Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Glia ; 36(1): 78-88, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11571786

RESUMO

Accumulating evidence, obtained largely in vitro, indicates that opioids regulate the genesis of neurons and glia and their precursors in the nervous system. Despite this evidence, few studies have assessed opioid receptor expression in identified cells within germinal zones or examined opioid effects on gliogenesis in vivo. To address this question, the role of opioids was explored in the subventricular zone (SVZ) and/or striatum of 2-5-day-old and/or adult ICR mice. The results showed that subpopulations of neurons, astrocytes, and oligodendrocytes in the SVZ and striatum differentially express mu-, delta-, and/or kappa-receptor immunoreactivity in a cell type-specific and developmentally regulated manner. In addition, DNA synthesis was assessed by examining 5-bromo-2'-deoxyuridine (BrdU) incorporation into glial and nonglial precursors. Morphine (a preferential mu-agonist) significantly decreased the number of BrdU-labeled GFAP(+) cells compared with controls or mice co-treated with naltrexone plus morphine. Alternatively, in S100beta(+) cells, morphine did not significantly decrease BrdU incorporation; however, significant differences were noted between mice treated with morphine and those treated with morphine plus naltrexone. Most cells were GFAP(-)/S100beta(-). When BrdU incorporation was assessed within the total population (glia and nonglia), morphine had no net effect, but naltrexone alone markedly increased BrdU incorporation. This finding suggests that DNA synthesis in GFAP(-)/S100beta(-) cells is tonically suppressed by endogenous opioids. Assuming that S100beta and GFAP, respectively, distinguish among younger and older astroglia, this implies that astroglial replication becomes increasingly sensitive to morphine during maturation, and suggests that opioids differentially regulate the development of distinct subpopulations of glia and glial precursors.


Assuntos
Astrócitos/metabolismo , Divisão Celular/fisiologia , Ventrículos Laterais/crescimento & desenvolvimento , Neostriado/crescimento & desenvolvimento , Neurônios/metabolismo , Oligodendroglia/metabolismo , Receptores Opioides/metabolismo , Proteínas S100 , Envelhecimento/fisiologia , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Animais Recém-Nascidos/anatomia & histologia , Animais Recém-Nascidos/crescimento & desenvolvimento , Animais Recém-Nascidos/metabolismo , Antígenos de Diferenciação/metabolismo , Antígenos de Superfície/metabolismo , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Bromodesoxiuridina/farmacocinética , Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Ventrículos Laterais/citologia , Ventrículos Laterais/metabolismo , Camundongos , Camundongos Endogâmicos ICR/anatomia & histologia , Camundongos Endogâmicos ICR/crescimento & desenvolvimento , Camundongos Endogâmicos ICR/metabolismo , Morfina/farmacologia , Naltrexona/farmacologia , Neostriado/citologia , Neostriado/metabolismo , Fatores de Crescimento Neural/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Oligodendroglia/citologia , Oligodendroglia/efeitos dos fármacos , Peptídeos Opioides/metabolismo , Receptores Opioides/efeitos dos fármacos , Subunidade beta da Proteína Ligante de Cálcio S100
2.
Glia ; 22(3): 249-59, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9482211

RESUMO

The diversity of opioid receptor expression was examined in astrocytes in low-density and non-dividing (confluent) cultures from the cerebral cortex, hippocampus, cerebellum, and striatum of 1-day-old mice. Mu, delta, and kappa opioid receptor expression was assessed in individual cells immunocytochemically, by using flow cytometry, and functionally by examining agonist-induced changes in intracellular calcium ([Ca2+]i). Significant spatial and temporal differences were evident in the pattern of expression of mu, delta, and kappa receptors among astrocytes. In low-density cultures, greater proportions of astrocytes expressed mu-opioid receptor immunoreactivity in the cerebral cortex and hippocampus (26-34%) than in the cerebellum or striatum (7-12%). At confluence, a greater percentage of astrocytes in cerebellar (26%) and striatal (30%) cultures expressed mu-immunoreactivity. Fewer astrocytes possessed delta-immunoreactivity in low-density striatal cultures (8%) compared to other regions (16-22%). The proportion of delta receptor-expressing astrocytes declined in the cerebellum but increased in the hippocampus. Kappa-opioid receptors were uniformly expressed by 27-34% of astrocytes from all regions, except in cortical cultures, where the proportion of kappa expressing cells was 38% at low-density and decreased to 22% at confluence. Selective mu (PLO 17; H-Tyr-Pro-Phe (N-Me) -D-Pro-NH2, delta ([D-Pen2, D-Pen5] enkephalin), or kappa (U50,488H; trans-(+/-)-3,4-Dichloro-N-methyl-N-[2-(1-pyrrolidinyl) cyclohexyl] benzeneacetamide methanesulfonate) opioid receptor agonists increased [Ca2+]i in subpopulations of astrocytes indicating the presence of functional receptors. Lastly, opioid receptor immunofluorescence varied during the cell division cycle. A greater proportion of astrocytes in the G2/M phase of the cell cycle were mu or delta receptor immunofluorescent than at G0/G1. When astrocytes were reversibly arrested in G1, significantly fewer cells expressed delta receptor immunofluorescence; however, upon reentry into the cell cycle immunofluorescent cells reappeared. In conclusion, opioid phenotype varies considerably among individual cultured astrocytes, and this diversity was determined by regional and developmental (age and cell cycle dependent) differences in the brain. These in vitro findings suggest astroglia contribute to regional and developmental idiosyncrasies in opioid function within the brain.


Assuntos
Astrócitos/química , Ciclo Celular/fisiologia , Receptores Opioides delta/biossíntese , Receptores Opioides kappa/biossíntese , Receptores Opioides mu/biossíntese , Receptores Opioides/biossíntese , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Fatores Etários , Analgésicos/farmacologia , Analgésicos não Narcóticos/farmacologia , Animais , Especificidade de Anticorpos , Astrócitos/citologia , Astrócitos/metabolismo , Química Encefálica/fisiologia , Cálcio/metabolismo , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Cerebelo/química , Cerebelo/citologia , Córtex Cerebral/química , Córtex Cerebral/citologia , Corpo Estriado/química , Corpo Estriado/citologia , Endorfinas/farmacologia , D-Penicilina (2,5)-Encefalina , Encefalinas/farmacologia , Citometria de Fluxo , Hipocampo/química , Hipocampo/citologia , Camundongos , Camundongos Endogâmicos ICR , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides/análise , Receptores Opioides/imunologia , Receptores Opioides delta/análise , Receptores Opioides delta/imunologia , Receptores Opioides kappa/análise , Receptores Opioides kappa/imunologia , Receptores Opioides mu/análise , Receptores Opioides mu/imunologia
3.
Brain Res ; 737(1-2): 175-87, 1996 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-8930364

RESUMO

To assess the role of kappa-opioid receptors in astrocyte development, the effect of kappa-agonists on the growth of astroglia derived from 1-2-day-old mouse cerebra was examined in vitro. kappa-Opioid receptor expression was assessed immunocytochemically (using KA8 and KOR1 antibodies), as well as functionally by examining the effect of kappa-receptor activation on intracellular calcium ([Ca2+]i) homeostasis and DNA synthesis. On days 6-7, as many as 50% of the astrocytes displayed kappa-receptor (KA8) immunoreactivity or exhibited increases in [Ca2+]i in response to kappa-agonist treatment (U69,593 or U50,488H). Exposure to U69,593 (100 nM) for 72 h caused a significant reduction in number and proportion of glial fibrillary acidic protein-immunoreactive astrocytes incorporating bromodeoxyuridine (BrdU) that could be prevented by co-administering the kappa-antagonist, nor-binaltorphimine (300 nM). In contrast, on day 14, only 5 or 14%, respectively, of the astrocytes were kappa-opioid receptor (KA8) immunoreactive or displayed functional increases in [Ca2+]i. Furthermore, U69,593 (100 nM) treatment failed to inhibit BrdU incorporation at 9 days in vitro. Experimental manipulations showed that kappa-receptor activation increases astroglial [Ca2+]i both through influx via L-type channels and through mobilization of intracellular stores (which is an important Ca2+ signaling pathway in cell division). Collectively, these results indicate that a subpopulation of developing astrocytes express kappa-opioid receptors in vitro, and suggest that the activation of kappa-receptors mobilizes [Ca2+]i and inhibits cell proliferation. Moreover, the proportion of astrocytes expressing kappa-receptors was greatest during a period of rapid cell growth suggesting that they are preferentially expressed by proliferating astrocytes.


Assuntos
Astrócitos/química , Benzenoacetamidas , Cálcio/metabolismo , Entorpecentes/farmacologia , Receptores Opioides kappa/genética , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida , Analgésicos/farmacologia , Animais , Astrócitos/citologia , Bromodesoxiuridina , Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Contagem de Células , Divisão Celular/efeitos dos fármacos , Células Cultivadas/química , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , DNA/biossíntese , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos ICR , Naloxona/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Nifedipino/farmacologia , Fenótipo , Pirrolidinas/farmacologia , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/análise , Tapsigargina/farmacologia , Fatores de Tempo
4.
Brain Res ; 720(1-2): 191-203, 1996 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-8782912

RESUMO

Morphine, a preferential mu-opioid receptor agonist, alters astroglial development by inhibiting cell proliferation and by promoting cellular differentiation. Although morphine affects cellular differentiation through a Ca(2+)-dependent mechanism, few studies have examined whether Ca2+ mediates the effect of opioids on cell proliferation, or whether a particular Ca2+ signal transduction pathway mediates opioid actions. Moreover, it is uncertain whether one or more opioid receptor types mediates the developmental effects of opioids. To address these questions, the present study examined the role of mu-opioid receptors and Ca2+ mobilization in morphine-induced astrocyte development. Morphine (1 microM) and non-morphine exposed cultures enriched in murine astrocytes were incubated in Ca(2+)-free media supplemented with < 0.005, 0.3, 1.0, or 3.0 mM Ca2+ ([Ca2+]o), or in unmodified media containing Ca2+ ionophore (A23187), nifedipine (1 microM), dantrolene (10 microM), thapsigargin (100 nM), or L-glutamate (100 microM) for 0-72 h. mu-Opioid receptor expression was examined immunocytochemically using specific (MOR1) antibodies. Intracellular Ca2+ ([Ca2+]i) was measured by microfluorometric analysis using fura-2. Astrocyte morphology and bromodeoxyuridine (BrdU) incorporation (DNA synthesis) were assessed in glial fibrillary acidic protein (GFAP) immunoreactive astrocytes. The results showed that morphine inhibited astroglial growth by activating mu-opioid receptors. Astrocytes expressed MOR1 immunoreactivity and morphine's actions were mimicked by the selective mu agonist PL017. In addition, morphine inhibited DNA synthesis by mobilizing [Ca2+]i in developing astroglia. At normal [Ca2+]o, morphine attenuated DNA synthesis by increasing [Ca2+]i; low [Ca2+]o (0.3 mM) blocked this effect, while treatment with Ca2+ ionophore or glutamate mimicked morphine's actions. At extremely low [Ca2+]o (< 0.005 mM), morphine paradoxically increased BrdU incorporation. Although opioids can increase [Ca2+]i in astrocytes through several pathways, not all affect DNA synthesis or cellular morphology. Nifedipine (which blocks L-type Ca2+ channels) did not prevent morphine-induced reductions in BrdU incorporation or cellular differentiation, while thapsigargin (which depletes IP3-sensitive Ca2+ stores) severely affected inhibited DNA synthesis and cellular differentiation-irrespective of morphine treatment. However, dantrolene (an inhibitor of Ca(2+)-dependent Ca2+ release) selectively blocked the effects of morphine. Collectively, the findings suggest that opioids suppress astroglial DNA synthesis and promote cellular hypertrophy by inhibiting Ca(2+)-dependent Ca2+ release from dantrolene-sensitive intracellular stores. This implies a fundamental mechanism by which opioids affect central nervous system maturation.


Assuntos
Astrócitos/metabolismo , Astrócitos/ultraestrutura , Cálcio/metabolismo , DNA/biossíntese , Morfina/farmacologia , Entorpecentes/farmacologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , Receptores Opioides mu/fisiologia , Animais , Antimetabólitos , Astrócitos/efeitos dos fármacos , Bromodesoxiuridina , Cálcio/fisiologia , Divisão Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Células Cultivadas , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos ICR , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo
5.
Brain Res Dev Brain Res ; 76(2): 189-96, 1993 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-8149585

RESUMO

Endogenous opioids and opiate drugs inhibit nervous system maturation, in part, by affecting the growth of astrocytes. Opiates inhibit astrocyte proliferation and cause premature differentiation. The emerging importance of Ca2+ in astrocyte function prompted us to explore whether opiates might affect astrocyte development by altering Ca2+ homeostasis. Astrocyte-enriched cultures were derived from newborn ICR mouse cerebra. Quantitative fluorescent measurements of intracellular free Ca2+ ([Ca2+]i) using Fura-2 as well as fluo-3 and computer-aided image analysis showed that 1 microM morphine significantly increased [Ca2+]i in flat, polyhedral, glial fibrillary acidic protein (GFAP) immunoreactive astrocytes at 2 and 6 min, and at 72 h. Co-administration of 3 microM naloxone blocked morphine-dependent increases in [Ca2+]i. Treatment with 1 microM concentrations of the kappa-opioid receptor agonist, U69,593, but not equimolar amounts of mu ([D-Ala2,MePhe4,Gly(ol)5]enkephalin)- or delta ([D-Pen2,D-Pen5]enkephalin)-opioid receptor agonists, significantly increased [Ca2+]i in astrocytes. To assess the role of Ca2+ in morphine-induced astrocyte differentiation, untreated and 1 microM morphine-treated astrocyte cultures were incubated for 5 days in < 0.01, 0.3, 1.0, or 3.0 mM extracellular Ca2+ ([Ca2+]o), or incubated with 1.0 mM [Ca2+]o in the presence of 1 microM of the Ca2+ ionophore, A23187. The areas of single astrocytes were measured and there was a positive correlation between astrocyte area and [Ca2+]o. Morphine had an additive effect on area and form factor measures when [Ca2+]o was 1.0 mM. High [Ca2+]o (3.0 mM) alone mimicked the action of morphine. Morphine alone had no effect on astrocyte area in the presence of 3.0 mM Ca2+.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Astrócitos/efeitos dos fármacos , Cálcio/fisiologia , Morfina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Astrócitos/citologia , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Tamanho Celular/efeitos dos fármacos , Células Cultivadas , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Endogâmicos ICR
6.
Neurosci Lett ; 157(1): 1-3, 1993 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-8233017

RESUMO

To determine whether there are regional differences in the ability of opiates to affect astrocyte proliferation, the effects of morphine were examined in astrocyte-enriched cultures from striatum, hippocampus and cerebral cortex derived from newborn mouse brains. Cultures from each region were continuously incubated in media alone (controls), or in media treated with 1 microM morphine, 1 microM morphine plus 3 microM naloxone, or 3 microM naloxone alone. Before harvesting at 6 days in vitro, cultures were exposed to [3H]thymidine (0.24 mu CI/ml for 16 h). Thymidine-labeling index was determined autoradiographically in flat, polyhedral (type 1) glial fibrillary acidic protein (GFAP)-immunoreactive astrocytes. Morphine significantly inhibited [3H]thymidine incorporation in astrocytes from all three brain regions, although regional differences in labeling indices were noted. The results show that opiates can intrinsically affect the proliferative rate of astrocytes from diverse brain regions.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , DNA/biossíntese , Morfina/farmacologia , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Corpo Estriado/citologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Naloxona/farmacologia , Timidina/metabolismo
7.
Brain Res Dev Brain Res ; 62(2): 245-55, 1991 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-1769103

RESUMO

Opioid-dependent changes in glial growth were characterized in primary dissociated and organotypic explant cultures of the developing mouse central nervous system (CNS) continuously grown in the presence of an endogenous opioid, [Met5]enkephalin, or an opiate drug, morphine. The glia in dissociated, astrocyte-enriched cultures derived from the cerebra of postnatal day 1, 3, or 5 mice, respectively, displayed age-dependent reductions in glial numbers that occurred at 3, 7, or 9 days in vitro (DIV) in response to continuous [Met5]enkephalin (10(-6) M) exposure. In contrast, in cultures derived from gestational day 19 mice, glial numbers were not reduced following continuous exposure to 10(-6) M [Met5]enkephalin during the first 7 days in vitro. An examination of [3H]thymidine incorporation by glial fibrillary acidic protein-(GFAP) immunoreactive astrocytes with flat (type 1) morphology in dissociated cultures derived from postnatal day 1 mice revealed that the reduction in glial numbers at 3 DIV was not immediately preceded by a reduction in the rate of [3H]thymidine incorporation at 2 DIV, although previous studies have shown that opioids inhibit the rate of [3H]thymidine incorporation by more mature astrocytes at 4 or 6 DIV. Early (i.e., at 2 to 3 DIV) changes in glial numbers may result from an inhibition of the proliferative rate of non-GFAP-containing glia or astrocyte precursors, or an enhanced rate of glial death. The rate of [3H]thymidine incorporation by GFAP-immunoreactive astrocytes with process-bearing (type 2) morphology was unchanged by opioid treatment. In separate experiments, a comparison of the area of growth of GFAP-immunoreactive astrocytes in paired symmetrical (right vs left) organotypic explant cultures demonstrated that opiates (i.e., 10(-5) M morphine) can inhibit astrocyte growth when the normal histiotypic organization of neurons and glia are maintained, and that there are regional differences in astrocyte responsiveness. Opioid-dependent alterations in astrocyte growth were mediated through specific opioid receptors since they were prevented by simultaneous treatment with (-)naloxone. The results suggest that the ability of opioids to modify glial growth is highly selective and varies depending on astrocyte type, as well as temporal and regional factors. Spatial and temporal differences in the response of developing glia to opioids may determine critical periods of CNS vulnerability to opioids in the maturing brain.


Assuntos
Encéfalo/citologia , Embrião de Mamíferos/metabolismo , Endorfinas/fisiologia , Neuroglia/fisiologia , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Astrócitos/fisiologia , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Contagem de Células , Técnicas de Cultura , DNA/metabolismo , Camundongos , Camundongos Endogâmicos ICR
8.
J Neurosci Res ; 29(4): 538-48, 1991 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1665190

RESUMO

To determine whether one or more opioid receptor types might be preferentially involved in gliogenesis, primary mixed glial cultures derived from mouse cerebra were continuously treated with varying concentrations of opioid agonists selective for mu (mu), i.e., DAGO ([D-Ala2, MePhe4, Gly(ol)5]enkephalin), delta (delta), i.e., DPDPE ([D-PEN2,D-PEN5]enkephalin), or kappa (kappa), i.e., U69,593, opioid receptor types. In addition, a group of cultures was treated with [Met5]-enkephalin, an agonist for delta opioid receptors as well as putative zeta (zeta) opioid receptors. Opioid-dependent changes in growth were assessed by examining alterations in (1) the number of cells in mixed glial cultures at 3, 6, and 8 days in vitro (DIV), (2) [3H]thymidine incorporation by glial fibrillary acidic protein (GFAP) immunoreactive, flat (type 1) astrocytes at 6 DIV, and (3) the area and form factor of GFAP-immunoreactive, flat (type 1) astrocytes. DPDPE at 10(-8) or 10(-10) M, as well as [Met5]-enkephalin at 10(-6), 10(-8), or 10(-10) M, significantly reduced the total number of glial cells in culture; but this effect was not observed with DAGO or U69,593 (both at 10(-6), 10(-8), or 10(-10) M). Equimolar concentrations (i.e., 10(-6) M) of [Met5]enkephalin or U69,593, but not DPDPE or DAGO, suppressed the rate of [3H]thymidine incorporation by GFAP-immunoreactive, flat (type 1) astrocytes. DAGO had no effect on growth, although in previous studies morphine was found to inhibit glial numbers and astrocyte DNA synthesis. [Met5]enkephalin (10(-6) M) was the only agonist to significantly influence astrocyte area. Collectively, these results indicate that delta (and perhaps mu) opioid receptor agonists reduce the total number of cells in mixed glial cultures; while [Met5]enkephalin-responsive (and perhaps kappa-responsive) opioid receptors mediate DNA synthesis in astrocytes. This implies that delta opioid receptors, as well as [Met5]enkephalin-sensitive, non-delta opioid receptors, mediate opioid-dependent regulation of astrocyte and astrocyte progenitor growth. These data support the concept that opioid-dependent changes in central nervous system growth are the result of endogenous opioid peptides acting through multiple opioid receptor types.


Assuntos
Analgésicos/farmacologia , Astrócitos/citologia , Benzenoacetamidas , Neuroglia/citologia , Receptores Opioides/fisiologia , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Células Cultivadas , Replicação do DNA/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina , D-Penicilina (2,5)-Encefalina , Encefalina Metionina/farmacologia , Encefalinas/farmacologia , Proteína Glial Fibrilar Ácida/análise , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos ICR , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Pirrolidinas/farmacologia , Receptores Opioides/efeitos dos fármacos , Receptores Opioides delta , Receptores Opioides kappa , Receptores Opioides mu , Timidina/metabolismo
9.
Brain Res Dev Brain Res ; 60(1): 1-7, 1991 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-1914143

RESUMO

To determine whether exogenous opiate drugs with abuse liability directly modify neural growth, the present study investigated the effects of morphine on astrocyte proliferation and differentiation in primary cultures of murine glial cells. The results indicate that morphine decreases glial cell production in a dose-dependent, naloxone-reversible manner. Most notably, gliogenesis virtually ceased in the presence of 10(-6) M morphine during the first week in culture, whereas 10(-8) M or 10(-10) M morphine caused an intermediate suppression of growth compared to control or 10(-6) M morphine treated cultures. Moreover, morphine treatment inhibited [3H]thymidine incorporation by glial fibrillary acidic protein (GFAP) immunoreactive, flat (type 1) astrocytes, suggesting that the decrease in glial cell production was due in part to an inhibition of astrocyte proliferation. Morphine also caused significant increases in both cytoplasmic area and process elaboration in flat (type 1) astrocytes indicating greater morphologic differentiation. In the above experiments, morphine-dependent alterations in astrocyte growth were antagonized by naloxone, indicating that morphine action was mediated by specific opioid receptors. These observations suggest that opiate drugs can directly modify neural growth by influencing two critical developmental events in astrocytes, i.e., inhibiting proliferation and inducing morphologic differentiation.


Assuntos
Astrócitos/efeitos dos fármacos , Morfina/farmacologia , Sistema Nervoso/crescimento & desenvolvimento , Neuroglia/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Proteína Glial Fibrilar Ácida , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos ICR , Naloxona/farmacologia , Sistema Nervoso/efeitos dos fármacos , Timidina/metabolismo
10.
J Neurosci Methods ; 36(2-3): 119-26, 1991 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-1712058

RESUMO

To determine whether cultured astrocytes express opioid gene mRNA, a method was developed for co-localizing a cell-type specific immunocytochemical marker for astrocytes, glial fibrillary acidic protein (GFAP), and proenkephalin mRNA in situ hybridization signal using high affinity cRNA probes. GFAP immunoreactivity and proenkephalin mRNA hybridization reaction were examined in intact glial cell preparations from neonatal mice that were cultured for 4-6 days prior to fixation. The double labelling method described herein permits the unambiguous identification of mRNA expression in specific populations of intact cultured cells using cell type-specific markers.


Assuntos
Astrócitos/metabolismo , Encefalinas/biossíntese , Precursores de Proteínas/biossíntese , RNA Mensageiro/biossíntese , Animais , Proteína Glial Fibrilar Ácida/biossíntese , Proteína Glial Fibrilar Ácida/imunologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos ICR , Hibridização de Ácido Nucleico , RNA , Sondas RNA , RNA Complementar
11.
Brain Res ; 522(2): 347-53, 1990 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-2224534

RESUMO

To identify the possible cellular sites of opioid gene expression during ontogeny, proenkephalin mRNA and enkephalin peptide expression were examined, respectively, by in situ hybridization and immunocytochemistry in organotypic explants of rat cerebellum and in astrocyte-enriched cultures of murine cerebral hemispheres. High levels of proenkephalin mRNA and enkephalin immunoreactivity were detected in immature cells identified as astrocytes. Double-labeling studies combining in situ hybridization and immunocytochemical localization of the astrocytic marker, glial fibrillary acidic protein, provided direct evidence that proenkephalin mRNA is expressed by astrocytes in culture. Based on previous studies that Met-enkephalin can inhibit astrocyte growth in vitro, the present results suggest that proenkephalin gene expression by astrocytes is important during central nervous system maturation.


Assuntos
Astrócitos/química , Encefalinas/análise , Encefalinas/genética , Precursores de Proteínas/genética , RNA Mensageiro/análise , Animais , Células Cultivadas , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos ICR , Hibridização de Ácido Nucleico , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos
12.
Life Sci ; 46(2): 91-8, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2299973

RESUMO

The action of met-enkephalin on the growth of astrocytes in mixed-glial cultures was examined. Primary, mixed-glial cultures were isolated from 1 day-old mouse cerebral hemispheres and continuously treated with either basal growth media (controls), 1 microM met-enkephalin, 1 microM met-enkephalin plus the opioid antagonist naloxone (3 microM), or naloxone alone (3 microM). Absolute numbers of neural cells were counted in unstained preparations, while combined [3H]-thymidine autoradiography and glial fibrillary acid protein (GFAP) immunocytochemistry was performed to identify specific changes in astrocytes. When compared to control and naloxone treated cultures, met-enkephalin caused a significant decrease in both total cell numbers, and in [3H]-thymidine incorporation by GFAP-positive cells with flat morphology. These results indicate that met-enkephalin suppresses astrocyte growth in culture.


Assuntos
Astrócitos/metabolismo , DNA/efeitos dos fármacos , Encefalina Metionina/farmacologia , Neuroglia/efeitos dos fármacos , Animais , Contagem de Células , Células Cultivadas , DNA/biossíntese , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos ICR , Neuroglia/citologia , Neuroglia/metabolismo , Timidina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA