Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Cogn Neurosci ; 57: 101141, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35933923

RESUMO

The rodent posterodorsal medial amygdala (MePD) evaluates and assigns valence to social sensory stimuli. The perception of social stimuli evolves during puberty, when the focus of social interactions shifts from kin to peers. Using the cell birthdate marker bromo-deoxyuridine (BrdU), we previously discovered that more pubertally born cells are added to the rat MePD in males than females. Here we addressed several questions that remained unanswered by our previous work. First, to determine whether there are sex differences in cell proliferation within the MePD, we examined BrdU-immunoreactive (-ir) cells at 2 and 4 h following BrdU administration on postnatal day 30 (P30). The density of BrdU-ir cells was greater in males than in females, indicating greater proliferation in males. Proliferation was substantiated by double-label immunohistochemistry showing that MePD BrdU-ir cells colocalize proliferating cell nuclear antigen, but not the cell death marker Caspase3. We next studied longer time points (2-21 days) following BrdU administration on P30 and found that the rate of cell attrition is higher in males. Finally, triple-label immunohistochemistry of P30-born MePD cells revealed that some of these cells differentiate into neurons or astrocytes within three weeks of cell birth, with no discernable sex differences. The demonstration of pubertal neuro- and glio-genesis in the MePD of male and female rats adds a new dimension to developmental plasticity of the MePD that may contribute to pubertal changes in the perception of social stimuli in both sexes.

2.
eNeuro ; 4(5)2017.
Artigo em Inglês | MEDLINE | ID: mdl-29098175

RESUMO

New cells are added during both puberty and adulthood to hypothalamic regions that govern reproduction, homeostasis, and social behaviors, yet the functions of these late-born cells remain elusive. Here, we pharmacologically inhibited cell proliferation in ventricular zones during puberty or in adulthood and determined subsequent effects on the hormone-induced surge of luteinizing hormone (LH) in female rats. Initial neuroanatomical analyses focused on verifying incorporation, activation, and pharmacological inhibition of pubertally or adult born cells in the anteroventral periventricular nucleus (AVPV) of the hypothalamus because of the essential role of the AVPV in triggering the preovulatory LH surge in females. We first showed that approximately half of the pubertally born AVPV cells are activated by estradiol plus progesterone (P) treatment, as demonstrated by Fos expression, and that approximately 10% of pubertally born AVPV cells express estrogen receptor alpha (ERα). Next, we found that mitotic inhibition through intracerebroventricular (ICV) administration of cytosine ß-D-arabinofuranoside (AraC), whether during puberty or in adulthood, decreased the number of new cells added to the AVPV and the suprachiasmatic nucleus (SCN), and also blunted and delayed the hormone-induced LH surge. These studies do not prove, but are highly suggestive, that ongoing postnatal addition of new cells in periventricular brain regions, including the AVPV and SCN, may be important to the integrity of female reproduction.


Assuntos
Hipotálamo Anterior/citologia , Hipotálamo Anterior/metabolismo , Hormônio Luteinizante/metabolismo , Maturidade Sexual/fisiologia , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/metabolismo , Animais , Antimitóticos/farmacologia , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Estradiol/administração & dosagem , Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Hipotálamo Anterior/efeitos dos fármacos , Hipotálamo Anterior/crescimento & desenvolvimento , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Progesterona/administração & dosagem , Progesterona/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Sprague-Dawley , Receptores de Progesterona/metabolismo , Núcleo Supraquiasmático/efeitos dos fármacos , Núcleo Supraquiasmático/crescimento & desenvolvimento
3.
Endocrinology ; 157(6): 2393-402, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27145006

RESUMO

The anteroventral periventricular nucleus (AVPV) orchestrates the neuroendocrine-positive feedback response that triggers ovulation in female rodents. The AVPV is larger and more cell-dense in females than in males, and during puberty, only females develop the capacity to show a positive feedback response. We previously reported a potential new mechanism to explain this female-specific gain of function during puberty, namely a female-biased sex difference in the pubertal addition of new cells to the rat AVPV. Here we first asked whether this sex difference is due to greater cell proliferation and/or survival in females. Female and male rats received the cell birthdate marker 5-bromo-2'-deoxyuridine (BrdU; 200 mg/kg, ip) on postnatal day (P) 30; brains were collected at short and long intervals after BrdU administration to assess cell proliferation and survival, respectively. Overall, females had more BrdU-immunoreactive cells in the AVPV than did males, with no sex differences in the rate of cell attrition over time. Thus, the sex difference in pubertal addition of AVPV cells appears to be due to greater cell proliferation in females. Next, to determine the phenotype of pubertally born AVPV cells, daily BrdU injections were given to female rats on P28-56, and tissue was collected on P77 to assess colocalization of BrdU and markers for mature neurons or glia. Of the pubertally born AVPV cells, approximately 15% differentiated into neurons, approximately 19% into astrocytes, and approximately 23% into microglia. Thus, both neuro- and gliogenesis occur in the pubertal female rat AVPV and potentially contribute to maturation of female reproductive function.


Assuntos
Hipotálamo Anterior/citologia , Hipotálamo Anterior/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Feminino , Masculino , Microglia/citologia , Microglia/metabolismo , Puberdade/fisiologia , Ratos , Ratos Sprague-Dawley , Maturidade Sexual/fisiologia
4.
Am J Physiol Heart Circ Physiol ; 309(2): H335-44, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25980022

RESUMO

The role of serotonin in the hemodynamic response to blood loss remains controversial. Caudal raphe serotonin neurons are activated during hypotensive hemorrhage, and their destruction attenuates sympathetic increases following blood loss in unanesthetized rats. Caudal raphe neurons provide serotonin-positive projections to the nucleus tractus solitarii (NTS), and disruption of serotonin-positive nerve terminals in the NTS attenuates sympathetic recovery following hemorrhage. Administration of 5-HT1A-receptor agonists following hemorrhage augments sympathetic-mediated increases in venous tone and tissue hypoxia. These findings led us to hypothesize that severe blood loss promotes activation of 5-HT1A receptors in the NTS, which facilitates sympathetic recovery and peripheral tissue perfusion. Here, we developed an adeno-associated viral vector encoding an efficacious small hairpin RNA sequence targeting the rat 5-HT1A receptor. Unanesthetized rats subjected to NTS injection of the anti-rat 5-HT1A small hairpin RNA-encoding vector 4 wk prior showed normal blood pressure recovery, but an attenuated recovery of renal sympathetic nerve activity (-6.4 ± 12.9 vs. 42.6 ± 15.6% baseline, P < 0.05) 50 min after 21% estimated blood volume withdrawal. The same rats developed increased tissue hypoxia after hemorrhage, as indicated by prolonged elevations in lactate (2.77 ± 0.5 vs. 1.34 ± 0.2 mmol/l, 60 min after start of hemorrhage, P < 0.05). 5-HT1A mRNA levels in the commissural NTS were directly correlated with renal sympathetic nerve activity (P < 0.01) and inversely correlated with lactate (P < 0.05) 60 min after start of hemorrhage. The data suggest that 5-HT1A receptors in the commissural NTS facilitate tissue perfusion after blood loss likely by increasing sympathetic-mediated venous return.


Assuntos
Barorreflexo , Hemorragia/metabolismo , Hipotensão/metabolismo , Rim/inervação , Receptor 5-HT1A de Serotonina/metabolismo , Neurônios Serotoninérgicos/metabolismo , Núcleo Solitário/metabolismo , Sistema Nervoso Simpático/metabolismo , Animais , Barorreflexo/efeitos dos fármacos , Pressão Sanguínea , Volume Sanguíneo , Modelos Animais de Doenças , Hemorragia/genética , Hemorragia/fisiopatologia , Hipotensão/genética , Hipotensão/fisiopatologia , Ácido Láctico/metabolismo , Masculino , Interferência de RNA , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Receptor 5-HT1A de Serotonina/efeitos dos fármacos , Receptor 5-HT1A de Serotonina/genética , Recuperação de Função Fisiológica , Neurônios Serotoninérgicos/efeitos dos fármacos , Agonistas do Receptor 5-HT1 de Serotonina/farmacologia , Transdução de Sinais , Núcleo Solitário/efeitos dos fármacos , Núcleo Solitário/fisiopatologia , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/fisiopatologia , Fatores de Tempo
5.
Dev Neurobiol ; 74(6): 633-42, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24339170

RESUMO

Adolescence involves shifts in social behaviors, behavioral flexibility, and adaptive risk-taking that coincide with structural remodeling of the brain. We previously showed that new cells are added to brain regions associated with sexual behaviors, suggesting that cytogenesis may be a mechanism for acquiring adult-typical behaviors during adolescence. Whether pubertal cell addition occurs in brain regions associated with behavioral flexibility or motivation and whether these patterns differ between pubertal and adult animals had not been determined. Therefore, we assessed patterns of cell proliferation or survival in the prefrontal cortex and nucleus accumbens. Pubertal and adult male rats were given injections of bromo-deoxyuridine (BrdU). To assess cell proliferation, half of the animals from each group were sacrificed 24 h following the last injection. The remaining animals were sacrificed at Day 30 following the last injection to evaluate cell survival. Adult animals had significantly lower densities of BrdU-immunoreactive (ir) cells in the prefrontal cortex, irrespective of post-BrdU survival time, whereas in the nucleus accumbens, adult animals had a lower density of BrdU-ir cells at the short survival time; however, the density of BrdU-ir cells was equivalent in pubertal and adult animals at the longer survival time. These data provide evidence that cell addition during puberty may contribute to the remodeling of brain regions associated with behavioral flexibility and motivation, and this cell addition continues into adulthood, albeit at lower levels. Higher levels of cell proliferation or survival in younger animals may reflect a higher level of plasticity, possibly contributing to the dynamic remodeling of the pubertal brain.


Assuntos
Envelhecimento , Neurogênese/fisiologia , Neurônios/fisiologia , Núcleo Accumbens/citologia , Núcleo Accumbens/crescimento & desenvolvimento , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/crescimento & desenvolvimento , Análise de Variância , Animais , Animais Recém-Nascidos , Bromodesoxiuridina/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
6.
Horm Behav ; 64(2): 203-10, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23998664

RESUMO

This article is part of a Special Issue "Puberty and Adolescence". Sexual differentiation is the process by which the nervous system becomes structurally and functionally dissimilar in females and males. In mammals, this process has been thought to occur during prenatal and early postnatal development, when a transient increase in testosterone secretion masculinizes and defeminizes the developing male nervous system. Decades of research have led to the views that structural sexual dimorphisms created during perinatal development are passively maintained throughout life, and that ovarian hormones do not play an active role in feminization of the nervous system. Furthermore, perinatal testosterone was thought to determine sex differences in neuron number by regulating cell death and cell survival, and not by regulating cell proliferation. As investigations of neural development during adolescence became more prominent in the late 20th century and revealed the extent of brain remodeling during this time, each of these tenets has been challenged and modified. Here we review evidence from the animal literature that 1) the brain is further sexually differentiated during puberty and adolescence; 2) ovarian hormones play an active role in the feminization of the brain during puberty; and 3) hormonally modulated, sex-specific addition of new neurons and glial cells, as well as loss of neurons, contribute to sexual differentiation of hypothalamic, limbic, and cortical regions during adolescence. This architectural remodeling during the adolescent phase of sexual differentiation of the brain may underlie the known sex differences in vulnerability to addiction and psychiatric disorders that emerge during this developmental period.


Assuntos
Encéfalo/crescimento & desenvolvimento , Hormônios/fisiologia , Roedores/fisiologia , Diferenciação Sexual/fisiologia , Maturidade Sexual/fisiologia , Animais , Feminino , Humanos , Hipotálamo/crescimento & desenvolvimento , Masculino
7.
J Comp Neurol ; 520(11): 2531-44, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22581688

RESUMO

The posterodorsal medial amygdala (MePD) exhibits numerous sex differences including differences in volume and in the number and morphology of neurons and astroctyes. In adulthood, gonadal hormones, including both androgens and estrogens, have been shown to play a role in maintaining the masculine character of many of these sex differences, but whether adult gonadal hormones maintain the increased number and complexity of astrocytes in the male MePD was unknown. To answer this question we examined astrocytes in the MePD of male and female Long Evans rats that were gonadectomized as adults and treated for 30 days with either testosterone or a control treatment. At the end of treatment brains were collected and immunostained for glial fibrillary acidic protein. Stereological analysis revealed that adult androgen levels influenced the number and complexity of astrocytes in the MePD of both sexes, but the specific effects of androgens were different in males and females. However, sex differences in the number and complexity of adult astrocytes persisted even in the absence of gonadal hormones in adulthood, suggesting that androgens also act earlier in life to determine these adult sex differences. Using immunofluorescence and confocal microscopy, we found robust androgen receptor immunostaining in a subpopulation of MePD astrocytes, suggesting that testosterone may act directly on MePD astrocytes to influence their structure and function.


Assuntos
Tonsila do Cerebelo/fisiologia , Astrócitos/fisiologia , Testosterona/fisiologia , Tonsila do Cerebelo/citologia , Animais , Contagem de Células , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Distribuição Aleatória , Ratos , Ratos Long-Evans , Caracteres Sexuais
8.
Psychoneuroendocrinology ; 35(7): 1023-33, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20138435

RESUMO

Estradiol regulates serotonin 1A (5-HT(1A)) receptor signaling. Since desensitization of 5-HT(1A) receptors may be an underlying mechanism by which selective serotonin reuptake inhibitors (SSRIs) mediate their therapeutic effects and combining estradiol with SSRIs enhances the efficacy of the SSRIs, it is important to determine which estrogen receptors are capable of desensitizating 5-HT(1A) receptor function. We previously demonstrated that selective activation of the estrogen receptor, GPR30, desensitizes 5-HT(1A) receptor signaling in rat hypothalamic paraventricular nucleus (PVN). However, since estrogen receptor-beta (ERbeta), is highly expressed in the PVN, we investigated the role of ERbeta in estradiol-induced desensitization of 5-HT(1A) receptor signaling. We first showed that a selective ERbeta agonist, diarylpropionitrile (DPN) has a 100-fold lower binding affinity than estradiol for GPR30. Administration of DPN did not desensitize 5-HT(1A) receptor signaling in rat PVN as demonstrated by agonist-stimulated hormone release. Second, we used a recombinant adenovirus containing ERbeta siRNAs to decrease ERbeta expression in the PVN. Reductions in ERbeta did not alter the estradiol-induced desensitization of 5-HT(1A) receptor signaling in oxytocin cells. In contrast, in animals with reduced ERbeta, estradiol administration, instead of producing desensitization, augmented the ACTH response to a 5-HT(1A) agonist. Combined with the results from the DPN treatment experiments, desensitization of 5-HT(1A) receptor signaling does not appear to be mediated by ERbeta in oxytocin cells, but that ERbeta, together with GPR30, may play a complex role in central regulation of 5-HT(1A)-mediated ACTH release. Determining the mechanisms by which estrogens induce desensitization may aid in the development of better treatments for mood disorders.


Assuntos
Estradiol/metabolismo , Receptor beta de Estrogênio/efeitos dos fármacos , Receptor beta de Estrogênio/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Hormônio Adrenocorticotrópico/sangue , Hormônio Adrenocorticotrópico/metabolismo , Animais , Estradiol/farmacologia , Feminino , Nitrilas/farmacologia , Ocitocina/sangue , Ocitocina/metabolismo , Propionatos/farmacologia , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo
9.
Horm Mol Biol Clin Investig ; 1(1): 21-6, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25961968

RESUMO

The nervous system is a steroidogenic tissue and several steroids synthesized locally in the brain, such as pregnenolone, progesterone and estradiol, modulate neuronal and glial physiology and are neuroprotective. The brain upregulates steroidogenesis at sites of injury as part of a program triggered by neural tissue to cope with neurodegenerative insults. Pharmacological targets to increase brain steroidogenesis and promote neuroprotection include the molecules that transport cholesterol to the inner mitochondrial membrane, where the first enzyme for steroidogenesis is located. Furthermore, the human gene encoding aromatase, the enzyme that synthesizes estradiol, is under the control of different tissue-specific promoters, and it is therefore conceivable that selective aromatase modulators can be developed that will enhance the expression of the enzyme and the consequent increase in estrogen formation in the brain but not in other tissues.

10.
Psychoneuroendocrinology ; 34 Suppl 1: S113-22, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19447561

RESUMO

Decreasing levels of sex hormones with aging may have a negative impact on brain function, since this decrease is associated with the progression of neurodegenerative disorders, increased depressive symptoms and other psychological disturbances. Extensive evidence from animal studies indicates that sex steroids, in particular estradiol, are neuroprotective. However, the potential benefits of estradiol therapy for the brain are counterbalanced by negative, life-threatening risks in the periphery. A potential therapeutic alternative to promote neuroprotection is the use of selective estrogen receptor modulators (SERMs), which may be designed to act with tissue selectivity as estrogen receptor agonists in the brain and not in other organs. Currently available SERMs act not only with tissue selectivity, but also with cellular selectivity within the brain and differentially modulate the activation of microglia, astroglia and neurons. Finally, SERMs may promote the interaction of estrogen receptors with the neuroprotective signaling of growth factors, such as the phosphatidylinositol 3-kinase/glycogen synthase kinase 3 pathway.


Assuntos
Fármacos Neuroprotetores/farmacologia , Receptores de Estrogênio/agonistas , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Doenças do Sistema Nervoso Central/tratamento farmacológico , Humanos , Modelos Biológicos , Neuroglia/metabolismo , Neurônios/metabolismo , Receptores de Estrogênio/metabolismo , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico
11.
Horm Behav ; 53(5): 753-64, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18402960

RESUMO

Androgen receptors are expressed in many different neuronal populations in the central nervous system where they often act as transcription factors in the cell nucleus. However, recent studies have detected androgen receptor immunoreactivity in neuronal and glial processes of the adult rat neocortex, hippocampal formation, and amygdala as well as in the telencephalon of eastern fence and green anole lizards. This review discusses previously published findings on extranuclear androgen receptors, as well as new experimental results that begin to establish a possible functional role for androgen receptors in axons within cortical regions. Electron microscopic studies have revealed that androgen receptor immunoreactive processes in the rat brain correspond to axons, dendrites and glial processes. New results show that lesions of the dorsal CA1 region by local administration of ibotenic acid reduce the density of androgen receptor immunoreactive axons in the cerebral cortex and the amygdala, suggesting that these axons may originate in the hippocampus. Androgen receptor immunoreactivity in axons is also decreased by the intracerebroventricular administration of colchicine, suggesting that androgen receptor protein is transported from the perikaryon to the axons by fast axonal transport. Androgen receptors in axons located in the cerebral cortex and amygdala and originating in the hippocampus may play an important role in the rapid behavioral effects of androgens.


Assuntos
Encéfalo/fisiologia , Receptores Androgênicos/fisiologia , Animais , Axônios/fisiologia , Axônios/ultraestrutura , Encéfalo/citologia , Encéfalo/ultraestrutura , Humanos , Neurônios/metabolismo , Neurônios/ultraestrutura , Receptores Androgênicos/ultraestrutura
12.
Reproduction ; 135(4): 419-29, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18367504

RESUMO

Neuron-to-glia, glia-to-neuron, and glia-to-glia communication are implicated in the modulation of neuronal activity and synaptic transmission relevant to reproduction. Glial cells play an important role in neuroendocrine regulation and participate in the sexual differentiation of neuronal connectivity of brain regions involved in the control of reproductive neuroendocrine output. During puberty, modifications in the morphology and chemistry of astrocytes and tanycytes in the hypothalamus and median eminence influence the maturation of the neuronal circuits controlling the secretion of GnRH. During adult reproductive life, the glial cells participate in the transient remodeling of neuronal connectivity in the preoptic area, the arcuate nucleus, the median eminence, and other brain regions involved in the control of reproduction. Gonadal hormones regulate glial plasticity by direct and indirect effects and regulate various other endocrine signals, local soluble factors and adhesion molecules that also affect glial function and glia-to-neuron communication. The glial cells, therefore, are central to the coordination of endocrine and local inputs that bring about neural plasticity and adapt reproductive capacity to homeostatic signals.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Hipotálamo , Neuroglia/fisiologia , Neurônios/fisiologia , Sistemas Neurossecretores/fisiologia , Reprodução/fisiologia , Adulto , Comunicação Celular , Humanos , Plasticidade Neuronal
13.
Nat Neurosci ; 11(9): 995-7, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19160494

RESUMO

New cells, including neurons, arise in several brain regions during puberty in rats. Sex differences in pubertal addition of cells coincide with adult sexual dimorphisms: for each region, the sex that gains more cells during puberty has a larger volume in adulthood. Removing gonadal hormones before puberty eliminates these sex differences, indicating that gonadal steroids direct the addition of new cells during puberty to maintain and accentuate sexual dimorphisms in the adult brain.


Assuntos
Envelhecimento/fisiologia , Neurogênese/fisiologia , Caracteres Sexuais , Fatores Etários , Animais , Animais Recém-Nascidos , Encéfalo/anatomia & histologia , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Bromodesoxiuridina/metabolismo , Castração , Contagem de Células , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Fosfopiruvato Hidratase/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
14.
Neurourol Urodyn ; 27(5): 440-5, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17957780

RESUMO

AIM: Urinary incontinence affects a significant number of post-menopausal women. There is conflicting evidence whether voiding symptoms in these women are related to hypoestrogenism or aging itself. This neuroanatomical study was designed to determine whether a specific central nervous system (CNS) pathway that projects to the pontine micturition center (PMC, also known as "Barrington's nucleus") is estrogen sensitive in a rat model. METHODS: A fluorescent retrograde tracer was injected into the dorsal pontine tegmentum of adult female Sprague-Dawley rats to identify neurons in the medial preoptic area (MPA) that project to the PMC. Immunohistochemistry was performed using antibodies directed against estrogen receptor-alpha (ERalpha) and estrogen receptor-beta (ERbeta) to identify estrogen-sensitive neurons. The brain sections were examined using fluorescence microscopy to identify cells that project to the PMC (contain fluorescent tracer) and also express ER (are immunoreactive for ER). RESULTS: There are neurons in the MPA that are double labeled (contain fluorescent tracer and express ERalpha, but not ERbeta), showing that a subset of neurons projecting from the MPA to the PMC is estrogen sensitive. CONCLUSIONS: A subset of estrogen-sensitive neurons projects from the MPA to the PMC in rats, raising the possibility that indirect estrogenic regulation of forebrain neuronal function may modulate the micturition reflex. Future development of drugs that alter the function of this estrogen-sensitive CNS pathway may provide therapeutic strategies to treat post-menopausal incontinence.


Assuntos
Estrogênios/fisiologia , Locus Cerúleo/fisiologia , Vias Neurais/fisiologia , Ponte/fisiologia , Área Pré-Óptica/fisiologia , Animais , Interpretação Estatística de Dados , Receptor alfa de Estrogênio/efeitos dos fármacos , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/efeitos dos fármacos , Receptor beta de Estrogênio/metabolismo , Feminino , Corantes Fluorescentes , Microscopia de Fluorescência , Neurônios/metabolismo , Ovariectomia , Ratos , Ratos Sprague-Dawley , Estilbamidinas
15.
Expert Rev Endocrinol Metab ; 2(3): 387-397, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-30743812

RESUMO

Although estradiol is a neuroprotective factor, estrogen therapy in older women increases the risk of adverse cognitive outcomes and poses additional peripheral risks, requiring careful use of estrogenic compounds as treatments for neurodegenerative conditions or neural injury. Potential alternatives to estrogen therapy to promote neuroprotection might include treatment with molecules that are able to interact with estrogen receptors, with alternative mechanisms of action, or with molecules that induce local estradiol synthesis in the brain, or a combination of all. However, before considering the broad clinical applications, more basic research is required to clarify the mechanisms of action and potential risks of some of these estrogen-based treatments.

16.
J Comp Neurol ; 492(4): 456-68, 2005 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-16228996

RESUMO

Androgen exposure during development and adulthood promotes cell-to-cell communication, modulates the size of specific brain nuclei, and influences hormone-dependent behavioral and neuroendocrine functions. Androgen action involves the activation of androgen receptors (AR). To elucidate the mechanisms involved in AR-mediated effects on forebrain development, double-label fluorescent immunohistochemistry and confocal microscopy were employed to identify the cellular phenotype of AR-immunoreactive (AR(+)) cells in the developing (embryonic day 20, postnatal days 0, 4, 10) and adult male rat forebrain. Sections were doubly labeled with antibodies directed against AR and one of the following: neurons (immature, nestin; mature, NeuN) or astrocytes [immature, vimentin; mature, glial fibrillary acidic protein (GFAP)] or mature oligodendrocytes (mGalC). In all brain regions examined, by far the majority of AR(+) cells were neurons. In addition, small subsets of AR(+) cells were identified as mature astrocytes (GFAP(+)) but only in specific brain regions at specific ages. AR(+)/GFAP(+) cells were observed in the cerebral cortex but only in postnatal day 10 rats and in the arcuate nucleus of the hypothalamus but only in adult rats. Immature neurons, immature astrocytes, and oligodendrocytes were not AR(+) at any age, in any region. Thus, both neurons and astrocytes in the male rat forebrain contain ARs, suggesting that androgens, via ARs, may exert effects on both cell types in an age- and region-dependent manner.


Assuntos
Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Receptores Androgênicos/metabolismo , Androgênios/farmacologia , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Biomarcadores/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Feminino , Imuno-Histoquímica , Masculino , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fenótipo , Ratos , Ratos Sprague-Dawley
17.
Endocrinology ; 146(10): 4340-8, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16002522

RESUMO

Reproductive and behavioral functions of progesterone receptors (PRs) in males were assessed by examining consequences of PR gene deletion. Basal hormone levels were measured in male progesterone receptor knockout (PRKO) mice and compared to wild-type (WT) counterparts. RIA of serum LH, testosterone, and progesterone levels revealed no significant differences. Levels of FSH were moderately but significantly lower and inhibin levels were higher in PRKOs; these differences were not accompanied by gross differences in testicular weight or morphology. PRKOs exhibited significant alterations in sexual behavior. In initial tests PRKOs exhibited reduced latency to mount, compared with WT. In second sessions, PRKOs again showed a significantly reduced latency to mount and increased likelihood of achieving ejaculation. RU486 treatment in WT produced increased mount and intromission frequency and decreased latency to intromission. In anxiety-related behavior tests, PRKO mice exhibited intermediate anxiety levels, compared with WT, suggesting that enhanced sexual behavior in PRKOs is not secondary to reduced anxiety. Immunohistochemical analysis revealed significantly enhanced androgen receptor expression in the medial preoptic nucleus and bed nucleus of the stria terminalis of PRKO. We conclude that testicular development and function and homeostatic regulation of the hypothalamic-pituitary testicular axis are altered to a lesser extent by PR gene deletion. In contrast, PR appears to play a substantial role in inhibiting the anticipatory/motivational components of male sexual behavior in the mouse. The biological significance of this inhibitory mechanism and the extent to which it is mediated by reduced androgen receptor expression remain to be clarified.


Assuntos
Receptores Androgênicos/fisiologia , Receptores de Progesterona/deficiência , Receptores de Progesterona/genética , Comportamento Sexual Animal , Animais , Ansiedade , Primers do DNA , Hormônio Foliculoestimulante/sangue , Hormônio Luteinizante/sangue , Masculino , Camundongos , Camundongos Knockout , Mifepristona/farmacologia , Atividade Motora , Tamanho do Órgão , Reação em Cadeia da Polimerase , Progesterona/farmacologia , Radioimunoensaio , Contagem de Espermatozoides , Testículo/anatomia & histologia
18.
J Neurosci ; 25(16): 4004-13, 2005 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-15843602

RESUMO

In the hamster facial nerve injury paradigm, we have established that androgens enhance both functional recovery from facial nerve paralysis and the rate of regeneration in the adult, through intrinsic effects on the nerve cell body response to injury and via an androgen receptor (AR)-mediated mechanism. Whether these therapeutic effects of gonadal steroids encompass neuroprotection from axotomy-induced cell death is the focus of the present study. Virtually 100% of adult hamster facial motoneurons (FMNs) survive axotomy at the stylomastoid foramen (SMF), whereas, before postnatal day 15 (P15), developing FMNs undergo substantial axotomy-induced cell death. The first part of the present study focuses on determining when ARs are first expressed in developing hamster FMNs. Using AR immunocytochemistry, it was found that males express ARs by P2 and females by P4, which is the earliest demonstration of AR expression in mammalian motoneurons reported thus far in the literature. The second half examines the neuroprotective effects of testosterone propionate, 17-beta estradiol, and dihydrotestosterone on FMNs of P7 hamsters after facial nerve transection at the SMF. The results demonstrate that androgens and estrogens are equally able to rescue approximately 20% of FMNs from axotomy-induced cell death, with the effects permanent. This study is the first to investigate the effects of both androgens and estrogens on axotomy-induced cell death in one system and, with our previously published work, to validate the hamster FMN injury paradigm as a model of choice in the investigation of both neurotherapeutic and neuroprotective actions of gonadal steroids.


Assuntos
Traumatismos do Nervo Facial/tratamento farmacológico , Traumatismos do Nervo Facial/patologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hormônios Esteroides Gonadais/farmacologia , Neurônios Motores/efeitos dos fármacos , Fatores Etários , Animais , Animais Recém-Nascidos , Axotomia/métodos , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/crescimento & desenvolvimento , Tronco Encefálico/patologia , Contagem de Células/métodos , Morte Celular/efeitos dos fármacos , Cricetinae , Di-Hidrotestosterona/farmacologia , Di-Hidrotestosterona/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Estradiol/farmacologia , Estradiol/uso terapêutico , Feminino , Lateralidade Funcional , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hormônios Esteroides Gonadais/uso terapêutico , Imuno-Histoquímica/métodos , Masculino , Mesocricetus , Neurônios Motores/patologia , Receptores Androgênicos/imunologia , Receptores Androgênicos/metabolismo , Fatores Sexuais , Testosterona/farmacologia , Testosterona/uso terapêutico
19.
Brain Res Brain Res Rev ; 48(2): 273-86, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15850667

RESUMO

Proteins involved in the intramitochondrial trafficking of cholesterol, the first step in steroidogenesis, such as the steroidogenic acute regulatory protein (StAR) and the peripheral-type benzodiazepine receptor (PBR), are upregulated in the nervous system after injury. Accordingly, a local increase in the levels of steroids, such as pregnenolone and progesterone, is observed following traumatic injury in the brain and spinal cord. The expression and activity of aromatase, the enzyme that synthesizes estradiol, is also increased in injured brain areas and its inhibition results in an increased neurodegeneration. These findings suggest that an increase in steroidogenesis is part of an overall mechanism used by the nervous tissue to cope with neurodegenerative conditions. Neural steroidogenesis is the result of a coordinated interaction of neurons and glia. For example, after neural injury, there is an upregulation of StAR in neurons and of PBR in microglia and astroglia. Aromatase is expressed in neurons under basal conditions and is upregulated in reactive astrocytes after injury. Some of the steroids produced by glia are neuroprotective. Progesterone and progesterone derivatives produced by Schwann cells, promote myelin formation and the remyelination and regeneration of injured nerves. In the central nervous system, the steroids produced by glia regulate synaptic function, affect anxiety, cognition, sleep and behavior, and exert neuroprotective and reparative roles. In addition, glial cells are targets for steroids and mediate some of the effects of these molecules on neurons, including the regulation of survival and regeneration.


Assuntos
Lesões Encefálicas/prevenção & controle , Hormônios Esteroides Gonadais/uso terapêutico , Neuroglia/fisiologia , Neurônios/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Animais , Aromatase/metabolismo , Lesões Encefálicas/complicações , Lesões Encefálicas/metabolismo , Comunicação Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Humanos , Modelos Biológicos , Neuroglia/classificação
20.
Exp Gerontol ; 39(11-12): 1623-31, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15582278

RESUMO

Sex steroids exert pleiotropic effects in the nervous system, preserving neural function and promoting neuronal survival. Therefore, the age-related decrease in sex steroids may have a negative impact on neural function. Progesterone, testosterone and estradiol prevent neuronal loss in the central nervous system in different experimental animal models of neurodegeneration. Furthermore, progesterone and its reduced derivatives dihydroprogesterone and tetrahydroprogesterone reduce aging-associated morphological abnormalities of myelin and aging-associated myelin fiber loss in rat peripheral nerves. However, the results from hormone replacement studies in humans are thus far inconclusive. A possible alternative to hormonal replacement therapy is to increase local steroidogenesis by neural tissues, which express enzymes for steroid synthesis and metabolism. Proteins involved in the intramitochondrial trafficking of cholesterol, the first step in steroidogenesis, such as the peripheral-type benzodiazepine receptor and the steroidogenic acute regulatory protein, are up-regulated in the nervous system after injury. Furthermore, steroidogenic acute regulatory protein expression is increased in the brain of 24-month-old rats compared with young adult rats. This suggests that brain steroidogenesis may be modified in adaptation to neurodegenerative conditions and to the brain aging process. Furthermore, recent studies have shown that local formation of estradiol in the brain, by the enzyme aromatase, is neuroprotective. Therefore, steroidogenic acute regulatory protein, peripheral-type benzodiazepine receptor and aromatase are attractive pharmacological targets to promote neuroprotection in the aged brain.


Assuntos
Envelhecimento/fisiologia , Encéfalo/fisiologia , Hormônios Esteroides Gonadais/fisiologia , Idoso , Animais , Aromatase/metabolismo , Encéfalo/metabolismo , Feminino , Terapia de Reposição Hormonal , Humanos , Masculino , Modelos Animais , Doenças Neurodegenerativas/metabolismo , Fosfoproteínas/metabolismo , Progesterona/metabolismo , Ratos , Receptores de GABA-A/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...