Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
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
2.
JAMA Netw Open ; 4(7): e2118223, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34297070

RESUMEN

Importance: Methadone access may be uniquely vulnerable to disruption during COVID-19, and even short delays in access are associated with decreased medication initiation and increased illicit opioid use and overdose death. Relative to Canada, US methadone provision is more restricted and limited to specialized opioid treatment programs. Objective: To compare timely access to methadone initiation in the US and Canada during COVID-19. Design, Setting, and Participants: This cross-sectional study was conducted from May to June 2020. Participating clinics provided methadone for opioid use disorder in 14 US states and territories and 3 Canadian provinces with the highest opioid overdose death rates. Statistical analysis was performed from July 2020 to January 2021. Exposures: Nation and type of health insurance (US Medicaid and US self-pay vs Canadian provincial). Main Outcomes and Measures: Proportion of clinics accepting new patients and days to first appointment. Results: Among 268 of 298 US clinics contacted as a patient with Medicaid (90%), 271 of 301 US clinics contacted as a self-pay patient (90%), and 237 of 288 Canadian clinics contacted as a patient with provincial insurance (82%), new patients were accepted for methadone at 231 clinics (86%) during US Medicaid contacts, 230 clinics (85%) during US self-pay contacts, and at 210 clinics (89%) during Canadian contacts. Among clinics not accepting new patients, at least 44% of 27 clinics reported that the COVID-19 pandemic was the reason. The mean wait for first appointment was greater among US Medicaid contacts (3.5 days [95% CI, 2.9-4.2 days]) and US self-pay contacts (4.1 days [95% CI, 3.4-4.8 days]) than Canadian contacts (1.9 days [95% CI, 1.7-2.1 days]) (P < .001). Open-access model (walk-in hours for new patients without an appointment) utilization was reported by 57 Medicaid (30%), 57 self-pay (30%), and 115 Canadian (59%) contacts offering an appointment. Conclusions and Relevance: In this cross-sectional study of 2 nations, more than 1 in 10 methadone clinics were not accepting new patients. Canadian clinics offered more timely methadone access than US opioid treatment programs. These results suggest that the methadone access shortage was exacerbated by COVID-19 and that changes to the US opioid treatment program model are needed to improve the timeliness of access. Increased open-access model adoption may increase timely access.


Asunto(s)
COVID-19 , Accesibilidad a los Servicios de Salud , Metadona/uso terapéutico , Tratamiento de Sustitución de Opiáceos , Trastornos Relacionados con Opioides/terapia , Pandemias , Listas de Espera , Instituciones de Atención Ambulatoria , Analgésicos Opioides , Canadá , Estudios Transversales , Financiación Personal , Servicios de Salud , Seguro de Salud , Medicaid , Estados Unidos
3.
Psychoneuroendocrinology ; 106: 111-116, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30974324

RESUMEN

Letrozole, a third-generation aromatase inhibitor, prevents the production of estrogens in the final step in conversion from androgens. Due to its efficacy at suppressing estrogens, letrozole has recently taken favor as a first-line adjuvant treatment for hormone-responsive breast cancer in middle-aged women. Though patient response to letrozole has generally been positive, there is conflicting evidence surrounding its effects on the development of depression. It is possible that the potential adverse effects of letrozole on mood are a result of the impact of hormonal fluctuations on neurogenesis in the hippocampus. Thus, to clarify the effects of letrozole on the hippocampus and behavior, we examined how chronic administration affects hippocampal neurogenesis and depressive-like behavior in middle-aged, intact female mice. Mice were given either letrozole (1 mg/kg) or vehicle by injection (i.p.) daily for 3 weeks. Depressive-like behavior was assessed during the last 3 days of treatment using the forced swim test, tail suspension test, and sucrose preference test. The production of new neurons was quantified using the immature neuronal marker doublecortin (DCX), and cell proliferation was quantified using the endogenous marker Ki67. We found that letrozole increased DCX and Ki67 expression and maturation in the dentate gyrus, but had no significant effect on depressive-like behavior. Our findings suggest that a reduction in estrogens in middle-aged females increases hippocampal neurogenesis without any adverse impact on depressive-like behavior; as such, this furthers our understanding of how estrogens modulate neurogenesis, and to the rationale for the utilization of letrozole in the clinical management of breast cancer.


Asunto(s)
Hipocampo/efectos de los fármacos , Letrozol/metabolismo , Neurogénesis/efectos de los fármacos , Animales , Aromatasa/metabolismo , Inhibidores de la Aromatasa/farmacología , Conducta Animal/efectos de los fármacos , Proliferación Celular , Giro Dentado/metabolismo , Depresión/tratamiento farmacológico , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Estrógenos/metabolismo , Femenino , Hipocampo/metabolismo , Letrozol/farmacología , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Células-Madre Neurales , Neuronas/metabolismo , Neuropéptidos/metabolismo
4.
Neurobiol Aging ; 78: 1-17, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30825663

RESUMEN

The maternal brain displays considerable plasticity, and motherhood is associated with changes in affective and cognitive function. Motherhood can alter the trajectory of brain aging, including modifications to neuroplasticity and cognition. Here, we investigated the short- and long-term effects of motherhood on hippocampal neurogenesis, microglial density and morphology, and circulating cytokines, domains known to be altered with age and implicated in cognition and mood. Female rats were bred then euthanized during gestation or at various postpartum time points, culminating in middle age, and nulliparous rats served as age-matched controls. Hippocampal neurogenesis was significantly suppressed during gestation and the postpartum period. Interestingly, neurogenesis declined significantly in middle-aged nulliparous rats but increased in primiparous rats across the same period. Transient postpartum adaptations to the neuroimmune environment of the hippocampus were evidenced, as Iba-1-immunoreactive microglia assumed a deramified morphology followed by increased density. Intriguingly, aging-related changes in circulating cytokines were dependent on parity. These adaptations in neurogenic and immune processes may have ramifications for maternal mood and cognition across the peripartum period and beyond.


Asunto(s)
Envejecimiento , Citocinas/metabolismo , Hipocampo/fisiología , Microglía/fisiología , Madres , Neurogénesis/fisiología , Afecto , Envejecimiento/inmunología , Envejecimiento/metabolismo , Envejecimiento/patología , Envejecimiento/fisiología , Animales , Proteínas de Unión al Calcio , Recuento de Células , Cognición , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Femenino , Hipocampo/inmunología , Humanos , Masculino , Proteínas de Microfilamentos , Microglía/patología , Proteínas Asociadas a Microtúbulos , Madres/psicología , Neuroinmunomodulación , Neuropéptidos , Periodo Posparto/fisiología , Periodo Posparto/psicología , Embarazo , Ratas Sprague-Dawley
5.
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
6.
Neuropharmacology ; 107: 278-293, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27018449

RESUMEN

Depression is more prevalent in women than in men, and women are at a heightened risk for depression during the postpartum and perimenopause. There is also evidence to suggest that the ovarian hormone milieu may dictate antidepressant efficacy. Thus, it is important to investigate the role of ovarian hormones in the pathogenesis of depression and in the mechanisms that may underlie antidepressant efficacy. In the present study, we used 10-month-old female Sprague-Dawley rats to examine the effects of long-term ovarian hormone deprivation on the development of depressive-like endophenotypes after chronic stress, and on antidepressant efficacy. Four months following ovariectomy (OVX) or sham surgery, all rats were subjected to 6 weeks of chronic unpredictable stress (CUS). During the last 3 weeks of CUS, rats received daily injections of fluoxetine (5 mg/kg) or vehicle. All rats were assessed on measures of anxiety- and depressive-like behavior, hypothalamic-pituitary-adrenal (HPA) negative feedback inhibition, and on markers of neurogenesis and microglia in the dentate gyrus. Our findings demonstrate that long-term ovarian hormone deprivation increased anxiety and depressive-like behavior, as seen by increased immobility in the forced swim test and latency to feed in the novelty suppressed feeding test, and decreased sucrose preference. Further, long-term OVX resulted in impaired HPA negative feedback inhibition, as seen in the dexamethasone suppression test. Fluoxetine treatment showed limited behavioral and neuroendocrine efficacy, however it reduced microglial (Iba-1) expression, and increased cell proliferation, neurogenesis (via cell survival), and the expression of the polysialylated neuronal cell adhesion molecule (PSA-NCAM) in the dentate gyrus, although these effects varied by region (dorsal, ventral) and ovarian status. Taken together, our findings demonstrate that ovarian hormones may impart resilience against the behavioral and neuroendocrine consequences of chronic unpredictable stress, and may modulate the effects of fluoxetine on cell proliferation, neurogenesis, and PSA-NCAM in the middle-aged female.


Asunto(s)
Corticosterona/sangre , Fluoxetina/uso terapéutico , Ovariectomía/efectos adversos , Ovario/metabolismo , Estrés Psicológico/sangre , Estrés Psicológico/tratamiento farmacológico , Factores de Edad , Animales , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Modelos Animales de Enfermedad , Estradiol/sangre , Femenino , Fluoxetina/farmacología , Ovariectomía/tendencias , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/psicología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA