Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Biol Chem ; 286(26): 23280-95, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21515676

RESUMO

Type I (α and ß) interferons (IFNs) elicit antiproliferative and antiviral activities via the surface receptor IFNAR. Serendipitous observations in transgenic mice in 1988 strongly suggested that IFNα/ß overexpression in the testis disrupts spermatogenesis. Here, we compare a new mouse strain transgenic for IFNß (Tg10) and a sister strain lacking the IFNAR1 subunit of IFNAR (Tg10-Ifnar1(-/-)), both strains expressing the transgene in the testis. The main source of IFNß RNA was the spermatid population. Importantly, the Tg10 mice, but not the double mutant Tg10-Ifnar1(-/-), showed altered spermatogenesis. The first IFNAR-dependent histological alteration was a higher apoptosis index in all germ cell categories apart from non-dividing spermatogonia. This occurred 3 weeks after the onset of IFNß production at postnatal day 20 and in the absence of somatic cell defects in terms of cell number, expression of specific cell markers, and hormonal activities. Several known interferon-stimulated genes were up-regulated in Tg10 Sertoli cells and prepachytene germ cells but not in pachytene spermatocytes and spermatids. In concordance with this, pachytene spermatocytes and spermatids isolated from wild-type testes did not display measurable amounts of IFNAR1 and phosphorylated STAT1 upon IFNß challenge in vitro, suggesting hyporesponsiveness of these cell types to IFN. At day 60, Tg10 males were sterile, and Sertoli cells showed increased amounts of anti-Mullerian hormone and decreased production of inhibin B, both probably attributable to the massive germ cell loss. Type I interferon signaling may lead to idiopathic infertilities by affecting the interplay between germ cells and Sertoli cells.


Assuntos
Infertilidade Masculina/metabolismo , Interferon beta/biossíntese , Túbulos Seminíferos/metabolismo , Transdução de Sinais , Espermatogênese , Espermatozoides/metabolismo , Animais , Hormônio Antimülleriano/genética , Hormônio Antimülleriano/metabolismo , Apoptose , Feminino , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Inibinas/genética , Inibinas/metabolismo , Interferon beta/genética , Masculino , Camundongos , Camundongos Transgênicos , Estágio Paquíteno/genética , Fosforilação/genética , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Túbulos Seminíferos/patologia , Células de Sertoli/metabolismo , Células de Sertoli/patologia , Espermatozoides/patologia , Fatores de Tempo
2.
J Neurosci ; 28(20): 5207-17, 2008 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-18480277

RESUMO

The barrel field of the somatosensory cortex constitutes a well documented example of anatomofunctional compartmentalization and activity-dependent interaction between neurons and astrocytes. In astrocytes, intercellular communication through gap junction channels composed by connexin 43 and 30 underlies a network organization. Immunohistochemical and electrophysiological experiments were undertaken to determine the coupling properties of astrocyte networks in layer IV of the developing barrel cortex. The expression of both connexins was found to be enriched within barrels compared with septa and other cortical layers. Combination of dye-coupling experiments performed with biocytin and immunostaining with specific cell markers demonstrated that astrocytic networks do not involve neurons, oligodendrocytes or NG2 cells. The shape of dye coupling was oval in the barrel cortex whereas it was circular in layer IV outside the barrel field. Two-dimensional analysis of these coupling areas indicated that gap junctional communication was restricted from a barrel to its neighbor. Such enrichment of connexin expression and transversal restriction were not observed in a transgenic mouse lacking the barrel organization, whereas they were both observed in a double-transgenic mouse with restored barrels. Direct observation of sulforhodamine B spread indicated that astrocytes located between two barrels were either weakly or not coupled, whereas coupling within a barrel was oriented toward its center. These observations indicated a preferential orientation of coupling inside the barrels resulting from subpopulations of astrocytes with different coupling properties that contribute to shaping astrocytic networks. Such properties confine intercellular communication in astrocytes within a defined barrel as previously reported for excitatory neuronal circuits.


Assuntos
Astrócitos/fisiologia , Comunicação Celular/fisiologia , Junções Comunicantes/fisiologia , Rede Nervosa/fisiologia , Córtex Somatossensorial/fisiologia , Vias Aferentes/fisiologia , Animais , Astrócitos/ultraestrutura , Conexina 30 , Conexina 43/metabolismo , Conexinas/metabolismo , Difusão , Junções Comunicantes/ultraestrutura , Imuno-Histoquímica , Lisina/análogos & derivados , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Rede Nervosa/ultraestrutura , Neurônios/fisiologia , Neurônios/ultraestrutura , Técnicas de Cultura de Órgãos , Rodaminas , Córtex Somatossensorial/ultraestrutura , Nervo Trigêmeo/fisiologia , Vibrissas/fisiologia
3.
J Mol Cell Cardiol ; 46(4): 587-95, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19162038

RESUMO

The potential role of serotonin (5-HT) in cardiac function has generated much interest in recent years. In particular, the need for a tight regulation of 5-HT to maintain normal cardiovascular activity has been demonstrated in different experimental models. However, it remains unclear how increased levels of 5-HT could contribute to the development of cardiac hypertrophy. Availability of 5-HT depends on the mitochondrial enzyme monoamine oxidase A (MAO-A). Therefore, we investigated the consequences of MAO-A deletion on ventricular remodeling in the model of aortic banding in mice. At baseline, MAO-A deletion was associated with an increase in whole blood 5-HT (39.4+/-1.9 microM vs. 24.0+/-0.9 microM in KO and WT mice, respectively). Cardiac 5-HT(2A), but not 5-HT(2B) receptors were overexpressed in MAO-A KO mice, as demonstrated by real-time PCR and Western-blot experiments. After aortic banding, MAO-A KO mice demonstrated greater increase in heart wall thickness, heart to body weight ratios, cardiomyocyte cross-section areas, and myocardial fibrosis compared to WT. Exacerbation of hypertrophy in KO mice was associated with increased amounts of 5-HT in the heart. In order to determine the role of 5-HT and 5-HT(2A) receptors in ventricular remodeling in MAO-A KO mice, we administered the 5-HT(2A) receptor antagonists ketanserin (1 mg/kg/day) or M100907 (0.1 mg/kg/day) during 4 weeks of aortic banding. Chronic administration of these antagonists strongly prevented exacerbation of ventricular hypertrophy in MAO-A KO mice. These results show for the first time that regulation of peripheral 5-HT by MAO-A plays a role in ventricular remodeling via activation of 5-HT(2A) receptors.


Assuntos
Cardiomegalia/enzimologia , Cardiomegalia/patologia , Deleção de Genes , Ventrículos do Coração/patologia , Monoaminoxidase/genética , Pressão , Serotonina/metabolismo , Estresse Fisiológico , Animais , Aorta/patologia , Pressão Sanguínea/efeitos dos fármacos , Cardiomegalia/diagnóstico por imagem , Cardiomegalia/fisiopatologia , Fibrose , Fluorbenzenos/administração & dosagem , Fluorbenzenos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/enzimologia , Ventrículos do Coração/fisiopatologia , Ketanserina/administração & dosagem , Ketanserina/farmacologia , Camundongos , Camundongos Knockout , Monoaminoxidase/metabolismo , Miocárdio/enzimologia , Miocárdio/patologia , Piperidinas/administração & dosagem , Piperidinas/farmacologia , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Serotonina/sangue , Estresse Fisiológico/efeitos dos fármacos , Ultrassonografia
4.
Circ Res ; 100(1): 41-9, 2007 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-17158340

RESUMO

The mitochondrial enzyme monoamine oxidase (MAO), its isoform MAO-A, plays a major role in reactive oxygen species-dependent cardiomyocyte apoptosis and postischemic cardiac damage. In the current study, we investigated whether sphingolipid metabolism can account for mediating MAO-A- and reactive oxygen species-dependent cardiomyocyte apoptosis. In H9c2 cardiomyoblasts, MAO-A-dependent reactive oxygen species generation led to mitochondria-mediated apoptosis, along with sphingosine kinase-1 (SphK1) inhibition. These phenomena were associated with generation of proapoptotic ceramide and decrease in prosurvival sphingosine 1-phosphate. These events were mimicked by inhibition of SphK1 with either pharmacological inhibitor or small interfering RNA, as well as by extracellular addition of C(2)-ceramide or H(2)O(2). In contrast, enforced expression of SphK1 protected H9c2 cells from serotonin- or H(2)O(2)-induced apoptosis. Analysis of cardiac tissues from wild-type mice subjected to ischemia/reperfusion revealed significant upregulation of ceramide and inhibition of SphK1. It is noteworthy that SphK1 inhibition, ceramide accumulation, and concomitantly infarct size and cardiomyocyte apoptosis were significantly decreased in MAO-A-deficient animals. In conclusion, we show for the first time that the upregulation of ceramide/sphingosine 1-phosphate ratio is a critical event in MAO-A-mediated cardiac cell apoptosis. In addition, we provide the first evidence linking generation of reactive oxygen species with SphK1 inhibition. Finally, we propose sphingolipid metabolites as key mediators of postischemic/reperfusion cardiac injury.


Assuntos
Apoptose/fisiologia , Monoaminoxidase/metabolismo , Miócitos Cardíacos/fisiologia , Estresse Oxidativo/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Ceramidas/metabolismo , Ceramidas/farmacologia , Regulação para Baixo , Resistência a Medicamentos/fisiologia , Peróxido de Hidrogênio/farmacologia , Lisofosfolipídeos/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias Cardíacas/fisiologia , Monoaminoxidase/deficiência , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Oxidantes/farmacologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Serotonina/farmacologia , Esfingolipídeos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Regulação para Cima
5.
Sleep ; 30(10): 1295-302, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17969463

RESUMO

STUDY OBJECTIVES: Alterations in the serotonin (5-HT) system have been suggested as a mechanism of sleep apnea in humans and rodents. The objective is to evaluate the contribution of 5-HT to this disorder. DESIGN: We studied sleep and breathing (whole-body plethysmography) in mutant mice that lack monoamine oxidase A (MAOA) and have increased concentrations of monoamines, including 5-HT. MEASUREMENTS AND RESULTS: Compared to wild-type mice, the mutants showed similar amounts of slow wave sleep (SWS) and rapid eye movement sleep (REMS), but exhibited a 3-fold increase in SWS and REMS apnea indices. Acute administration of the MAOA inhibitor clorgyline decreased REMS amounts and increased the apnea index in wild-type but not mutant mice. Parachlorophenylalanine, a 5-HT synthesis inhibitor, reduced whole brain concentrations of 5-HT in both strains, and induced a decrease in apnea index in mutant but not wild-type mice. CONCLUSION: Our results show that MAOA deficiency is associated with increased sleep apnea in mice and suggest that an acute or chronic excess of 5-HT contributes to this phenotype.


Assuntos
Monoaminoxidase/deficiência , Síndromes da Apneia do Sono/enzimologia , Fases do Sono , Animais , Encéfalo/enzimologia , Clorgilina/farmacologia , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Monoaminoxidase/genética , Neurônios/enzimologia , Pletismografia , Receptores 5-HT1 de Serotonina/metabolismo , Fases do Sono/efeitos dos fármacos
6.
Aggress Behav ; 33(1): 1-6, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17441000

RESUMO

Monoamine oxidase A (MAO A) degrades serotonin, dopamine and noradrenaline, factors critically involved in the regulation of aggression. Different kinds of aggression were investigated in Tg8, a transgenic mouse strain lacking a functional MAO A gene. MAO A-deficient mice differ from wild-type C3H/HeJ (C3H) in terms of showing higher territorial, predatory and isolation-induced aggression. Tg8 demonstrated shorter latencies to cricket killing and to the first attack after 6 weeks isolation than C3H mice. In the resident-intruder paradigm, MAO A-lacking mice were more aggressive than C3H when tested as intruders. In contrast to C3H, attack in Tg8 mice did not depend on different aggressiveness of intruders of BALB/c, A/Sn and C3H strains. Tg8 mice displayed no increase in aggression but demonstrated reduced social investigation towards anesthetized, as well as towards juvenile BALB/c males. Thus, MAO A deficiency in Tg8 mice is accompanied by increased expression of different kinds of aggression, as well as by disruption of normal pattern of social interaction.


Assuntos
Agressão , Monoaminoxidase/deficiência , Comportamento Social , Animais , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Comportamento Predatório , Isolamento Social
7.
J Neurosci ; 25(3): 706-10, 2005 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-15659608

RESUMO

In the mouse somatosensory cortex, thalamocortical axons (TCAs) corresponding to individual whiskers cluster into restricted barrel domains during the first days of life. If whiskers are lesioned before that time, the cortical space devoted to the afferents from the damaged whisker shrinks and becomes occupied by thalamocortical afferents from neighboring unlesioned whiskers. This plasticity ends by postnatal day 3 (P3) to P4 when barrels emerge. To test whether TCA development and lesion-induced plasticity are linked, we used monoamine oxidase A knock-out (MAOA-KO) mice in which normal TCA development is halted by an excess of serotonin. Normal TCA development can be restored when serotonin levels are lowered by parachlorophenylalanine (PCPA). By varying the time of PCPA administration, we found that barrel development can be reinitiated until P11, although the emergence of TCA clusters becomes gradually slower and less complete. In mice in which barrels emerge 3 d later than the normal schedule, at P6 instead of P3, we examined lesion-induced plasticity. We find a progressive decline of the lesion-induced plasticity and a closure at P3, similar to normal mice, showing that this plasticity is not influenced by an excess of serotonin levels. Thus, in MAOA-KO mice, the emergence of barrel patterning can be delayed without a concomitant delay in lesion-induced plasticity, and the cortical space devoted to one whisker representation cannot be modified by the periphery once patterning is imprinted in the subcortical relays. We conclude that the closure of the lesion-induced plasticity period in the barrelfield is probably not determined at the cortical level.


Assuntos
Plasticidade Neuronal/fisiologia , Córtex Somatossensorial/crescimento & desenvolvimento , Tálamo/crescimento & desenvolvimento , Vibrissas/inervação , Animais , Axônios/fisiologia , Padronização Corporal , Mapeamento Encefálico , Fenclonina/farmacologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Monoaminoxidase/genética , Vias Neurais/crescimento & desenvolvimento , Serotonina/metabolismo , Fatores de Tempo , Triptofano Hidroxilase/antagonistas & inibidores , Vibrissas/lesões
8.
J Comp Neurol ; 495(5): 607-23, 2006 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-16498683

RESUMO

Abnormally high brain 5-HT levels in monoamine oxidase-A knockout (MAO-A KO) mouse neonates raise the question of whether the distribution and density of the 5-HT1A receptors (5-HT1AR) expressed in the brain by postnatal day P7 are affected and, if so, whether the 5-HT1A autoreceptors in the dorsal raphe are modified in the same way as the postsynaptic 5-HT1AR present in raphe target structures. [3H]8-OH-DPAT binding and quantitative autoradiography were performed to answer these questions. Binding specificity was first confirmed in adult wild-type mice and rat brain sections. 5-HT1AR binding was then analyzed in four MAO-A mutant vs. five wild-type neonatal brains, from olfactory bulb to cervical cord. Among 12 structures expressing postsynaptic 5-HT1AR in wild-type neonates, the highest densities involved the retrosplenial cortex, entorhinal cortex, and septum (52-46 fmol/mg tissue); low densities occurred in the hippocampus and spinal cord (24 fmol/mg tissue); in addition, the raphe autoreceptor density was only 20 fmol/mg tissue. In mutants, the distribution of postsynaptic 5-HT1AR was unchanged, but an overall decrease in density occurred (-32% to -63%); the raphe autoreceptors decreased in mutants by at least -79%. Data are discussed with reference to the ectopic 5-HT uptake and accumulation reported to occur during the first 10 postnatal days in wild-type and MAO-A KO mice. As previously suggested to explain the raphe autoreceptor loss in 2-month-old MAO-A KO mice, the overall 5-HT1AR down-regulation in mutant pups probably results from extracellular 5-HT excess in both raphe and target structures. The greater the 5-HT excess, the more the functional receptor density decreases.


Assuntos
Encéfalo/metabolismo , Bulbo/metabolismo , Monoaminoxidase/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Medula Espinal/metabolismo , Animais , Animais Recém-Nascidos , Sítios de Ligação , Encéfalo/anatomia & histologia , Bulbo/citologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Monoaminoxidase/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Medula Espinal/citologia , Proteínas Vesiculares de Transporte de Monoamina/metabolismo
9.
Psychoneuroendocrinology ; 31(2): 179-86, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16112493

RESUMO

The effect of a lack of the gene encoding monoamine oxidase A (MAO A) in transgenic Tg 8 mice on the corticosterone response to restraint, cold, water deprivation-induced, or social acute stress as well as chronic variable stress was studied. It was found that Tg 8 mice with genetic MAO A knockout and wild-type C3H/HeJ (C3H) strain showed similar plasma corticosterone resting level. MAO A knockout mice differed from C3H mice by attenuated response to restraint (60 min), cold (4 degrees C, 60 min), and water deprivation (48 h) as well as to a chronic (15 days) variable stress. No difference between Tg 8 and C3H strains in the response to psychosocial stress (encounters for 30 min of six previously isolated mice) has been found. ACTH administration to dexamethasone-pretreated mice produced a similar corticosterone effect in Tg 8 and C3H mice, indicating that the decreased stress response in MAO A-deficient mice was due rather to the central mechanisms regulating stress-induced ACTH release than to adrenocortical responsiveness to ACTH.


Assuntos
Córtex Suprarrenal/enzimologia , Corticosterona/sangue , Monoaminoxidase/metabolismo , Estresse Fisiológico/enzimologia , Estresse Psicológico/enzimologia , Córtex Suprarrenal/fisiopatologia , Hormônio Adrenocorticotrópico/fisiologia , Análise de Variância , Animais , Crioterapia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Monoaminoxidase/genética , Meio Social , Estresse Fisiológico/sangue , Estresse Fisiológico/genética , Estresse Psicológico/sangue , Estresse Psicológico/genética
10.
Neurosci Lett ; 401(1-2): 49-54, 2006 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-16638624

RESUMO

Gene targeting approaches greatly facilitate insight into the functioning of monoamine transporters, the targets of potent antidepressants. The serotonin transporter (5-HTT) is the molecular target of a large number of antidepressants. To assess the clearance of serotonin (5-HT) in the absence of the 5-HTT, we have generated double knockout mice lacking both the 5-HTT and the catabolizing enzyme monoamine oxidase A (MAOA). We found aberrant 5-HT accumulation in the striatum of these MAOA/5-HTT double knockout mice. By additional ablation of the dopamine transporter (DAT), this aberrant 5-HT accumulation was abolished in MAOA/5-HTT/DAT triple knockout mice. Thus, aberrant uptake of 5-HT occurs in dopaminergic terminals under conditions of elevated 5-HT levels, and this aberrant uptake is mediated by the DAT. These findings have important consequences for antidepressant therapy, since during treatment of depression with selective serotonin reuptake inhibitors, clearance of 5-HT by dopaminergic neurons may reduce the desired therapeutic elevation of extracellular 5-HT levels. This provides a molecular rationale for improving antidepressant efficacy by additional pharmacological inhibition of the DAT.


Assuntos
Encéfalo/metabolismo , Dopamina/metabolismo , Monoaminoxidase/genética , Neurônios/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Serotonina/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Química Encefálica/genética , Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/metabolismo , Transtorno Depressivo/fisiopatologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Feminino , Masculino , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
11.
Neuropharmacology ; 105: 142-153, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26764241

RESUMO

Pharmacological studies have yielded valuable insights into the role of the serotonin 2A (5-HT2A) receptor in major depressive disorder (MDD) and antidepressant drugs (ADs) response. However, it is still unknown whether genetic variants in the HTR2A gene affect the therapeutic outcome of ADs and the mechanism underlying the regulation of such response remains poorly described. In this context, a translational human-mouse study offers a unique opportunity to address the possibility that variations in the HTR2A gene may represent a relevant marker to predict the efficacy of ADs. In a first part of this study, we investigated in depressed patients the effect of three HTR2A single nucleotide polymorphisms (SNPs), selected for their potential functional consequences on 5-HT2A receptor (rs6313, rs6314 and rs7333412), on response and remission rates after 3 months of antidepressant treatments. We also explored the consequences of the constitutive genetic inactivation of the 5-HT2A receptor (i.e. in 5-HT2A(-/-) mice) on the activity of acute and prolonged administration of SSRIs. Our clinical data indicate that GG patients for the rs7333412 SNP were less prone to respond to ADs than AA/AG patients. In the preclinical study, we demonstrated that the 5-HT2A receptor exerts an inhibitory influence on the neuronal activity of the serotonergic system after acute administration of SSRIs. However, while the chronic administration of the SSRIs escitalopram or fluoxetine elicited a progressive increased in the firing rate of 5-HT neurons in 5-HT2A(+/+) mice, it failed to do so in 5-HT2A(-/-) mutants. These electrophysiological impairments were associated with a decreased ability of the chronic administration of fluoxetine to stimulate hippocampal plasticity and to produce antidepressant-like activities. Genetic loss of the 5-HT2A receptor compromised the activity of chronic treatment with SSRIs, making this receptor a putative marker to predict ADs response.


Assuntos
Antidepressivos/uso terapêutico , Transtorno Depressivo Maior/tratamento farmacológico , Transtorno Depressivo Maior/genética , Receptor 5-HT2A de Serotonina/genética , Receptor 5-HT2A de Serotonina/fisiologia , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Adolescente , Adulto , Idoso , Animais , Sobrevivência Celular/efeitos dos fármacos , Citalopram/administração & dosagem , Núcleo Dorsal da Rafe/efeitos dos fármacos , Núcleo Dorsal da Rafe/fisiologia , Fluoxetina/administração & dosagem , Genótipo , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Humanos , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Polimorfismo de Nucleotídeo Único , Pesquisa Translacional Biomédica , Adulto Jovem
12.
J Neurosci ; 22(19): 8541-52, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12351728

RESUMO

In the rodent primary somatosensory cortex, the thalamocortical axons (TCAs) are organized into clusters that correspond to functional units in the periphery. Around these axons, neurons in layer IV aggregate as barrels. To understand how this organization emerges, we analyzed TCA development in mice that do not form barrels, the monoamine oxidase A knock-out (MAOA-KO), and in MAOA/5-HT(1B) receptor double-KO mice, which have a restored barrel field. We show that TCAs already attain cortical layer IV on the day of birth. They are uniformly distributed in this layer from postnatal day 0 (P0) to P2 and secondarily coalesce into barrel domains in layer IV, over a 3 d period (P3-P5), with no prepatterning in the deeper layers. In MAOA-KO mice, the uniform distribution of the TC projection is maintained, and no axon clusters emerge. Individual TCA arbors were traced after carbocyanine injections. At P1, TCAs were poorly branched and covered variable tangential widths, encompassing one to two prospective barrels. At P7 the number of TCA branches increased 10-fold in layer IV and became restricted to one barrel. In MAOA-KO mice, there was a 50% reduction of the TCA terminal branches in layer IV, with a 40% increase in their tangential extent. These defects were corrected in the MAOA/5-HT(1B) double knock-out mice, indicating an effect of the presynaptic 5-HT(1B) receptor on axon branching. Our results indicate that the barrel-deficient phenotype of MAOA-KO mice results from an altered refinement of the TCA arbors in their target layer IV, involving branch elaboration and collateral retraction during early postnatal life.


Assuntos
Axônios/ultraestrutura , Proteínas de Membrana Transportadoras , Monoaminoxidase/deficiência , Proteínas do Tecido Nervoso , Córtex Somatossensorial/citologia , Tálamo/citologia , Animais , Axônios/metabolismo , Proteínas de Transporte/biossíntese , Corantes , Imuno-Histoquímica , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Monoaminoxidase/biossíntese , Monoaminoxidase/genética , Neurônios/metabolismo , Neurônios/ultraestrutura , Fenótipo , Receptor 5-HT1B de Serotonina , Receptores de Serotonina/biossíntese , Receptores de Serotonina/deficiência , Receptores de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina , Córtex Somatossensorial/crescimento & desenvolvimento
13.
J Neurosci ; 22(12): 4987-5000, 2002 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12077195

RESUMO

Mice lacking monoamine oxidase A (MAOA) display high levels of brain serotonin during the first postnatal week, causing an exuberant outgrowth of thalamocortical axons (TCAs) in layer IV of the somatosensory cortex (S1). We asked whether this exuberance is attributable to abnormal TrkB signaling, because modulation of TrkB signaling during a critical period dramatically influences the segregation and the morphology of TCAs in layer IV of the visual cortex. Using in situ hybridization and ELISA immunoassays, we showed that the levels of trkB mRNA and BDNF and neurotrophin-4 (NT-4) proteins are normal in the thalamus and the cortex of mice lacking MAOA during barrel field formation. Because the release of BDNF and NT-4 could be abnormal in MAOA knock-out (KO) mice, we tested whether abnormal TrkB signaling is required for TCA exuberance in MAOA-KO mice by generating mice lacking both trkB and MAOA. Surprisingly, these mice exhibited more severe phenotypes than those found in MAOA-KO mice: a widespread tangential expansion of TCAs in layer IV of the cortex, resulting in a fusion of all sensory representations and a radial expansion of TCAs in layers II-III of the cortex. Careful examination of mice lacking trkB alone revealed subtle alterations of TCAs, with abnormal invasion of layer III. This study reveals the following: (1) expression of trkB, BDNF, and NT-4 are not modulated by an excess of serotonin during barrel formation, (2) TrkB signaling limits branching of TCAs in inappropriate supragranular cortical layers, and (3) serotonin and TrkB signaling act together to cluster thalamocortical axons in layer IV.


Assuntos
Axônios/ultraestrutura , Receptor trkB/metabolismo , Serotonina/biossíntese , Córtex Somatossensorial/crescimento & desenvolvimento , Córtex Somatossensorial/metabolismo , Tálamo/crescimento & desenvolvimento , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Morte Celular , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/análise , Ácido Hidroxi-Indolacético/metabolismo , Cinética , Camundongos , Camundongos Knockout , Monoaminoxidase/genética , Fatores de Crescimento Neural/metabolismo , RNA Mensageiro/biossíntese , Receptor trkB/genética , Transdução de Sinais , Córtex Somatossensorial/citologia , Tálamo/citologia , Tálamo/metabolismo
14.
J Comp Neurol ; 442(4): 331-47, 2002 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-11793338

RESUMO

Monoamine oxidases A (MAOA) and B (MAOB) are key players in the inactivation pathway of biogenic amines. Their cellular localization has been well established in the mature brain, but nothing is known concerning the localization of both enzymes during development. We have combined in situ hybridization and histochemistry to localize MAOA and MAOB in the developing nervous system of mice. Our observations can be summarized as five key features. (1) MAOA is tightly linked to catecholaminergic traits. MAOA is expressed in all noradrenergic and adrenergic neurons early on, and in several dopaminergic cell groups such as the substantia nigra. MAOA is also expressed in all the neurons that display a transient tyrosine hydroxylase expression in the brainstem and the amygdala and in neurons with transient dopamine-beta-hydroxylase expression in the cranial sensory ganglia. (2) MAOA and MAOB are coexpressed in the serotoninergic neurons of the raphe from E12 to P7. During postnatal life, MAOA expression declines, whereas MAOB expression remains stable. (3) MAOA is transiently expressed in the cholinergic motor nuclei of the hindbrain, and MAOB is expressed in the forebrain cholinergic neurons. (4) MAOA- and MAOB-expressing neurons are also detected in structures that do not contain aminergic neurons, such as the thalamus, hippocampus, and claustrum. (5) Starting at birth, MAOB expression is found in a variety of nonneuronal cells, the choroid plexus, the ependyma, and astrocytes. These localizations are of importance for understanding the effects of monoaminergic transmission during development.


Assuntos
Diferenciação Celular/fisiologia , Sistema Nervoso Central/enzimologia , Camundongos Endogâmicos C3H/metabolismo , Monoaminoxidase/metabolismo , Neurônios/enzimologia , Sistema Nervoso Periférico/enzimologia , Acetilcolina/metabolismo , Envelhecimento/fisiologia , Animais , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/crescimento & desenvolvimento , Dopamina/metabolismo , Epinefrina/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Histamina/metabolismo , Imuno-Histoquímica , Melatonina/metabolismo , Camundongos , Camundongos Endogâmicos C3H/embriologia , Camundongos Endogâmicos C3H/crescimento & desenvolvimento , Camundongos Knockout , Monoaminoxidase/genética , Neurônios/citologia , Norepinefrina/metabolismo , Sistema Nervoso Periférico/embriologia , Sistema Nervoso Periférico/crescimento & desenvolvimento , Fenótipo , RNA Mensageiro/metabolismo , Serotonina/metabolismo
15.
J Interferon Cytokine Res ; 22(4): 457-62, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12034028

RESUMO

The aim of this study was to investigate the contribution of endogenous - that is, without the addition of any interferon (IFN) inducer - type I IFN production in the defense against tumor development. To this purpose, the IFN-alpha receptor (IFNAR) knockout (KO)-induced mutation, resulting in the complete absence of IFN-alpha/beta activity, was introduced into a C3H genetic background by 10 backcross generations, followed by brother-sister matings for at least four generations. The resulting mice were inoculated either with syngeneic C3H melanoma K1735 cells, with allogeneic 3LL carcinoma cells, or with allogeneic B16F10 melanoma cells. With all three tumor cell lines, tumor development and ensuing mortality were enhanced in the IFNAR KO animals. This indicates that endogenous IFN-alpha/beta production is a mediator of natural immunity to tumor development.


Assuntos
Neoplasias Experimentais/etiologia , Receptores de Interferon/fisiologia , Animais , Carcinoma Pulmonar de Lewis/patologia , Divisão Celular , Feminino , Injeções Intramusculares , Injeções Subcutâneas , Cinética , Masculino , Melanoma Experimental/patologia , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Transplante de Neoplasias , Receptor de Interferon alfa e beta , Receptores de Interferon/genética , Taxa de Sobrevida , Células Tumorais Cultivadas
16.
Int J Neuropsychopharmacol ; 2(3): 241-243, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11281992

RESUMO

A line of transgenic mice was isolated in which transgene integration had caused a deletion in the gene encoding monoamine oxidase A, an enzyme that degrades serotonin and norepinephrine. This has provided an animal model of MAOA deficiency in humans, a condition characterized by borderline mental retardation and impulsive aggression.

17.
Brain Res Dev Brain Res ; 142(1): 19-29, 2003 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-12694941

RESUMO

Genetic inactivation of monoamine oxidase-A (MAO-A) significantly elevates levels of serotonin (5-HT) during early development and causes a disruption in the compartmented organization of thalamocortical axon terminals in layer 4 of the somatosensory cortex. In order to determine whether corticocortical innervation of the primary somatosensory cortex is also affected by this mutation, we examined the distribution of zinc-containing axon terminals (terminals known to originate from within the cortex) in the developing somatosensory cortex of MAO-A knockout mice, at postnatal days (PD) 3, 5, 6, 8, 10, 12, 15, 28, and 60. In layer 4 of wild-type mice, histochemical staining for zinc respected barrel-specific compartments at all ages beyond PD 5. By contrast, zinc staining in MAO-A knockout mice did not exhibit signs of barrel compartmentation at any age. Across cortical layers, substantial developmental changes in the distribution of zinc-containing terminals were observed in wild-type mice up until PD 12, at which time the mature lamina-specific pattern of zinc staining was achieved. Similar changes were observed in the somatosensory cortex of MAO-A knockout mice, except that its developmental time course was significantly compressed, with zincergic innervation achieving a mature appearance by PD 8. These results provide evidence that an excess of monoamines, most likely 5-HT, dramatically perturbs the columnar organization of intracortical zincergic afferents in layer 4 and significantly accelerates the appearance of a mature laminar pattern of zinc-containing corticocortical terminals.


Assuntos
Monoaminoxidase/deficiência , Terminações Pré-Sinápticas/metabolismo , Serotonina/metabolismo , Córtex Somatossensorial/crescimento & desenvolvimento , Zinco/metabolismo , Animais , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Monoaminoxidase/genética , Mutação , Córtex Somatossensorial/fisiologia
18.
Epilepsy Res ; 59(1): 25-34, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15135164

RESUMO

The role of elevated neuroactive amine exposure during embryonic and early postnatal development on seizure threshold and epileptogenesis was examined using both electrical and pentylenetetrazol (PTZ) kindling in monoamine oxidase A knockout (MAO(A) KO) mice and their wildtype, parental strain (C3H). In the first experiment permanent bilateral electrodes were implanted in the amygdala of both C3H and MAO(A) KO mice. The mice had their afterdischarge threshold determined and then seizures were kindled daily for a total of 20 days. We observed that the MAO(A) KO mice had lower afterdischarge thresholds and less severe seizures compared to the C3H mice. In the second experiment, seizures were elicited in experimentally naive mice using 50mg/kg of PTZ once daily for 7 days. We observed that the MAO(A) KO mice had shorter latencies to the onset of the first seizure, shorter total duration of seizures and fewer seizures per day. Overall the results of both experiments suggest that MAO(A) KO mice have an increased susceptibility to seizures, but are more resistant to epileptogenesis. We conclude that the high levels of neuroactive amines in the MAO(A) KO mice reorganize the brain to make the mice more susceptible to seizures but the remaining high levels of serotonin and norepinephrine likely inhibit epileptogenesis.


Assuntos
Excitação Neurológica/genética , Monoaminoxidase/deficiência , Convulsões/genética , Animais , Estimulação Elétrica/métodos , Epilepsia/induzido quimicamente , Epilepsia/genética , Epilepsia/fisiopatologia , Feminino , Excitação Neurológica/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Monoaminoxidase/genética , Pentilenotetrazol , Convulsões/induzido quimicamente , Convulsões/fisiopatologia
19.
Brain Res Bull ; 78(6): 283-9, 2009 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-19111597

RESUMO

Elevated brain monoamine concentrations resulting from monoamine oxidase A genetic ablation (MAOA knock-out mice) lead to changes in other neurotransmitter systems. To investigate the consequences of MAOA deficiency on the cholinergic system, we measured ligand binding to the high-affinity choline transporter (CHT1) and to muscarinic and nicotinic receptors in brain sections of MAOA knock-out (KO) and wild-type mice. A twofold increase in [(3)H]-hemicholinium-3 ([(3)H]-HC-3) binding to CHT1 was observed in the caudate putamen, nucleus accumbens, and motor cortex in MAOA KO mice as compared with wild-type (WT) mice. There was no difference in [(3)H]-HC-3 labeling in the hippocampus (dentate gyrus) between the two genotypes. Binding of [(125)I]-epibatidine ([(125)I]-Epi), [(125)I]-alpha-bungarotoxin ([(125)I]-BGT), [(3)H]-pirenzepine ([(3)H]-PZR), and [(3)H]-AFDX-384 ([(3)H]-AFX), which respectively label high- and low-affinity nicotinic receptors, M1 and M2 muscarinic cholinergic receptors, was not modified in the caudate putamen, nucleus accumbens, and motor cortex. A small but significant decrease of 19% in M1 binding densities was observed in the hippocampus (CA1 field) of KO mice. Next, we tested acetylcholinesterase activity and found that it was decreased by 25% in the striatum of KO mice as compared with WT mice. Our data suggest that genetic deficiency in MAOA enzyme is associated with changes in cholinergic activity, which may account for some of the behavioral alterations observed in mice and humans lacking MAOA.


Assuntos
Encéfalo/metabolismo , Monoaminoxidase/deficiência , Receptores Colinérgicos/metabolismo , Acetilcolinesterase/metabolismo , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/metabolismo , Bungarotoxinas/metabolismo , Hemicolínio 3/metabolismo , Radioisótopos do Iodo/metabolismo , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Monoaminoxidase/genética , Monoaminoxidase/metabolismo , Pirenzepina/análogos & derivados , Pirenzepina/metabolismo , Piridinas/metabolismo , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M2/metabolismo , Receptores Nicotínicos/metabolismo , Trítio/metabolismo
20.
J Biol Chem ; 282(1): 115-23, 2007 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-17090537

RESUMO

Previous studies have established that abrogation of monoamine oxidase (MAO) A expression leads to a neurochemical, morphological, and behavioral specific phenotype with increased levels of serotonin (5-HT), norepinephrine, and dopamine, loss of barrel field structure in mouse somatosensory cortex, and an association with increased aggression in adults. Forebrain-specific MAO A transgenic mice were generated from MAO A knock-out (KO) mice by using the promoter of calcium-dependent kinase IIalpha (CaMKIIalpha). The presence of human MAO A transgene and its expression were verified by PCR of genomic DNA and reverse transcription-PCR of mRNA and Western blot, respectively. Significant MAO A catalytic activity, autoradiographic labeling of 5-HT, and immunocytochemistry of MAO A were found in the frontal cortex, striatum, and hippocampus but not in the cerebellum of the forebrain transgenic mice. Also, compared with MAO A KO mice, lower levels of 5-HT, norepinephrine, and DA and higher levels of MAO A metabolite 5-hydroxyindoleacetic acid were found in the forebrain regions but not in the cerebellum of the transgenic mice. These results suggest that MAO A is specifically expressed in the forebrain regions of transgenic mice. This forebrain-specific differential expression resulted in abrogation of the aggressive phenotype. Furthermore, the disorganization of the somatosensory cortex barrel field structure associated with MAO A KO mice was restored and became morphologically similar to wild type. Thus, the lack of MAO A in the forebrain of MAO A KO mice may underlie their phenotypes.


Assuntos
Monoaminoxidase/genética , Monoaminoxidase/fisiologia , Neurotransmissores/metabolismo , Prosencéfalo/metabolismo , Animais , Encéfalo/metabolismo , Catálise , Genótipo , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Monoaminoxidase/metabolismo , Fenótipo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA