Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Neurosci ; 29(25): 8198-205, 2009 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-19553459

RESUMO

Growth hormone (GH) secretion in male rats exhibits a 3.3 h ultradian rhythm generated by the reciprocal interaction of GH-releasing hormone (GHRH) and somatostatin (SRIF). SRIF receptor subtypes sst(1) and sst(2) are highly expressed in GHRH neurons of the hypothalamic arcuate nucleus (ARC). We previously demonstrated an ultradian oscillation in binding of SRIF analogs to the ARC in relation to GH peaks and troughs. Here we tested the hypothesis that these ultradian changes in SRIF binding are due to differential plasma membrane targeting of sst(1) receptors in ARC neurons using immunocytochemistry and electron microscopy. We found that 87% of sst(1)-positive ARC neurons also synthesized GHRH. Subcellularly, 80% of sst(1) receptors were located intracellularly and 20% at the plasma membrane regardless of GH status. However, whereas 30% of the cell-surface sst(1) receptors were located perisynaptically or subsynaptically following exposure to high GH secretion, this fraction was increased to 42% following a GH trough period (p = 0.05). Furthermore, the relative abundance of symmetric and asymmetric synapses on sst(1)-positive dendrites also varied significantly, depending on the GH cycle, from approximately equal numbers following GH troughs to 70:30 in favor of symmetric, i.e., inhibitory, inputs after GH peaks (p < 0.02). These findings suggest that postsynaptic localization of sst(1) receptors and synaptic connectivity in the ARC undergo pronounced remodeling in parallel with the GH rhythm. Such synaptic plasticity may be an important mechanism by which sst(1) mediates SRIF's cyclical effects on ARC GHRH neurons to generate the ultradian rhythm of GH secretion.


Assuntos
Ciclos de Atividade/fisiologia , Núcleo Arqueado do Hipotálamo/metabolismo , Hormônio Liberador de Hormônio do Crescimento/sangue , Hormônio do Crescimento/sangue , Neurônios/metabolismo , Receptores de Somatostatina/metabolismo , Sinapses/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/ultraestrutura , Imunofluorescência , Imuno-Histoquímica , Masculino , Microscopia Eletrônica , Neurônios/ultraestrutura , Radioimunoensaio , Ratos , Ratos Sprague-Dawley , Somatostatina/sangue , Sinapses/ultraestrutura
2.
Mol Biol Cell ; 16(1): 306-15, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15496457

RESUMO

The neuronal isoform of vesicular monoamine transporter, VMAT2, is responsible for packaging dopamine and other monoamines into synaptic vesicles and thereby plays an essential role in dopamine neurotransmission. Dopamine neurons in mice lacking VMAT2 are unable to store or release dopamine from their synaptic vesicles. To determine how VMAT2-mediated filling influences synaptic vesicle morphology and function, we examined dopamine terminals from VMAT2 knockout mice. In contrast to the abnormalities reported in glutamatergic terminals of mice lacking VGLUT1, the corresponding vesicular transporter for glutamate, we found that the ultrastructure of dopamine terminals and synaptic vesicles in VMAT2 knockout mice were indistinguishable from wild type. Using the activity-dependent dyes FM1-43 and FM2-10, we also found that synaptic vesicles in dopamine neurons lacking VMAT2 undergo endocytosis and exocytosis with kinetics identical to those seen in wild-type neurons. Together, these results demonstrate that dopamine synaptic vesicle biogenesis and cycling are independent of vesicle filling with transmitter. By demonstrating that such empty synaptic vesicles can cycle at the nerve terminal, our study suggests that physiological changes in VMAT2 levels or trafficking at the synapse may regulate dopamine release by altering the ratio of fillable-to-empty synaptic vesicles, as both continue to cycle in response to neural activity.


Assuntos
Dopamina/metabolismo , Glicoproteínas de Membrana/genética , Proteínas de Membrana Transportadoras/genética , Neurônios/metabolismo , Vesículas Sinápticas/fisiologia , Animais , Encéfalo/metabolismo , Corantes/farmacologia , Endocitose , Ácido Glutâmico/metabolismo , Cinética , Mesencéfalo/metabolismo , Mesencéfalo/ultraestrutura , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Neurônios/ultraestrutura , Isoformas de Proteínas , Sinapses/metabolismo , Fatores de Tempo , Proteínas Vesiculares de Transporte de Aminas Biogênicas , Proteínas Vesiculares de Transporte de Monoamina
3.
Glob Heart ; 13(2): 65-72, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29716847

RESUMO

Four decades ago, U.S. life expectancy was within the same range as other high-income peer countries. However, during the past decades, the United States has fared worse in many key health domains resulting in shorter life expectancy and poorer health-a health disadvantage. The National Heart, Lung, and Blood Institute convened a panel of national and international health experts and stakeholders for a Think Tank meeting to explore the U.S. health disadvantage and to seek specific recommendations for implementation research opportunities for heart, lung, blood, and sleep disorders. Recommendations for National Heart, Lung, and Blood Institute consideration were made in several areas including understanding the drivers of the disadvantage, identifying potential solutions, creating strategic partnerships with common goals, and finally enhancing and fostering a research workforce for implementation research. Key recommendations included exploring why the United States is doing better for health indicators in a few areas compared with peer countries; targeting populations across the entire socioeconomic spectrum with interventions at all levels in order to prevent missing a substantial proportion of the disadvantage; assuring partnership have high-level goals that can create systemic change through collective impact; and finally, increasing opportunities for implementation research training to meet the current needs. Connecting with the research community at large and building on ongoing research efforts will be an important strategy. Broad partnerships and collaboration across the social, political, economic, and private sectors and all civil society will be critical-not only for implementation research but also for implementing the findings to have the desired population impact. Developing the relevant knowledge to tackle the U.S. health disadvantage is the necessary first step to improve U.S. health outcomes.


Assuntos
Pesquisa Biomédica , Doenças Cardiovasculares/prevenção & controle , Longevidade/fisiologia , National Heart, Lung, and Blood Institute (U.S.) , Guias de Prática Clínica como Assunto , Congressos como Assunto , Humanos , Estados Unidos
4.
J Neurosci ; 26(3): 953-62, 2006 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-16421315

RESUMO

The present study demonstrates that perikaryaldelta-opioid receptors (deltaORs) in rat dorsal root ganglion (DRG) neurons bind and internalize opioid ligands circulating in the CSF. Using confocal and electron microscopy, we found that prolonged morphine treatment increased the cell surface density of these perikaryal deltaORs and, by way of consequence, receptor-mediated internalization of the fluorescent deltorphin (DLT) analog omega-Bodipy 576/589 deltorphin-I 5-aminopentylamide (Fluo-DLT) in all three types of DRG neurons (small, medium, and large). In contrast, chronic inflammatory pain induced by the injection of complete Freund's adjuvant (CFA) into one hindpaw selectively increased Fluo-DLT internalization in small and medium-sized DRG neurons ipsilateral to the inflammation. Based on our previous studies in the spinal cord of mu-opioid receptor (muOR) knock-out mice, it may be assumed that the enhanced membrane recruitment of deltaORs observed after sustained morphine is attributable to stimulation of muORs. However, the selectivity of the effect induced by inflammatory pain suggests that it involves a different mechanism, namely a modality-specific and pain-related activation of C and Adelta fibers. Indeed, stimulation by capsaicin of transient receptor potential vanilloid 1 receptors, which are selectively expressed by small diameter (< 600 microm2) DRG neurons, increased Fluo-DLT internalization exclusively in this cell population. The present results, therefore, demonstrate that DRG neurons express perikaryal deltaORs accessible to CSF-circulating ligands and that the density and, hence, presumably also the responsiveness, of these receptors may be modulated by both pain-related stimuli and sustained exposure to muOR agonists.


Assuntos
Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Morfina/farmacologia , Medição da Dor/efeitos dos fármacos , Receptores Opioides delta/análise , Receptores Opioides delta/biossíntese , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Gânglios Espinais/citologia , Masculino , Morfina/uso terapêutico , Dor/tratamento farmacológico , Dor/metabolismo , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/fisiologia
5.
Endocrinology ; 148(5): 2095-105, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17272399

RESUMO

The somatostatin [somatotropin release-inhibiting factor (SRIF)] receptor subtypes sst(2A) and sst(5) are frequently coexpressed in SRIF-responsive cells, including endocrine pituitary cells. We previously demonstrated that sst(2A) and sst(5) exhibit different subcellular localizations and regulation of cell surface expression, although they have similar signaling properties. We investigated here whether sst(2A) and sst(5) functionally interact in cells coexpressing the two receptor subtypes. We stimulated both transfected cells stably expressing sst(2A) alone (CHO-sst(2A)) or together with sst(5) (CHO-sst(2A+5)) and the pituitary cell line AtT20, which endogenously expresses the two receptor subtypes, with either the nonselective agonist [D-Trp(8)]-SRIF-14 or the sst(2)-selective agonist L-779,976. In CHO-sst(2A) cells, stimulation with either ligand resulted in the loss of approximately 75% of cell surface SRIF binding sites and massive internalization of sst(2A) receptors. The cells were desensitized to subsequent stimulation with [D-Trp(8)]-SRIF-14, which failed to inhibit forskolin-evoked cAMP accumulation. Similarly, in CHO-sst(2A+5) and AtT20 cells, [D-Trp(8)]-SRIF-14 induced the loss of 60-70% of SRIF binding sites as well as massive sst(2A) endocytosis. By contrast, in cells expressing both sst(2A) and sst(5), selective stimulation of sst(2A) with L-779,976 resulted in only 20-40% loss of cell surface binding and markedly reduced sst(2A) internalization. Consequently, whereas CHO-sst(2A+5) and AtT20 cells stimulated with [D-Trp(8)]-SRIF-14 were desensitized to a second stimulation with the same agonist, cells prestimulated with L-779,976 were not desensitized to subsequent [D-Trp(8)]-SRIF-14 stimulation. These findings indicate that the presence of sst(5) in the same cells modulates trafficking and cell surface regulation of sst(2A) and cellular desensitization to the effects of SRIF.


Assuntos
Endocitose/fisiologia , Transporte Proteico/fisiologia , Receptores de Somatostatina/metabolismo , Amidas/metabolismo , Amidas/farmacologia , Animais , Células CHO , Colforsina/farmacologia , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Indóis/metabolismo , Indóis/farmacologia , Radioisótopos do Iodo , Camundongos , Ensaio Radioligante , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/genética , Transdução de Sinais/fisiologia , Somatostatina/análogos & derivados , Somatostatina/metabolismo , Somatostatina/farmacologia , Transfecção
6.
J Neurosci ; 25(36): 8188-96, 2005 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-16148226

RESUMO

Intrathecal administration of the neuropeptide neurotensin (NT) was shown previously to exert antinociceptive effects in a variety of acute spinal pain paradigms including hotplate, tail-flick, and writhing tests. In the present study, we sought to determine whether some of these antinociceptive effects might be elicited via stimulation of low-affinity NTS2 receptors. We first established, using immunoblotting and immunohistochemical techniques, that NTS2 receptors were extensively associated with putative spinal nociceptive pathways, both at the level of the dorsal root ganglia and of the superficial layers of the dorsal horn of the spinal cord. We then examined the effects of intrathecal administration of NT or selective NTS2 agonists on acute thermal pain. Both NT and NTS2 agonists, levocabastine and Boc-Arg-Arg-Pro-Tyrpsi(CH2NH)Ile-Leu-OH (JMV-431), induced dose-dependent antinociceptive responses in the tail-flick test. The effects of levocabastine and of JMV-431 were unaffected by coadministration of the NTS1-specific antagonist 2-[(1-(7-chloro-4-quinolinyl)-5-(2,6-dimethoxy-phenyl)pyrazol-3-yl)carboxylamino]tricyclo)3.3.1.1.(3.7))-decan-2-carboxylic acid (SR48692), confirming that they were NTS2 mediated. In contrast, the antinociceptive effects of NT were partly abolished by coadministration of SR48692, indicating that NTS1 and NTS2 receptors were both involved. These results suggest that NTS2 receptors play a role in the regulation of spinal nociceptive inputs and that selective NTS2 agonists may offer new avenues for the treatment of acute pain.


Assuntos
Analgésicos/farmacologia , Gânglios Espinais/fisiologia , Receptores de Neurotensina/fisiologia , Medula Espinal/fisiologia , Analgésicos/administração & dosagem , Animais , Gânglios Espinais/efeitos dos fármacos , Temperatura Alta , Técnicas In Vitro , Injeções Espinhais , Vértebras Lombares , Modelos Animais , Neurotensina/farmacologia , Dor , Piperidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Neurotensina/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Cauda
7.
J Neurosci ; 23(12): 4888-98, 2003 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12832511

RESUMO

We recently demonstrated that prolonged treatment with morphine increases the antinociceptive potency of the delta-opioid receptor (deltaOR) agonist deltorphin and promotes cell surface targeting of deltaORs in neurons of the dorsal horn of the rat spinal cord (Cahill et al., 2001b). In the present study we examined whether these effects were mediated selectively via muOR. Using the same intermittent treatment regimen as for morphine, we found that methadone and etorphine, but not fentanyl, enhanced [D-Ala2]-deltorphin-mediated antinociception. However, continuous delivery of fentanyl for 48 hr resulted in augmented deltaOR-mediated antinociception when compared with saline-infused animals. Time course studies confirmed that a 48 hr treatment with morphine was necessary for the establishment of enhanced deltaOR-mediated antinociception. The observed increases in deltaOR agonist potency and deltaOR plasma membrane density were reversed fully 48 hr after discontinuation of morphine injections. Wild-type C57BL/6 mice pretreated with morphine for 48 hr similarly displayed enhanced deltaOR-mediated antinociception in a tonic pain paradigm. Accordingly, the percentage of plasma membrane-associated deltaOR in the dorsal horn of the spinal cord, as assessed by immunogold electron microscopy, increased from 6.6% in naive to 12.4% in morphine-treated mice. In contrast, morphine treatment of muOR gene knock-out (KO) mice did not produce any change in deltaOR plasma membrane density. These results demonstrate that selective activation of muOR is critical for morphine-induced targeting of deltaOR to neuronal membranes, but not for basal targeting of this receptor to the cell surface.


Assuntos
Receptores Opioides delta/metabolismo , Receptores Opioides mu/deficiência , Animais , Área Sob a Curva , Comportamento Animal/efeitos dos fármacos , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , Etorfina/farmacologia , Feminino , Fentanila/administração & dosagem , Fentanila/farmacologia , Masculino , Metadona/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfina/farmacologia , Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos , Células do Corno Posterior/citologia , Células do Corno Posterior/efeitos dos fármacos , Células do Corno Posterior/metabolismo , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/agonistas , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Tempo , Regulação para Cima/efeitos dos fármacos
8.
J Neurosci ; 24(24): 5549-59, 2004 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-15201327

RESUMO

An in vivo fluorescent deltorphin (Fluo-DLT) internalization assay was used to assess the distribution and regulation of pharmacologically available delta opioid receptors (deltaORs) in the rat lumbar (L4-5) spinal cord. Under basal conditions, intrathecal injection of Fluo-DLT resulted in the labeling of numerous deltaOR-internalizing neurons throughout dorsal and ventral horns. The distribution and number of Fluo-DLT-labeled perikaryal profiles were consistent with that of deltaOR-expressing neurons, as revealed by in situ hybridization and immunohistochemistry, suggesting that a large proportion of these cells was responsive to intrathecally administered deltaOR agonists. Pretreatment of rats with morphine for 48 hr resulted in a selective increase in Fluo-DLT-labeled perikaryal profiles within the dorsal horn. These changes were not accompanied by corresponding augmentations in either deltaOR mRNA or (125)I-deltorphin-II binding levels, suggesting that they were attributable to higher densities of cell surface deltaOR available for internalization rather than to enhanced production of the receptor. Unilateral dorsal rhizotomy also resulted in increased Fluo-DLT internalization in the ipsilateral dorsal horn when compared with the side contralateral to the deafferentation or to non-deafferented controls, suggesting that deltaOR trafficking in dorsal horn neurons may be regulated by afferent inputs. Furthermore, morphine treatment no longer increased Fluo-DLT internalization on either side of the spinal cord after unilateral dorsal rhizotomy, indicating that microOR-induced changes in the cell surface availability of deltaOR depend on the integrity of primary afferent inputs. Together, these results suggest that regulation of deltaOR responsiveness through microOR activation in this region is linked to somatosensory information processing.


Assuntos
Morfina/farmacologia , Entorpecentes/farmacologia , Receptores Opioides delta/metabolismo , Medula Espinal/metabolismo , Animais , Corantes Fluorescentes/química , Corantes Fluorescentes/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Hibridização In Situ , Região Lombossacral , Masculino , Microscopia de Fluorescência , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Células do Corno Posterior/metabolismo , Transporte Proteico , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/agonistas , Receptores Opioides delta/biossíntese , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Rizotomia , Medula Espinal/anatomia & histologia , Regulação para Cima
9.
J Mol Neurosci ; 25(3): 207-14, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15800374

RESUMO

In recent years, we demonstrated that prolonged (48-h) treatment of rats or mice with selective m-opioid receptor ((mu)OR) agonists induced a translocation of delta-opioid receptors ((delta)ORs) from intracellular compartments to neuronal plasma membranes in the dorsal horn of the spinal cord. It remained to be determined whether this phenomenon also occurred in the brain. To resolve this issue, we analyzed by immunogold histochemistry the subcellular distribution of (delta)ORs in the nucleus accumbens, dorsal neostriatum, and frontal cortex in mice treated or not with morphine (48 h). We observed that prolonged treatment with morphine induced a translocation of (delta)ORs from intracellular to subplasmalemmal and membrane compartments in dendrites from both the nucleus accumbens and the dorsal neostriatum but not from the frontal cortex. We propose that this (mu)OR-(delta)OR interaction might prolong and modulate the sensitivity of neurons to opiates in specific target regions.


Assuntos
Analgésicos Opioides/farmacologia , Morfina/farmacologia , Neostriado/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Receptores Opioides delta/metabolismo , Animais , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Feminino , Lobo Frontal/citologia , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Neostriado/citologia , Neostriado/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/ultraestrutura , Núcleo Accumbens/citologia , Núcleo Accumbens/metabolismo , Transporte Proteico/efeitos dos fármacos
10.
Med Sci (Paris) ; 21(8-9): 741-6, 2005.
Artigo em Francês | MEDLINE | ID: mdl-16115460

RESUMO

Apelin is a peptide that was recently isolated as the endogenous ligand for the human orphan APJ receptor, a G protein-coupled receptor which shares 31 % amino-acid sequence identity with the angiotensin type 1 receptor. Apelin naturally occurs in the brain and plasma as 13 (pE13F) and 17 amino-acid (K17F) fragments of a single pro-peptide precursor. In transfected CHO cells, K17F and pE13F bind with high affinity to the rat APJ receptor, promote receptor internalization, and inhibit forskolin-induced cAMP formation. In the same cells, pE13F activates MAP kinase and PI3 kinase pathways. Apelin and APJ receptors are both widely distributed in the brain but are particularly highly expressed in the supraoptic (SON) and paraventricular (PVN) hypothalamic nuclei. Dual labeling studies demonstrate that within these two nuclei, apelin and its receptor are colocalized with vasopressin (AVP) in a subset of magnocellular neurons. In lactating rats, characterized by increases in both synthesis and release of AVP, central injection of apelin inhibits the phasic electrical activity of AVP neurons, reduces plasma AVP levels, and increases aqueous diuresis. Moreover, water deprivation, while increasing the activity of AVP neurons, reduces plasma apelin concentrations and induces an intra-neuronal pile up of the peptide, thereby decreasing the inhibitory effect of apelin on AVP release and preventing additional water loss at the kidney level. Taken together, these data demonstrate that apelin counteracts the effects of AVP in the maintenance of body fluid homeostasis. In addition, apelin and its receptor are present in the cardiovascular system, i.e. heart, kidney and vessels. Systemically administered apelin reduces arterial blood pressure, increases cardiac contractility and reduces cardiac loading. The development of non peptidic analogs of apelin may therefore offer new therapeutic avenues for the treatment of cardiovascular disorders.


Assuntos
Proteínas de Transporte/fisiologia , Neuropeptídeos/fisiologia , Vasopressinas/metabolismo , Sequência de Aminoácidos , Animais , Apelina , Líquidos Corporais/fisiologia , Encéfalo/fisiologia , Sequência Conservada , Homeostase , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Ligantes , Dados de Sequência Molecular , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Vasopressinas/antagonistas & inibidores
11.
Int J Biochem Cell Biol ; 36(11): 2153-68, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15313463

RESUMO

The neurotensin receptor-3, originally identified as sortilin, is unique among neuropeptide receptors in that it is a single trans-membrane domain, type I receptor. To gain insight into the functionality of neurotensin receptor-3, we examined the neurotensin-induced intracellular trafficking of this receptor in the human carcinoma cell line HT29, which expresses both neurotensin receptor-1 and -3 sub-types. At steady state, neurotensin receptor-3 was found by sub-cellular fractionation and electron microscopic techniques to be predominantly associated with intracellular elements. A small proportion (approximately 10%) was associated with the plasma membrane, but a significant amount (approximately 25%) was observed inside the nucleus. Following stimulation with neurotensin (NT), neurotensin/neurotensin receptor-3 complexes were internalized via the endosomal pathway. This internalization entailed no detectable loss of cell surface receptors, suggesting compensation through either recycling or intracellular receptor recruitment mechanisms. Internalized ligand and receptors were both sorted to the pericentriolar recycling endosome/Trans-Golgi Network (TGN), indicating that internalized neurotensin is sorted to this compartment via neurotensin receptor-3. Furthermore, within the Trans-Golgi Network, neurotensin was bound to a lower molecular form of the receptor than at the cell surface or in early endosomes, suggesting that signaling and transport functions of neurotensin receptor-3 may be mediated through different molecular forms of the protein. In conclusion, the present work suggests that the neurotensin receptor-3 exists in two distinct forms in HT29 cells: a high molecular weight, membrane-associated form responsible for neurotensin endocytosis from the cell surface and a lower molecular weight, intracellular form responsible for the sorting of internalized neurotensin to the Trans-Golgi Network.


Assuntos
Membrana Celular/metabolismo , Endocitose/fisiologia , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transporte Proteico/fisiologia , Receptores de Neurotensina/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Fracionamento Celular , Núcleo Celular/metabolismo , Complexo de Golgi/metabolismo , Células HT29 , Humanos , Microscopia Imunoeletrônica , Neurotensina/metabolismo , Transdução de Sinais/fisiologia
12.
Endocrinology ; 145(9): 4392-400, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15166125

RESUMO

Apelin, a neuropeptide recently identified as the endogenous ligand for the G protein-coupled receptor APJ, is highly concentrated in brain structures involved in the control of body fluid homeostasis including the supraoptic (SON) and paraventricular (PVN) hypothalamic nuclei. To clarify the implication of apelin in the regulation of water balance, we sought to determine whether apelin colocalized with arginine vasopressin (AVP) in the rat SON and PVN. We also investigated the effects of water deprivation on the levels of apelin within these two nuclei by comparison with those of AVP. Using dual immunolabeling confocal microscopy, we found that a large proportion of apelin-immunoreactive neurons colocalized AVP within both the SON and PVN, but that the two peptides were segregated within distinct subcellular compartments inside these cells. Both the number and labeling intensity of magnocellular apelin-immunoreactive cells increased significantly after 24- or 48-h dehydration, whereas the number and labeling density of AVP-immunoreactive neurons significantly decreased. The dehydration-induced increase in apelin immunoreactivity was markedly diminished by central injection of a selective vasopressin-1 receptor antagonist. Conversely, the effect of dehydration was mimicked by a 16-min intracerebroventricular infusion of AVP, again in a vasopressin-1 receptor antagonist-reversible manner. These results provide additional evidence for the involvement of the neuropeptide apelin in the control of body fluid homeostasis. They further suggest that the dehydration-induced release of AVP from magnocellular hypothalamic neurons may be responsible for the observed increase in immunoreactive apelin levels within the same neurons and thus that the release of one peptide may block that of another peptide synthesized in the same cells.


Assuntos
Arginina Vasopressina/metabolismo , Proteínas de Transporte/metabolismo , Desidratação/metabolismo , Hipotálamo Anterior/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Animais , Anticorpos , Apelina , Arginina Vasopressina/imunologia , Água Corporal/metabolismo , Proteínas de Transporte/imunologia , Homeostase/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Coelhos , Ratos , Receptores de Vasopressinas/metabolismo , Privação de Água/fisiologia
13.
Eur J Neurosci ; 4(12): 1309-1319, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-12106394

RESUMO

The topographic and cellular distribution of the neurotensin-hydrolysing neutral metalloendopeptidase 24.16 (EC 3.4.24.16) was examined by light and electron microscopic immunohistochemistry in adult rat mesencephalon. Light microscopic immunoradioautography revealed a ubiquitous distribution of the enzyme throughout the midbrain with a relative enrichment of grey matter areas including the substantia nigra, ventral tegmental area, interfascicular nucleus, interpeduncular nucleus, rostral and caudal linear raphe nuclei, central grey and superficial grey of the superior colliculus. Peroxidase - antiperoxidase immunocytochemistry revealed two distinct cellular patterns of immunostaining: (1) weakly labelled neuronal perikarya more or less uniformly distributed throughout the grey matter, and (2) intensely immunoreactive glial cells heterogeneously distributed across the mesencephalon. Areas exhibiting dense concentrations of endopeptidase 24.16-containing glial cells corresponded to those displaying enhanced immunoreactivity in immunoradioautographs, suggesting that a major proportion of brain endopeptidase 24.16 is associated with glia. Electron microscopic examination of the substantia nigra and ventral tegmental area confirmed the association of the enzyme with a subpopulation of neurons and allowed identification of labelled glial cells as protoplasmic astrocytes. In neurons, endopeptidase 24.16 immunoreactivity was distributed heterogeneously within the cytoplasm of perikarya, dendrites and axons. Reaction product was also characteristically associated with restricted zones of the plasma membrane and underlying neuroplasm. In astrocytes, endopeptidase 24.16 immunostaining was densely and uniformly distributed throughout the cytoplasm of cell bodies and processes. Many of these processes were in direct contact with endopeptidase 24.16-immunopositive neuronal elements. The present results demonstrate that within the midbrain, endopeptidase 24.16 is both intracytoplasmic and membrane-associated in neurons and predominantly intracytoplasmic in glia. The presence of a large number of immunostained elements within areas of the midbrain known to display high levels of neurotensin and/or neurotensin receptors, together with the demonstrated catabolic activity of the enzyme on neurotensin in vitro, is consistent with a role of endopeptidase 24.16 in the functional inactivation of endogenous neurotensin in this region of the brain.

14.
J Comp Neurol ; 461(4): 520-38, 2003 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-12746866

RESUMO

In the present study, we localized the levocabastine-sensitive neurotensin receptor (NTS2) protein in adult rat brain by using an N-terminally-directed antibody. NTS2-like immunoreactivity was broadly distributed throughout the rat brain. At the cellular level, the reaction product was exclusively associated with neurons and predominantly, although not exclusively, with their dendritic arbors. No NTS2 signal was observed over astrocytes, as confirmed by dual confocal microscopic immunofluorescence studies using the astrocytic marker S100beta. High densities of NTS2-like immunoreactive nerve cell bodies and/or processes were detected in many regions documented to receive a dense neurotensinergic innervation, such as the olfactory bulb, bed nucleus of the stria terminalis, magnocellular preoptic nucleus, amygdaloid complex, anterodorsal thalamic nucleus, substantia nigra, ventral tegmental area, and several brainstem nuclei. Most conspicuous among the latter were structures implicated in the descending control of nociceptive inputs (e.g., the periaqueductal gray, dorsal raphe, gigantocellular reticular nucleus, pars alpha, lateral paragigantocellular, and raphe magnus), in keeping with the postulated role of NTS2 receptors in the mediation of neurotensin's supraspinal antinociceptive actions. However, the distribution of NTS2-like immunoreactivity largely exceeded that of neurotensin terminal fields, and some of the highest concentrations of the receptor were found in areas devoid of neurotensinergic inputs such as the cerebral cortex, the hippocampus, and the cerebellum, suggesting that neurotensin may not be the exclusive endogenous ligand for this receptor subtype.


Assuntos
Mapeamento Encefálico , Encéfalo/metabolismo , Receptores de Neurotensina/metabolismo , Animais , Células COS/metabolismo , Células Cultivadas , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
15.
J Comp Neurol ; 465(3): 349-60, 2003 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-12966560

RESUMO

To assess the validity of rodent models for investigating the role of delta opioid receptors (DOR) in analgesia, the distribution of DOR binding and mRNA were compared between rodent and primate spinal cord and dorsal root ganglia (DRG), using receptor autoradiography and in situ hybridization, respectively. In mouse and rat spinal cord, [(125)I]-deltorphin-labeled DOR binding sites were detected throughout the gray matter. In contrast, in primate and particularly in human spinal cord, DOR binding was mainly present in laminae I-II, with little to no binding in deeper layers. Accordingly, in rodent spinal cord, DOR mRNA was expressed by a large number of neurons distributed throughout the ventral and dorsal horns, whereas in the primate, DOR expression was significantly lower, as evidenced by a moderate number of labeled cells throughout the gray matter in monkey and by only few labeled cells in human, mainly in Clarke's column and lamina IX. Major species differences in DOR expression were also observed in primary afferent cells bodies. In rat DRG, intense DOR mRNA hybridization was primarily observed over large ganglion cells immunopositive for neurofilament 200. In contrast, in monkey and human DRG, DOR mRNA was primarily detected over small and medium-sized ganglion cells. These results demonstrate major differences in the expression and distribution of DOR in the spinal cord and DRG between mammalian species. Specifically, they point to a progressive specialization of DOR toward the regulation of primary somatosensory, namely nociceptive, inputs during phylogeny and suggest that the effects of DOR agonists in rodents may not be entirely predictive of their action in humans.


Assuntos
Gânglios Espinais/metabolismo , Regulação da Expressão Gênica/fisiologia , Filogenia , Receptores Opioides delta/biossíntese , Medula Espinal/metabolismo , Animais , Sítios de Ligação/fisiologia , Feminino , Humanos , Macaca fascicularis , Masculino , Camundongos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/genética , Especificidade da Espécie
16.
J Comp Neurol ; 461(4): 483-505, 2003 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-12746864

RESUMO

The neurotensin (NT) receptor, NTS3, originally identified as the intracellular sorting protein sortilin, is a member of a recently discovered family of receptors characterized by a single transmembrane domain. The present study provides the first comprehensive description of the distribution of NTS3/sortilin mRNA and protein in adult rat brain using in situ hybridization and immunocytochemistry. Both NTS3/sortilin mRNA and immunoreactivity displayed a widespread distribution throughout the brain. High levels of NTS3/sortilin expression and immunoreactivity were found in neuronal cell bodies and dendrites of allocortical areas such as the piriform cortex and hippocampus. Regions expressing both high levels of NTS3/sortilin mRNA and protein also included several neocortical areas, the islands of Calleja, medial and lateral septal nuclei, amygdaloid nuclei, thalamic nuclei, the supraoptic nucleus, the substantia nigra, and the Purkinje cell layer of the cerebellar cortex. In the brainstem, all cranial nerve motor nuclei were strongly labeled. NTS3/sortilin mRNA and immunoreactivity were also detected over oligodendrocytes in major fiber tracts. Subcellularly, NTS3/sortilin was predominantly concentrated over intracytoplasmic membrane-bound organelles. Many of the areas exhibiting high levels of NTS3/sortilin (e.g., olfactory cortex, medial septum, and periaqueductal gray) have been documented to contain high concentrations of NT nerve cell bodies and axons, supporting the concept that NTS3/sortilin may play a role in NT sorting and/or signaling. Other areas (e.g., hippocampal CA fields, cerebellar cortex, and cranial nerve motor nuclei), however, are NT-negative, suggesting that NTS3/sortilin also exerts functions unrelated to NT signaling.


Assuntos
Mapeamento Encefálico , Encéfalo/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de Neurotensina/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Animais , Imuno-Histoquímica , Hibridização In Situ , Masculino , Glicoproteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Neuroglia/metabolismo , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Neurotensina/genética , Distribuição Tecidual
17.
Pain ; 109(3): 266-273, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15157687

RESUMO

Previous studies from our laboratory have demonstrated that both chronic inflammatory pain, induced by intraplantar injection of complete Freund's adjuvant (CFA), and prolonged (48 h) stimulation of mu-opioid receptors (muOR) by systemic administration of a variety of selective agonists, resulted in enhanced plasma membrane targeting of delta-opioid receptors (deltaOR) in neurons of the dorsal spinal cord. To determine whether deltaOR trafficking induced by chronic inflammation was dependent on the activation of muOR, we investigated by immunogold cytochemistry the effects of intraplantar CFA injection on the plasma membrane density of deltaOR in muOR knockout (KO) mice. In untreated wild-type (WT) mice, only a small proportion of deltaOR was associated with neuronal plasma membranes in the dorsal horn of the spinal cord. The CFA-induced inflammation produced a significantly higher ratio of plasma membrane to intracellular receptors, as well as a 75% increase in the membrane density of immunoreactive deltaOR, in dendrites of the ipsilateral dorsal horn as compared to untreated mice. This increase in the membrane density of deltaOR was likely due to a recruitment of receptors from intracellular stores since no difference in the overall deltaOR immunolabeling density was evident between CFA-treated and untreated mice. Most importantly, the CFA-induced changes in deltaOR plasma membrane insertion seen in WT animals were not present in the spinal cord of muOR KO mice. These results demonstrate that the integrity of muOR is necessary for CFA-induced changes in deltaOR trafficking to occur and suggest that these changes could be elicited by stimulation of muOR by endogenous opioids released in response to chronic inflammatory pain.


Assuntos
Inflamação/genética , Dor/genética , Receptores Opioides delta/metabolismo , Receptores Opioides mu/genética , Animais , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Doença Crônica , Dendritos/metabolismo , Dendritos/ultraestrutura , Modelos Animais de Doenças , Feminino , Adjuvante de Freund , Lateralidade Funcional/genética , Imuno-Histoquímica , Inflamação/metabolismo , Inflamação/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica , Dor/metabolismo , Dor/fisiopatologia , Células do Corno Posterior/metabolismo , Células do Corno Posterior/ultraestrutura , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Receptores Opioides mu/deficiência , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
18.
Neuropharmacology ; 42(8): 1089-98, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12128010

RESUMO

Following its binding to somatodendritic receptors, the neuropeptide neurotensin (NT) internalizes via a clathrin-mediated process. In the present study, we investigated whether NT also internalizes presynaptically using synaptosomes from rat neostriatum, a region in which NT1 receptors are virtually all presynaptic. Binding of [(3)H]-NT to striatal synaptosomes in the presence of levocabastine to block NT2 receptors is specific, saturable, and has NT1 binding properties. A significant fraction of the bound radioactivity is resistant to hypertonic acid wash indicating that it is internalized. Internalization of [(3)H]-NT, like that of [(125)I]-transferrin, is blocked by sucrose and low temperature, consistent with endocytosis occurring via a clathrin-dependent pathway. However, contrary to what was reported at the somatodendritic level, neither [(3)H]-NT nor [(125)I]-transferrin internalization in synaptosomes is sensitive to the endocytosis inhibitor phenylarsine oxide. Moreover, treatment of synaptosomes with monensin, which prevents internalized receptors from recycling to the plasma membrane, reduces [(3)H]-NT binding and internalization, suggesting that presynaptic NT1 receptors, in contrast to somatodendritic ones, are recycled back to the plasma membrane. Taken together, these results suggest that NT internalizes in nerve terminals via an endocytic pathway that is related to, but is mechanistically distinct from that responsible for NT internalization in nerve cell bodies.


Assuntos
Endocitose/fisiologia , Neostriado/metabolismo , Neurotensina/metabolismo , Receptores de Neurotensina/fisiologia , Transdução de Sinais/fisiologia , Sinaptossomos/metabolismo , Animais , Masculino , Neostriado/ultraestrutura , Terminações Pré-Sinápticas/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Neurotensina/ultraestrutura , Sinaptossomos/ultraestrutura , Trítio
19.
Br J Pharmacol ; 139(4): 695-704, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12812992

RESUMO

1 Neuropeptide Y Y(1) receptors are known to internalize following the binding of agonists. In the present study, a pseudopeptide Y(1) receptor antagonist, homodimeric Ile-Glu-Pro-Dpr-Tyr-Arg-Leu-Arg-Tyr-CONH(2) (GR231118), also induced Y(1) receptor internalization in human embryonic kidney (HEK293) cells. 2 We demonstrated first that both specifically bound radiolabeled antagonist ([(125)I]GR231118) and agonist ([(125)I][Leu(31), Pro(34)]PYY) underwent receptor-mediated sequestration/internalization in transfected HEK293 cells. 3 Agonist-induced Y(1) receptor internalization was dependent on clathrin-coated pits and was regulated in part by Gi/o-protein activation as revealed by pertussin toxin sensitivity. In contrast, antagonist-induced sequestration of Y(1) receptors was partly dependent on clathrin-coated pits, but independent from Gi/o-protein activation. 4 Exposure to high concentrations of agonist or antagonist caused a 50 and 75% loss of cell surface binding, respectively. The loss caused by the agonist rapidly recovered. This phenomenon was blocked by monensin, an inhibitor of endosome acidification, suggesting that cell surface receptor recovery is due to recycling. In contrast to the agonist, GR231118 induced a long-lasting sequestration of Y(1) receptors in HEK293 cells. 5 Immunofluorescence labeling indicated that following 40 min of incubation with either the agonist or the antagonist, Y(1) receptors followed markedly different intercellular trafficking pathways. 6 Taken together, these findings provided evidence that a pseudopeptide Y(1) receptor antagonist can induce long-lasting disappearance of cell surface receptors through a pathway distinct from the classical endocytic/recycling pathway followed by stimulation with an agonist.


Assuntos
Linhagem Celular , Receptores de Neuropeptídeo Y/agonistas , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Animais , Clatrina/metabolismo , Vesículas Revestidas por Clatrina/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Humanos , Radioisótopos do Iodo , Rim/citologia , Rim/embriologia , Peptídeo YY/farmacocinética , Peptídeos Cíclicos/farmacocinética , Radioisótopos , Ratos , Receptores de Neuropeptídeo Y/efeitos dos fármacos , Receptores de Neuropeptídeo Y/genética , Fatores de Tempo , Transfecção
20.
Can J Public Health ; 104(1): e80-1, 2012 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-23618110

RESUMO

Sex and gender have been demonstrated to influence all domains of health, from basic mechanisms of disease development to health service utilization. It is therefore no longer acceptable to ignore sex and gender issues in health research reports if these reports are to be deemed accurate. Funding agencies and journals have been identified as primary change agents in health research systems. Canada is making progress on the funding side of the equation--applicants to Canada's federal health research funding agency are required to justify why sex and gender are relevant or not to their research designs. We argue that it is now time for Canada's leading health research journals to follow suit. We have a unique opportunity in Canada to demonstrate leadership in doing science better with sex and gender--and we should not let it be missed.


Assuntos
Pesquisa Biomédica/organização & administração , Liderança , Publicações Periódicas como Assunto/normas , Canadá , Feminino , Humanos , Masculino , Projetos de Pesquisa
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa