NR1 receptors may affect opioid-sensitive neurons through multiple cellular mechanisms, i.e., through presynaptic, postsynaptic, or glial actions.


Subject(s)
Enkephalins/metabolism , Glutamic Acid/metabolism , Locus Coeruleus/metabolism , Neurons/metabolism , Presynaptic Terminals/metabolism , Rats/metabolism , Animals , Locus Coeruleus/ultrastructure , Male , Neurons/ultrastructure , Presynaptic Terminals/ultrastructure , Protein Isoforms/metabolism , Rats, Sprague-Dawley , Receptors, Glutamate/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Opioid/physiology , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Tissue Distribution
17.
J Comp Neurol ; 408(4): 549-59, 1999 Jun 14.
Article in English | MEDLINE | ID: mdl-10340504

ABSTRACT

In the ventrolateral periaqueductal gray (PAG), endogenous pathways which dampen pain transmission can be activated by either opioids or excitatory amino acids such as N-methyl D-aspartate (NMDA). The effects of these ligands may converge, because morphine-produced analgesia in the PAG can be blocked by NMDA receptor antagonists. To determine the relationship between the subcellular sites where opioid ligands of the mu opioid receptor (MOR) and NMDA receptor ligands may act, we studied the ultrastructural distribution of immunolabeling for MOR and the R1 subunit of the NMDA receptor (NR1) in the ventrolateral PAG. MOR labeling was most commonly distributed along extrasynaptic regions of the plasma membrane of neuronal dendrites (80% or 245/306). In addition, MOR labeling was found presynaptically in axon terminals (13% or 39/306) which preferentially formed symmetric (inhibitory-type) synapses. NR1 immunoreactivity was also prevalent in dendrites (72% or 242/335), but in contrast to MOR, was usually associated with a subset of postsynaptic densities. Axon terminals (5%, 17/335) and glial processes (18%, 61/335) comprised the remainder of NR1-labeled profiles. There was a striking colocalization of MOR and NR1 labeling within dendrites. The majority of NR1-labeled dendrites contained MOR labeling (72%, 176/242) and likewise, the majority of MOR-labeled dendrites contained NR1 labeling (72%, 176/245). Thus, mu opioid and NMDA receptor ligands may act at several overlapping subcellular sites to modulate behaviors subserved by the ventrolateral PAG, such as antinociception.


Subject(s)
Neurons/physiology , Periaqueductal Gray/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Receptors, Opioid, mu/physiology , Synapses/physiology , Animals , Axons/physiology , Axons/ultrastructure , Immunohistochemistry , Male , Microscopy, Electron , Periaqueductal Gray/anatomy & histology , Rats , Rats, Sprague-Dawley , Subcellular Fractions/physiology , Subcellular Fractions/ultrastructure
18.
Brain Res ; 816(1): 58-67, 1999 Jan 16.
Article in English | MEDLINE | ID: mdl-9878688

ABSTRACT

Immunohistochemical-labeling for the neurochemicals gamma-aminobutyric acid (GABA) and enkephalin are abundant in the ventromedial nucleus of the hypothalamus (VMN). In VMN, both GABA and enkephalin may function to regulate feeding behavior, as well as other hormone-controlled behaviors. Importantly, in several brain areas, enkephalin is often thought to modulate GABAergic neurotransmission. Therefore, we used dual-labeling immunohistochemistry with electron microscopic analysis to study the circuitry of neurons containing GABA- and/or enkephalin-labeling within the VMN. Somato-dendritic profiles containing GABA-labeling were three fold more abundant than GABA-labeled axon terminals (117 soma or dendrites vs. 34 axons). In addition, axon terminals containing GABA-labeling sometimes synapsed onto GABA-labeled somata or dendrites (25% or 9/34). In contrast, under these conditions labeling for enkephalin was primarily restricted to axon terminals, which were very abundant throughout VMN. Enkephalin-containing terminals accounted for a large fraction (25% 23/92) of the axons in contact with GABA-labeled dendrites, although they also contacted unlabeled dendrites. These observations suggest that a population of VMN neurons are GABAergic. These may be either local circuit 'interneurons' or projection neurons. In addition, GABA-labeled VMN neurons may be regulated by either enkephalin or GABA. These morphologic observations provide the basis for disinhibitory mechanisms to function within the VMN.


Subject(s)
Enkephalins/metabolism , Neurons/metabolism , Presynaptic Terminals/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Dendrites/metabolism , Dendrites/ultrastructure , Immunohistochemistry , Interneurons/metabolism , Male , Microscopy, Electron , Neurons/physiology , Neurons/ultrastructure , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Synapses/metabolism , Synapses/ultrastructure
19.
J Comp Neurol ; 388(4): 575-86, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9388017

ABSTRACT

The three classes of opioid receptors, mu, delta, and kappa, are distributed within the locus coeruleus (LC) of the rat brain. We have recently shown with immunoelectron microscopy that the mu-opioid receptor (muOR) is localized prominently to extrasynaptic sites on the plasma membranes of noradrenergic perikarya and dendrites of the LC. To further characterize the cellular distribution of other opioid receptors in this region, in this study, we examined the ultrastructural localization of an antipeptide sequence unique to the delta-opioid receptor (deltaOR) in sections that were also dual labeled for methionine-enkephalin (M-ENK), an opioid peptide known to be an endogenous ligand of the deltaOR. Immunoperoxidase labeling for deltaOR was localized primarily to the plasma membranes of presynaptic axon terminals and was also associated with large dense core vesicles. The deltaOR-labeled axon terminals formed both excitatory (asymmetric) and inhibitory (symmetric) type synaptic specializations with unlabeled dendrites and were frequently apposed by astrocytic processes. Dual labeling showed that, of 180 deltaOR-labeled axon terminals, 16% showed colocalization with M-ENK. These formed both types of synaptic junctions. Peroxidase labeling for deltaOR was also observed occasionally within dendrites, unmyelinated axons, and glial processes. The deltaOR-labeled dendrites were usually postsynaptic to unlabeled axon terminals that contained both small clear and large dense core vesicles. These results provide the first ultrastrucutral evidence that, in the LC, deltaOR may play a role in the presynaptic modulation of release of both excitatory and inhibitory neurotransmitters. They also suggest involvement of deltaOR in autoregulation of M-ENK release from axon terminals in this region.


Subject(s)
Brain Mapping , Enkephalin, Methionine/analysis , Locus Coeruleus/cytology , Presynaptic Terminals/chemistry , Receptors, Opioid, delta/analysis , Animals , Astrocytes/physiology , Immunohistochemistry , Locus Coeruleus/ultrastructure , Male , Microscopy, Electron , Myelin Sheath/physiology , Rats , Rats, Sprague-Dawley
20.
J Comp Neurol ; 381(3): 373-87, 1997 May 12.
Article in English | MEDLINE | ID: mdl-9133574

ABSTRACT

To study possible cellular targets and subcellular sites of action of opioid ligands in the rat hippocampus, we examined the distribution of the delta opioid receptor (DOR) by immunocytochemistry. By light microscopy, numerous interneurons, or non-principal cells, were intensely labeled for DOR, whereas the CA1 and CA3 pyramidal cells were lightly labeled. DOR-immunoreactive interneurons were found throughout the hippocampus but were particularly concentrated in stratum oriens of the CA1 region. Double labeling immunofluorescence revealed that DOR-immunoreactivity was found in a subpopulation of gamma-aminobutyric acid (GABA)-containing interneurons, which included most somatostatin-immunoreactive cells. Electron microscopic analysis of sections singly labeled for DOR revealed that DOR-immunoreactive profiles were abundant and widespread throughout all hippocampal lamina and had a similar distribution in CA1 and CA3. DOR-immunoreactivity was sometimes found in dendrites, which corresponded in morphology to those of interneurons. In addition, DOR-labeling was found in the shafts and spines of many dendrites, which exhibited the morphology of pyramidal cell dendrites. Within dendrites, dense DOR-immunoreactivity was associated with the plasmalemmal surface at or near the postsynaptic density, usually of asymmetric synapses. In addition, DOR labeling was present in a heterogeneous population of axon terminals, as well as in astrocytic profiles. At mossy fiber synapses, DOR labeling was occasionally found at both pre-and post-synaptic sites. These studies demonstrate that DOR is present at multiple sites on diverse cell types where it may function to regulate neuronal activity in the hippocampus.


Subject(s)
Interneurons/chemistry , Pyramidal Cells/chemistry , Rats, Sprague-Dawley/physiology , Receptors, Opioid, delta/analysis , Animals , Antibody Specificity , Dendrites/chemistry , Dendrites/ultrastructure , Enkephalins/analysis , Fluorescent Antibody Technique , Hippocampus/chemistry , Hippocampus/cytology , Interneurons/ultrastructure , Male , Microscopy, Electron , Pyramidal Cells/ultrastructure , Rats , Receptors, Opioid, delta/immunology , Somatostatin/analysis , Somatostatin/immunology , Synapses/chemistry , Synapses/ultrastructure , gamma-Aminobutyric Acid/analysis , gamma-Aminobutyric Acid/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...