Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Brain Behav Immun ; 120: 54-70, 2024 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-38772427

RESUMEN

Female sex and Apolipoprotein E (APOE) ε4 genotype are top non-modifiable risk factors for Alzheimer's disease (AD). Although female-unique experiences like parity (pregnancy and motherhood) have positive effects on neuroplasticity at middle age, previous pregnancy may also contribute to AD risk. To explore these seemingly paradoxical long-term effects of parity, we investigated the impact of parity with APOEε4 genotype by examining behavioural and neural biomarkers of brain health in middle-aged female rats. Our findings show that primiparous (parous one time) hAPOEε4 rats display increased use of a non-spatial cognitive strategy and exhibit decreased number and recruitment of new-born neurons in the ventral dentate gyrus of the hippocampus in response to spatial working memory retrieval. Furthermore, primiparity and hAPOEε4 genotype synergistically modulate inflammatory markers in the ventral hippocampus. Collectively, these findings demonstrate that previous parity in hAPOEε4 rats confers an added risk to present with reduced activity and engagement of the hippocampus as well as elevated pro-inflammatory signaling, and underscore the importance of considering female-specific factors and genotype in health research.

2.
Neurobiol Aging ; 109: 192-194, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34775209

RESUMEN

Sex differences are well-established in Alzheimer's disease (AD) frequency and pathogenesis, but are not mechanistically understood. Accelerated epigenetic age has been associated with both cognitive aging and AD pathophysiology, but has not been studied by sex in AD or related cognitive impairment. Using the ADNI cohort, we found that none of sex, cognitive impairment diagnosis, nor load of APOEε4 alleles (strongest genetic AD risk factor) were associated with epigenetic age acceleration (DNAmAge, Intrinsic DNAmAge, PhenoAge, or GrimAge), although females exhibit more accelerated epigenetic aging using the Skin & Blood clock in the transition from normal cognition to cognitive impairment than males.


Asunto(s)
Envejecimiento/genética , Enfermedad de Alzheimer/genética , Envejecimiento Cognitivo , Disfunción Cognitiva/genética , Epigénesis Genética/genética , Caracteres Sexuales , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/etiología , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad
3.
Brain Behav Immun ; 97: 394-409, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34174336

RESUMEN

Untreated perinatal depression can have severe consequences for the mother and her children. However, both the efficacy to mothers and safety to exposed infants of pharmacological antidepressants such as selective serotonin reuptake inhibitors (SSRIs), have been questioned. We previously reported that maternal SSRI exposure increased hippocampal IL-1ß levels, which may be tied to limited efficacy of SSRIs during the postpartum to the dam but is not yet known whether maternal postpartum SSRIs affect the neuroinflammatory profile of adult offspring. In addition, although controversial, perinatal SSRI exposure has been linked to increased risk of autism spectrum disorder (ASD) in children. Oxytocin (OT) is under investigation as a treatment for ASD, but OT is a large neuropeptide that has difficulty crossing the blood-brain barrier (BBB). TriozanTM is a nanoformulation that can facilitate OT to cross the BBB. Thus, we investigated the impact of maternal postpartum SSRIs and offspring preadolescent OT treatment on adult offspring neuroinflammation, social behavior, and neurogenesis in the hippocampus. Using a model of de novo postpartum depression, corticosterone (CORT) was given in the postpartum to the dam with or without treatment with the SSRI, fluoxetine (FLX) for 21 days postpartum. Offspring were then subsequently treated with either OT, OT + TriozanTM, or vehicle for 10 days prior to adolescence (PD25-34). Maternal FLX decreased hippocampal IL-10 and IL-13 and neurogenesis in both sexes, whereas maternal CORT increased hippocampal IL-13 in both sexes. Maternal CORT treatment shifted the neuroimmune profile towards a more proinflammatory profile in offspring hippocampus, whereas oxytocin, independent of formulation, normalized this profile. OT treatment increased hippocampal neurogenesis in adult males but not in adult females, regardless of maternal treatment. OT treatment increased the time spent with a novel social stimulus animal (social investigation) in both adult male and female offspring, although this effect depended on maternal CORT. These findings underscore that preadolescent exposure to OT can reverse some of the long-lasting effects of postpartum maternal CORT and FLX treatments in the adult offspring. In addition, we found that maternal treatments that reduce (CORT) or increase (FLX) hippocampal inflammation in dams resulted in opposing patterns of hippocampal inflammation in adult offspring.


Asunto(s)
Trastorno del Espectro Autista , Efectos Tardíos de la Exposición Prenatal , Hijos Adultos , Animales , Trastorno del Espectro Autista/tratamiento farmacológico , Proteína Doblecortina , Femenino , Fluoxetina/farmacología , Hipocampo , Humanos , Inflamación/tratamiento farmacológico , Masculino , Neurogénesis , Oxitocina , Embarazo , Ratas , Ratas Sprague-Dawley , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Estrés Psicológico
4.
Psychoneuroendocrinology ; 129: 105248, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33962245

RESUMEN

Alzheimer's disease (AD) is characterized by severe cognitive decline and pathological changes in the brain (brain atrophy, hyperphosphorylation of tau, and deposition of amyloid-beta protein). Females have greater neuropathology (AD biomarkers and brain atrophy rates) and cognitive decline than males, however these effects can depend on diagnosis (amnestic mild cognitive impairment (aMCI) or AD) and APOE genotype (presence of ε4 alleles). Using the ADNI database (N = 630 females, N = 830 males), we analyzed the effect of sex, APOE genotype (non-carriers or carriers of APOEε4 alleles), and diagnosis (cognitively normal (CN), early aMCI (EMCI), late aMCI (LMCI), probable AD) on cognition (memory and executive function), hippocampal volume, and AD biomarkers (CSF levels of amyloid beta, tau, and ptau). Regardless of APOE genotype, memory scores were higher in CN, EMCI, and LMCI females compared to males but this sex difference was absent in probable AD, which may suggest a delay in the onset of cognitive decline or diagnosis and/or a faster trajectory of cognitive decline in females. We found that, regardless of diagnosis, CSF tau-pathology was disproportionately elevated in female carriers of APOEε4 alleles compared to males. In contrast, male carriers of APOEε4 alleles had reduced levels of CSF amyloid beta compared to females, irrespective of diagnosis. We also detected sex differences in hippocampal volume but the direction was dependent on the method of correction. Altogether results suggest that across diagnosis females show greater memory decline compared to males and APOE genotype affects AD neuropathology differently in males and females which may influence sex differences in incidence and progression of aMCI and AD.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteínas E , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Apolipoproteínas E/genética , Biomarcadores/sangre , Disfunción Cognitiva/genética , Disfunción Cognitiva/fisiopatología , Femenino , Genotipo , Humanos , Masculino , Memoria/fisiología , Factores Sexuales
5.
J Alzheimers Dis ; 78(2): 627-641, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33016923

RESUMEN

BACKGROUND: Alzheimer's disease (AD) disproportionately affects females with steeper cognitive decline and more neuropathology compared to males, which is exacerbated in females carrying the APOEɛ4 allele. The risk of developing AD is also higher in female APOEɛ4 carriers in earlier age groups (aged 65-75), and the progression from cognitively normal to mild cognitive impairment (MCI) and to AD may be influenced by sex. Inflammation is observed in AD and is related to aging, stress, and neuroplasticity, and although studies are scarce, sex differences are noted in inflammation. OBJECTIVE: The objective of this study was to investigate underlying physiological inflammatory mechanisms that may help explain why there are sex differences in AD and APOEɛ4 carriers. METHODS: We investigated, using the ADNI database, the effect of sex and APOE genotype (non-carriers or carriers of 1 and 2 APOEɛ4 alleles) and sex and diagnosis (cognitively normal (CN), MCI, AD) on CSF (N = 279) and plasma (N = 527) markers of stress and inflammation. RESULTS: We found CSF IL-16 and IL-8 levels differed by sex and APOE genotype, as IL-16 was higher in female APOEɛ4 carriers compared to non-carriers, while the opposite pattern was observed in males with IL-8. Furthermore, females had on average higher levels of plasma CRP and ICAM1 but lower levels of CSF ICAM1, IL-8, IL-16, and IgA than males. Carrying APOEɛ4 alleles and diagnosis (MCI and AD) decreased plasma CRP in both sexes. CONCLUSION: Sex and APOE genotype differences in CSF and plasma inflammatory biomarkers support that the underlying physiological changes during aging differ by sex and tissue origin.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/metabolismo , Apolipoproteína E4/metabolismo , Mediadores de Inflamación/metabolismo , Caracteres Sexuales , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Apolipoproteína E4/genética , Biomarcadores/metabolismo , Bases de Datos Factuales/tendencias , Femenino , Humanos , Masculino
6.
Neuropharmacology ; 175: 108174, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32497592

RESUMEN

Perinatal depression (PND) affects approximately 15% of women, and de novo postpartum depression affects approximately 40% of PND cases. Selective serotonin reuptake inhibitors (SSRIs) are a common class of antidepressants prescribed to treat PND. However, the safety and efficacy of SSRIs have been questioned in both clinical and preclinical research. Here, using a preclinical rodent model of de novo postpartum depression, we aim to better understand neuroinflammatory cytokines and tryptophan mechanisms that may be related to SSRI efficacy. Rat dams were treated with high corticosterone (CORT; 40 mg/kg, s.c.) for 22 days in the postpartum period to simulate a depressive-like endophenotype. Concurrently, a subset of dams was treated with the SSRI, fluoxetine (FLX; 10 mg/kg, s.c.), in the postpartum period. We showed, consistent with previous studies, that although maternal FLX treatment prevented CORT-induced disturbances in maternal care behavior during the early postpartum, it failed to prevent the expression of CORT-induced passive coping behavior in the late postpartum. Furthermore, FLX treatment, regardless of CORT treatment, increased maternal hippocampal IL-1ß, plasma CXCL1, and decreased maternal plasma tryptophan, 4'-pyridoxic acid, and pyridoxal concentrations. Maternal CORT treatment reduced maternal hippocampal IFN-γ, and both hippocampal and plasma TNF-α. Our work suggests that the limited efficacy of FLX in the late postpartum may be associated with elevated levels of the proinflammatory cytokine IL-1ß in the maternal hippocampus, elevated plasma CXCL1, decreased plasma tryptophan concentration, and changes in vitamin B6 dependent tryptophan-kynurenine pathway. These findings suggest novel pathways for improving SSRI efficacy in alleviating perinatal depression.


Asunto(s)
Depresión Posparto/metabolismo , Fluoxetina/administración & dosificación , Mediadores de Inflamación/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Transducción de Señal/efectos de los fármacos , Triptófano/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Periodo Posparto , Ratas Sprague-Dawley
7.
Horm Behav ; 122: 104734, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32169583

RESUMEN

Oxytocin regulates social behaviours, pair bonding and hippocampal neurogenesis but most studies have used adult males. Our study investigated the effects of oxytocin on social investigation and adult hippocampal neurogenesis in male and female rats. Oxytocin has poor penetration of the blood-brain barrier, therefore we tested a nanoparticle drug, TRIOZAN™ (Ovensa Inc.), which permits greater blood-brain-barrier penetration. Adult male and female rats were injected daily (i.p.) for 10 days with either: oxytocin in PBS (0.5 or 1.0 mg/kg), oxytocin in TRIOZAN™ (0.5 or 1.0 mg/kg), or vehicle (PBS) and tested for social investigation. Oxytocin decreased body mass and increased social investigation and number of oxytocin-immunoreactive cells in the supraoptic nucleus (SON) of the hypothalamus in male rats only. In both sexes, oxytocin decreased the number of immature neurons (doublecortin+ cells) in the ventral hippocampus and reduced plasma 17ß-estradiol levels in a dose- and delivery-dependent way. Oxytocin in TRIOZAN™ reduced "sedation" observed post-injection and increased certain central effects (oxytocin levels in the hypothalamus and neurogenesis in the ventral hippocampus) relative to oxytocin in PBS, indicating that the nanoparticle may be used as an alternative brain delivery system. We showed that oxytocin has sex-specific effects on social investigation, body mass, "sedation", and the oxytocin system. In contrast, similar effects were observed in both sexes in neurogenesis and plasma 17ß-estradiol. Our work suggests that sex differences in oxytocin regulation of brain endpoints is region-specific (hypothalamus versus hippocampus) and that oxytocin does not promote social investigation in females.


Asunto(s)
Hipocampo/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Oxitocina/farmacología , Conducta Social , Animales , Conducta Animal/efectos de los fármacos , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Femenino , Hipocampo/citología , Hipocampo/fisiología , Hipotálamo/citología , Hipotálamo/metabolismo , Inmunohistoquímica , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Neuropéptidos/metabolismo , Oxitocina/metabolismo , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales , Núcleo Supraóptico/citología , Núcleo Supraóptico/efectos de los fármacos , Núcleo Supraóptico/metabolismo
8.
Horm Behav ; 119: 104651, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31790664

RESUMEN

The estrogen receptor (ER) mechanisms by which 17ß-estradiol influences depressive-like behaviour have primarily been investigated acutely and not within an animal model of depression. Therefore, the current study aimed to dissect the contribution of ERα and ERß to the effects of 17ß-estradiol under non-stress and chronic stress conditions. Ovariectomized (OVX) or sham-operated mice were treated chronically (47 days) with 17ß-estradiol (E2), the ERß agonist diarylpropionitrile (DPN), the ERα agonist propylpyrazole-triol (PPT), or vehicle. On day 15 of treatment, mice from each group were assigned to chronic unpredictable stress (CUS; 28 days) or non-CUS conditions. Mice were assessed for anxiety- and depressive-like behaviour and hypothalamic-pituitary-adrenal (HPA) axis function. Cytokine and chemokine levels, and postsynaptic density protein 95 were measured in the hippocampus and frontal cortex, and adult hippocampal neurogenesis was assessed. Overall, the effects of CUS were more robust that those of estrogenic treatments, as seen by increased immobility in the tail suspension test (TST), reduced PSD-95 expression, reduced neurogenesis in the ventral hippocampus, and HPA axis negative feedback dysregulation. However, we also observe CUS-dependent and -independent effects of ovarian status and estrogenic treatments. The effects of CUS on PSD-95 expression, the cytokine milieu, and in TST were largely driven by PPT and DPN, indicating that these treatments were not protective. Independent of CUS, estradiol increased neurogenesis in the dorsal hippocampus, blunted the corticosterone response to an acute stressor, and increased anxiety-like behaviour. These findings provide insights into the complexities of estrogen signaling in modulating depressive-like phenotypes under non-stress and chronic stress conditions.


Asunto(s)
Depresión/metabolismo , Receptor alfa de Estrógeno/agonistas , Receptor beta de Estrógeno/agonistas , Estrés Psicológico/metabolismo , Animales , Enfermedad Crónica , Corticosterona/metabolismo , Depresión/etiología , Depresión/psicología , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Reacción Cataléptica de Congelación/efectos de los fármacos , Reacción Cataléptica de Congelación/fisiología , Hipocampo/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Ratones , Ratones Endogámicos C57BL , Nitrilos/farmacología , Ovariectomía , Fenoles/farmacología , Fenotipo , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/metabolismo , Propionatos/farmacología , Pirazoles/farmacología , Estrés Psicológico/complicaciones , Estrés Psicológico/patología , Estrés Psicológico/psicología
9.
Endocrinology ; 160(9): 2128-2136, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31219567

RESUMEN

Androgens (testosterone and DHT) increase adult hippocampal neurogenesis by increasing survival of new neurons in male rats and mice via an androgen receptor pathway, but it is not known whether androgens regulate neurogenesis in female rats and whether the effect is age-dependent. We investigated the effects of DHT, a potent androgen, on neurogenesis in young adult and middle-aged male and female rats. Rats were gonadectomized and injected with the DNA synthesis marker bromodeoxyuridine (BrdU). The following day, rats began receiving daily injections of oil or DHT for 30 days. We evaluated cell proliferation (Ki67) and survival of new neurons (BrdU and BrdU/NeuN) in the hippocampus of male and female rats by using immunohistochemistry. As expected, DHT increased the number of BrdU+ cells in young males but surprisingly not in middle-aged males or in young and middle-aged females. In middle age, DHT increased the proportion of BrdU/NeuN cells, an effect driven by females. Androgen receptor expression also increased with aging in both female and male rats, which may contribute to a lack of DHT neurogenic effect in middle age. Our results indicate that DHT regulates adult hippocampal neurogenesis in a sex- and age-dependent manner.


Asunto(s)
Dihidrotestosterona/farmacología , Hipocampo/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Factores de Edad , Animales , Femenino , Hipocampo/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Factores Sexuales
10.
Front Neuroendocrinol ; 53: 100740, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30826374

RESUMEN

Becoming a mother is associated with dramatic changes in physiology, endocrinology, immune function, and behaviour that begins during pregnancy and persists into the postpartum. Evidence also suggests that motherhood is accompanied by long-term changes in brain function. In this review, we summarize the short (pregnancy and postpartum) and long-term (beyond the postpartum and into middle age) effects of pregnancy and motherhood on cognition, neuroplasticity, and neuroimmune signalling. We also discuss the effects of previous history of pregnancy and motherhood (parity) on brain health and disease (neurodegenerative diseases and stroke outcomes) and on efficacy of hormone and antidepressant treatments. Finally, we argue that pregnancy and motherhood are unique female experiences that need to be taken into account to better understand female brain function and aging.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Cognición/fisiología , Madres , Periodo Posparto/fisiología , Animales , Humanos , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología
11.
Neurobiol Aging ; 70: 291-307, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30056312

RESUMEN

Menopause is associated with cognitive decline, and hormone therapies (HTs) may improve cognition depending on type and timing of HTs. Previous parity may influence cognition in later life. We investigated how primiparity and long-term ovariectomy influence cognition, neurogenesis, hormones, cytokines, and neuronal activation in middle-aged rats in response to Premarin, an HT. Nulliparous and primiparous rats were sham-ovariectomized or ovariectomized, administered vehicle or Premarin 6 months later, and all rats were trained in the Morris water maze. Premarin improved early spatial learning and memory in nulliparous rats but impaired early learning in primiparous rats. With training, primiparity increased hippocampal neurogenesis, and Premarin decreased immature neurons, regardless of parity. Moreover, Premarin increased serum tumor necrosis factor α and the CXC chemokine ligand 1 (CXCL1) in trained nulliparous, but not primiparous, rats. However, Premarin decreased the expression of the immediate early gene zif268 in the dorsal CA3 region in primiparous rats after training. Thus, primiparity alters how Premarin affects spatial learning, neuronal activation, and serum cytokines. These findings have implications for the treatment of age-associated cognitive decline in women.


Asunto(s)
Citocinas/sangre , Estrógenos Conjugados (USP)/administración & dosificación , Estrógenos/administración & dosificación , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Paridad , Aprendizaje Espacial/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Proteínas de Dominio Doblecortina , Femenino , Hipocampo/fisiología , Terapia de Reemplazo de Hormonas , Conducta Materna/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/fisiología , Neuropéptidos/metabolismo , Ovariectomía , Ratas Sprague-Dawley , Aprendizaje Espacial/fisiología , Memoria Espacial/efectos de los fármacos , Memoria Espacial/fisiología
13.
Horm Behav ; 74: 37-52, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26122299

RESUMEN

This article is part of a Special Issue "Estradiol and Cognition". There are sex differences in hippocampus-dependent cognition and neurogenesis suggesting that sex hormones are involved. Estrogens modulate certain forms of spatial and contextual memory and neurogenesis in the adult female rodent, and to a lesser extent male, hippocampus. This review focuses on the effects of sex and estrogens on hippocampal learning, memory, and neurogenesis in the young and aged adult rodent. We discuss how factors such as the type of estrogen, duration and dose of treatment, timing of treatment, and type of memory influence the effects of estrogens on cognition and neurogenesis. We also address how reproductive experience (pregnancy and mothering) and aging interact with estrogens to modulate hippocampal cognition and neurogenesis in females. Given the evidence that adult hippocampal neurogenesis plays a role in long-term spatial memory and pattern separation, we also discuss the functional implications of regulating neurogenesis in the hippocampus.


Asunto(s)
Envejecimiento/efectos de los fármacos , Envejecimiento/fisiología , Estrógenos/farmacología , Hipocampo/crecimiento & desarrollo , Memoria/fisiología , Neurogénesis/fisiología , Animales , Cognición/efectos de los fármacos , Cognición/fisiología , Estradiol/farmacología , Femenino , Hipocampo/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Embarazo , Roedores , Caracteres Sexuales
14.
PLoS One ; 10(6): e0129880, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26075609

RESUMEN

Estradiol increases cell proliferation in the dentate gyrus of the female rodent but it is not known whether the G protein-coupled estrogen receptor (GPER), a membrane receptor, is involved in this process, nor whether there are regional differences in estradiol's effects on cell proliferation. Thus, we investigated whether estradiol exerts its effects on cell proliferation in the dorsal and ventral dentate gyrus through GPER, using the GPER agonist, G1, and antagonist, G15. Ovariectomized adult female rats received a single injection of either: 17ß-estradiol (10 µg), G1 (0.1, 5, 10 µg), G15 (40 µg), G15 and estradiol, or vehicle (oil, DMSO, or oil+DMSO). After 30 min, animals received an injection of bromodeoxyuridine (BrdU) and were perfused 24 h later. Acute treatment with estradiol increased, while the GPER agonist G1 (5 µg) decreased, the number of BrdU+ cells in the dentate gyrus relative to controls. The GPER antagonist, G15 increased the number of BrdU+ cells relative to control in the dorsal region and decreased the number of BrdU+ cells in the ventral region. However, G15 treatment in conjunction with estradiol partially eliminated the estradiol-induced increase in cell proliferation in the dorsal dentate gyrus. Furthermore, G1 decreased the expression of GPER in the dentate gyrus but not the CA1 and CA3 regions of the hippocampus. In summary, we found that activation of GPER decreased cell proliferation and GPER expression in the dentate gyrus of young female rats, presenting a potential and novel estrogen-independent role for this receptor in the adult hippocampus.


Asunto(s)
Estradiol/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Animales , Benzodioxoles/farmacología , Peso Corporal , Proliferación Celular/efectos de los fármacos , Ciclopentanos/farmacología , Giro Dentado/anatomía & histología , Giro Dentado/efectos de los fármacos , Giro Dentado/metabolismo , Estradiol/administración & dosificación , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/anatomía & histología , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Tamaño de los Órganos , Quinolinas/farmacología , Ratas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
15.
Gen Comp Endocrinol ; 203: 69-85, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24685768

RESUMEN

Thyroid hormones (THs) are well-known regulators of development and metabolism in vertebrates. There is increasing evidence that THs are also involved in gonadal differentiation and reproductive function. Changes in TH status affect sex ratios in developing fish and frogs and reproduction (e.g., fertility), hormone levels, and gonad morphology in adults of species of different vertebrates. In this review, we have summarized and compared the evidence for cross-talk between the steroid hormone and thyroid axes and present a comparative model. We gave special attention to TH regulation of sex steroid synthesis and action in both the brain and gonad, since these are important for gonad development and brain sexual differentiation and have been studied in many species. We also reviewed research showing that there is a TH system, including receptors and enzymes, in the brains and gonads in developing and adult vertebrates. Our analysis shows that THs influences sex steroid hormone synthesis in vertebrates, ranging from fish to pigs. This concept of crosstalk and conserved hormone interaction has implications for our understanding of the role of THs in reproduction, and how these processes may be dysregulated by environmental endocrine disruptors.


Asunto(s)
Hormonas Esteroides Gonadales/metabolismo , Gónadas/metabolismo , Receptor Cross-Talk/fisiología , Diferenciación Sexual/fisiología , Hormonas Tiroideas/metabolismo , Vertebrados/metabolismo , Animales , Femenino , Masculino , Reproducción/fisiología , Glándula Tiroides/metabolismo
16.
Gen Comp Endocrinol ; 177(1): 195-204, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22487252

RESUMEN

Gonadal differentiation in frogs is affected by sex steroids and thyroid hormones (THs); however, the genes controlling differentiation and the molecular effects of THs in the gonad are not clear and have only been investigated in a few anuran species. In this study, we established developmental profiles and TH regulation of sex steroid- and TH-related genes in the gonad-mesonephros complex (GMC) of the túngara frog (Physalaemus pustulosus), and compared the results to our previous research in another tropical frog, Silurana tropicalis. The developmental profiles allowed us to identify three genes as markers of ovarian development. During metamorphosis, aromatase (cyp19), estrogen receptor α, and steroid 5α-reductase 1 (srd5alpha1) were higher in the GMC of putative and morphological females. Acute exposure to triiodothyronine (T3) decreased GMC expression of srd5alpha1 and cyp19, while increasing TH-related genes in premetamorphic tadpoles. The regulation of sex steroid-related genes differed significantly from our previous study in S. tropicalis. P. pustulosus and S. tropicalis share ecological, developmental, and reproductive characteristics; however, they are not closely related. These results along with our previous research in the tadpole brain support the hypothesis that evolutionary convergence is not important in understanding differences in the effects of TH on sex steroid-related genes in frogs. Finally, we propose that T3 induces male gonadal development but this can be achieved through different mechanisms depending on the species.


Asunto(s)
Gónadas/metabolismo , Hormonas Tiroideas/metabolismo , Triyodotironina/metabolismo , Animales , Anuros , Receptor alfa de Estrógeno/metabolismo , Femenino , Masculino , Oxidorreductasas/metabolismo
17.
Brain Behav Evol ; 79(2): 98-112, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22142812

RESUMEN

In amphibians, thyroid hormones (THs) are considered key regulators of brain remodeling during metamorphosis, while sex steroids (estrogens and androgens) control sexual differentiation and gonadal development. However, these two endocrine axes can interact during tadpole brain development. Previously, we demonstrated that THs affect sex steroid-related gene expression in the developing brain of Silurana tropicalis and Rana pipiens; however, the gene expression changes differed between species. We chose to study a third anuran species, Physalaemus pustulosus, to test new hypotheses about the role of THs in the regulation of brain gene expression. We first established developmental transcript profiles of TH- and sex steroid-related genes in the brain of P. pustulosus. Then, following the same protocols as in our previous studies, we investigated triiodothyronine (T3) regulation of brain transcripts in premetamorphic P. pustulosus and then compared the results with our previous two studies. In the case of TH-related genes, TH receptor beta (trbeta) and deiodinase type 3 (dio3), mRNA developmental profiles were similar in the three species and with respect to other species in the published literature. However, the profiles of TH receptor alpha (tralpha) and deiodinase type 2 (dio2) mRNA revealed differences between anuran species. Among the three anurans we have studied, the direction of the T3 regulation of TH-related genes was overall similar, but the magnitude of gene expression change differed depending on the rate of metamorphosis in a given species. For the sex steroid-related genes, each species exhibited similar developmental profiles but differed in their response to T3. In P. pustulosus, T3 reduced the expression of aromatase (cyp19) while increasing mRNA levels of androgen and estrogen receptors. These results are similar to previous research in R. pipiens but differ from data for S. tropicalis, for which we found an increase in androgen synthesis enzymes but no effect on cyp19. Together, we propose that T3 has the potential to induce the brain androgen system in anurans. This could be achieved by increasing androgen synthesis enzymes (S. tropicalis) or by decreasing estrogen synthesis (due to a decrease in cyp19 in P. pustulosus and R. pipiens). In conclusion, we demonstrated that mechanisms of hormone interactions differ between anuran species, but in all cases T3 appears to affect the balance of sex steroids in the brain, stimulating the androgen system. We have shown that the regulation of sex steroid-related genes by T3 is more similar among closely related species than species with similar reproductive and developmental characteristics.


Asunto(s)
Anuros/crecimiento & desarrollo , Encéfalo/crecimiento & desarrollo , Larva/crecimiento & desarrollo , Metamorfosis Biológica/fisiología , Hormonas Tiroideas/fisiología , Transcripción Genética , Andrógenos/genética , Andrógenos/fisiología , Animales , Anuros/genética , Anuros/fisiología , Encéfalo/fisiología , Estrógenos/genética , Estrógenos/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Larva/genética , Metamorfosis Biológica/genética , Rana pipiens/genética , Rana pipiens/crecimiento & desarrollo , Rana pipiens/fisiología , Receptores de Hormona Tiroidea/fisiología , Especificidad de la Especie , Hormonas Tiroideas/genética
18.
Mol Cell Endocrinol ; 331(1): 143-9, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20837100

RESUMEN

In amphibians, the main role of thyroid hormones (THs) is to regulate metamorphosis; however, there is evidence that THs also affect gonadal sexual differentiation. In this study, Silurana (Xenopus) tropicalis tadpoles were exposed to triiodothyronine (T3; 0, 0.5, 5 and 50 nM), the bioactive form of THs for 48h. Real-time RT-PCR analyses in the gonad-mesonephros complex (GMC) revealed that TH- and androgen-related genes were positively regulated, while estrogen receptor ß was negatively regulated by T3. Together, these results are in agreement with the masculinizing effect of THs in amphibians. Profiles of TH- and sex steroid-related genes in the GMC during metamorphosis of S. tropicalis suggest that THs are important regulators of sex steroid-related gene expression in the GMC. This study provides evidence that the GMC is a target of THs but that a complex interplay exists between THs and sex steroids during gonadal sexual development.


Asunto(s)
Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hormonas Esteroides Gonadales/genética , Gónadas/metabolismo , Mesonefro/metabolismo , Triyodotironina/farmacología , Xenopus/genética , Animales , Estrógenos/metabolismo , Femenino , Hormonas Esteroides Gonadales/metabolismo , Gónadas/efectos de los fármacos , Humanos , Masculino , Mesonefro/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Diferenciación Sexual/efectos de los fármacos , Diferenciación Sexual/genética , Triyodotironina/administración & dosificación , Triyodotironina/genética , Triyodotironina/metabolismo , Xenopus/embriología
19.
Environ Toxicol Chem ; 29(8): 1804-15, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20821635

RESUMEN

Exposure to multiple environmental stressors is negatively impacting the health of amphibians worldwide. Increased exposure to ultraviolet B radiation (UVBR) and chemical pollutants may affect amphibian populations by disrupting metamorphosis; however, the actual mechanisms by which these stressors affect development remain unknown. Because amphibian metamorphosis is controlled by thyroid hormones (TH), changes in developmental rates by environmental stress suggest a disruption of the thyroid system. Tadpoles were chronically exposed to environmental levels of UVBR (average of 0.15 W/m2) and 4-tert-octylphenol (OP; 10 nM), alone and combined, prior to being challenged to exogenous TH triiodothyronine (T3; 5 or 50 nM). This experimental approach was taken to determine whether exposure to these stressors affects the ability of T3 to elicit specific molecular and morphological responses. Exposure to OP increased mRNA levels of thyroid receptors (TRs) alpha and beta, deiodinase type 2 (D2), and corticotropin releasing hormone in the brain and of D2 in the tail of tadpoles. 4-tert-octylphenol also enhanced T3-induced expression of D2 in the brain. The combination of UVBR and OP affected the expression of TR alpha in the brain and the responses of TR alpha and beta genes to T3 in the tail, demonstrating the importance of considering the effects of multiple stressors on amphibians. Tadpoles exposed to UVBR were developmentally delayed and exhibited slowed tail resorption and accelerated hindlimb development following exposure to T3. Together, these findings indicate that UVBR alters the rate of development and TH-dependent morphological changes at metamorphosis, and that exposure to UVBR and/or OP disrupts the expression of genes important for development and the biological action of T3 in peripheral tissues. Our group is the first to demonstrate that environmental levels of UVBR and/or OP can affect the thyroid system of amphibians.


Asunto(s)
Fenoles/toxicidad , Rana pipiens/metabolismo , Tensoactivos/toxicidad , Triyodotironina/toxicidad , Rayos Ultravioleta , Contaminantes Químicos del Agua/toxicidad , Animales , Larva/efectos de los fármacos , Larva/efectos de la radiación , Metamorfosis Biológica/efectos de los fármacos , Metamorfosis Biológica/efectos de la radiación , ARN Mensajero/metabolismo , Rana pipiens/crecimiento & desarrollo , Estrés Fisiológico , Triyodotironina/genética , Triyodotironina/metabolismo
20.
Aquat Toxicol ; 100(4): 354-64, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20864192

RESUMEN

Fluoxetine (FLX) is a pharmaceutical acting as a selective serotonin reuptake inhibitor and is used to treat depression in humans. Fluoxetine and the major active metabolite norfluoxetine (NFLX) are released to aquatic systems via sewage-treatment effluents. They have been found to bioconcentrate in wild fish, raising concerns over potential endocrine disrupting effects. The objective of this study was to determine effects of waterborne FLX, including environmental concentrations, on the reproductive axis in sexually mature male goldfish. We initially cloned the goldfish serotonin transporter to investigate tissue and temporal expression of the serotonin transporter, the FLX target, in order to determine target tissues and sensitive exposure windows. Sexually mature male goldfish, which showed the highest levels of serotonin transporter expression in the neuroendocrine brain, were exposed to FLX at 0.54µg/L and 54µg/L in a 14-d exposure before receiving vehicle or sex pheromone stimulus consisting of either 4.3nM 17,20ß-dihydroxy-4-pregnene-3-one (17,20P) or 3nM prostaglandin F2(α) (PGF2(α)). Reproductive endpoints assessed included gonadosomatic index, milt volume, and blood levels of the sex steroids testosterone and estradiol. Neuroendocrine function was investigated by measuring blood levels of luteinizing hormone, growth hormone, pituitary gene expression of luteinizing hormone, growth hormone and follicle-stimulating hormone and neuroendocrine brain expression of isotocin and vasotocin. To investigate changes at the gonadal level of the reproductive axis, testicular gene expression of the gonadotropin receptors, both the luteinizing hormone receptor and the follicle-stimulating hormone receptor, were measured as well as expression of the growth hormone receptor. To investigate potential impacts on spermatogenesis, testicular gene expression of the spermatogenesis marker vasa was measured and histological samples of testis were analyzed qualitatively. Estrogen indices were measured by expression and activity analysis of gonadal aromatase, as well as liver expression analysis of the estrogenic marker, esr1. After 14d, basal milt volume significantly decreased at 54µg/L FLX while pheromone-stimulated milt volume decreased at 0.54µg/L and 54µg/L FLX. Fluoxetine (54µg/L) inhibited both basal and pheromone-stimulated testosterone levels. Significant concentration-dependent reductions in follicle-stimulating hormone and isotocin expression were observed with FLX in the 17,20P- and PGF2(α)-stimulated groups, respectively. Estradiol levels and expression of esr1 concentration-dependently increased with FLX. This study demonstrates that FLX disrupts reproductive physiology of male fish at environmentally relevant concentrations, and potential mechanisms are discussed.


Asunto(s)
Fluoxetina/toxicidad , Carpa Dorada/fisiología , Reproducción/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/toxicidad , Contaminantes Químicos del Agua/toxicidad , Animales , Aromatasa/metabolismo , Secuencia de Bases , Receptor alfa de Estrógeno/metabolismo , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Expresión Génica/efectos de los fármacos , Carpa Dorada/genética , Hígado/metabolismo , Masculino , Datos de Secuencia Molecular , Oxitocina/análogos & derivados , Oxitocina/genética , Oxitocina/metabolismo , Prostaglandinas F/genética , Prostaglandinas F/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/efectos de los fármacos , Testículo/metabolismo , Testosterona/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...