Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Horm Behav ; 147: 105281, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36434852

RESUMEN

Aromatase inhibitors (AIs) are a class of drugs commonly given to patients with estrogen receptor (ER)-dependent breast cancers to reduce estrogenic stimulation. However, AIs like Letrozole are associated with negative side effects such as cognitive deficits, sleep disturbances and hot flashes. We have previously shown that these negative effects can be recapitulated in common marmosets (Callithrix jacchus) treated with Letrozole (20 µg daily) for 4 weeks and that marmosets treated with Letrozole show increased levels of estradiol in the hippocampus (Gervais et al., 2019). In order to better understand the mechanisms through which AIs affect cognitive function and increase steroid levels in the hippocampus, we used bulk, paired-end RNA-sequencing to examine differentially expressed genes among Letrozole-treated (LET; n = 8) and vehicle-treated (VEH; n = 8) male and female animals. Gene ontology results show significant reduction across hundreds of categories, some of the most significant being inflammatory response, stress response, MHC Class II protein complex binding, T-cell activation, carbohydrate binding and signaling receptor binding in LET animals. GSEA results indicate that LET females, but not LET males, show enrichment for hormonal gene sets. Based on the transcriptional changes observed, we conclude that AIs may differentially affect the sexes in part due to processes mediated by the CYP-450 superfamily. Ongoing studies will further investigate the longitudinal effects of AIs on behavior and whether AIs increase the risk of stress-induced neurodegeneration.


Asunto(s)
Callithrix , Nitrilos , Masculino , Animales , Femenino , Letrozol/farmacología , Nitrilos/farmacología , Triazoles/farmacología , Inhibidores de la Aromatasa/farmacología , Estrona , Hipocampo , Expresión Génica
2.
Am J Primatol ; 84(9): e23427, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35942572

RESUMEN

Olfactory dysfunction has been identified as an early biomarker for dementia risk but has rarely been assessed in nonhuman primate models of human aging. To better characterize common marmosets as such models, we assessed olfactory discrimination performance in a sample of 10 animals (5 females), aged 2.5-8.9 years old. The monkeys were proficient in the discrimination and reversal of visual stimuli but naïve to odor stimuli. For olfactory discrimination, the monkeys performed a series of six discriminations of increasing difficulty between two odor stimuli. We found no evidence for an age-related decline as both young and older individuals were able to perform the discriminations in roughly the same number of trials. In addition, the older monkeys had faster responses than the younger animals. However, we noted that when adjusted for age, the speed of acquisition of the first discrimination in the olfactory modality was inversely correlated to the speed of acquisition of their first discrimination of two visual stimuli months earlier. These results suggest that marmosets may compensate for sensory deficits in one modality with higher sensory performance in another. These data have broad implications for the assessment of age-related cognitive decline and the categorization of animals as impaired or nonimpaired.


Asunto(s)
Callithrix , Disfunción Cognitiva , Animales , Callithrix/fisiología , Aprendizaje Discriminativo , Femenino , Humanos , Aprendizaje , Percepción Visual
3.
Am J Primatol ; 83(11): e23309, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34403529

RESUMEN

Aging across the Primate Order is poorly understood because ages of individuals are often unknown, there is a dearth of aged animals available for study, and because aging is best characterized by longitudinal studies which are difficult to carry out in long-lived species. The human population is aging rapidly, and advanced age is a primary risk factor for several chronic diseases and conditions that impact healthspan. As lifespan has increased, diseases and disorders of the central nervous system (CNS) have become more prevalent, and Alzheimer's disease and related dementias have become epidemic. Nonhuman primate (NHP) models are key to understanding the aging primate CNS. This Special Issue presents a review of current knowledge about NHP CNS aging across the Primate Order. Similarities and differences to human aging, and their implications for the validity of NHP models of aging are considered. Topics include aging-related brain structure and function, neuropathologies, cognitive performance, social behavior and social network characteristics, and physical, sensory, and motor function. Challenges to primate CNS aging research are discussed. Together, this collection of articles demonstrates the value of studying aging in a breadth of NHP models to advance our understanding of human and nonhuman primate aging and healthspan.


Asunto(s)
Disfunción Cognitiva , Primates , Envejecimiento , Animales , Biología , Enfermedad Crónica , Estados Unidos
4.
Am J Primatol ; 83(11): e23271, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34018622

RESUMEN

Age-related cognitive decline has been extensively studied in humans, but the majority of research designs are cross-sectional and compare across younger and older adults. Longitudinal studies are necessary to capture variability in cognitive aging trajectories but are difficult to carry out in humans and long-lived nonhuman primates. Marmosets are an ideal primate model for neurocognitive aging as their naturally short lifespan facilitates longitudinal designs. In a longitudinal study of marmosets tested on reversal learning starting in middle-age, we found that, on average, the group of marmosets declined in cognitive performance around 8 years of age. However, we found highly variable patterns of cognitive aging trajectories across individuals. Preliminary analyses of brain tissues from this cohort also show highly variable degrees of neuropathology. Future work will tie together behavioral trajectories with brain pathology and provide a window into the factors that predict age-related cognitive decline.


Asunto(s)
Envejecimiento , Callithrix , Animales , Estudios Transversales , Longevidad , Estudios Longitudinales
5.
Am J Primatol ; 83(11): e23299, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34255875

RESUMEN

While humans exhibit a significant degree of neuropathological changes associated with deficits in cognitive and memory functions during aging, non-human primates (NHP) present with more variable expressions of pathological alterations among individuals and species. As such, NHP with long life expectancy in captivity offer an opportunity to study brain senescence in the absence of the typical cellular pathology caused by age-related neurodegenerative illnesses commonly seen in humans. Age-related changes at neuronal population, single cell, and synaptic levels have been well documented in macaques and marmosets, while age-related and Alzheimer's disease-like neuropathology has been characterized in additional species including lemurs as well as great apes. We present a comparative overview of existing neuropathologic observations across the primate order, including classic age-related changes such as cell loss, amyloid deposition, amyloid angiopathy, and tau accumulation. We also review existing cellular and ultrastructural data on neuronal changes, such as dendritic attrition and spine alterations, synaptic loss and pathology, and axonal and myelin pathology, and discuss their repercussions on cellular and systems function and cognition.


Asunto(s)
Envejecimiento , Encéfalo/patología , Primates , Enfermedad de Alzheimer , Animales , Angiopatía Amiloide Cerebral
6.
J Neurosci ; 39(5): 918-928, 2019 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-30587540

RESUMEN

Breast cancer patients using aromatase inhibitors (AIs) as an adjuvant therapy often report side effects, including hot flashes, mood changes, and cognitive impairment. Despite long-term use in humans, little is known about the effects of continuous AI administration on the brain and cognition. We used a primate model of human cognitive aging, the common marmoset, to examine the effects of a 4-week daily administration of the AI letrozole (20 µg, p.o.) on cognition, anxiety, thermoregulation, brain estrogen content, and hippocampal pyramidal cell physiology. Letrozole treatment was administered to both male and female marmosets and reduced peripheral levels of estradiol (E2), but unexpectedly increased E2 levels in the hippocampus. Spatial working memory and intrinsic excitability of hippocampal neurons were negatively affected by the treatment possibly due to increased hippocampal E2. While no changes in hypothalamic E2 were observed, thermoregulation was disrupted by letrozole in females only, indicating some impact on hypothalamic activity. These findings suggest adverse effects of AIs on the primate brain and call for new therapies that effectively prevent breast cancer recurrence while minimizing side effects that further compromise quality of life.SIGNIFICANCE STATEMENT Aromatase inhibitors (AIs) are used as an adjuvant therapy for estrogen-receptor-positive breast cancer and are associated with side effects, including hot flashes, depression/anxiety, and memory deficits severe enough for many women to discontinue this life-saving treatment. AIs are also used by men, yet sex differences in the reported side effects have not been systematically studied. We show that AI-treated male and female marmosets exhibit behavioral changes consistent with these CNS symptoms, as well as elevated hippocampal estradiol and compromised hippocampal physiology. These findings illustrate the need for (1) a greater understanding of the precise mechanisms by which AIs impact brain function and (2) the development of new treatment approaches for breast cancer patients that minimize adverse effects on the brain.


Asunto(s)
Inhibidores de la Aromatasa/efectos adversos , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Letrozol/efectos adversos , Animales , Ansiedad/inducido químicamente , Ansiedad/psicología , Regulación de la Temperatura Corporal/efectos de los fármacos , Química Encefálica/efectos de los fármacos , Callithrix , Cognición/efectos de los fármacos , Estradiol/metabolismo , Estrógenos/metabolismo , Hipocampo/citología , Hipocampo/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Caracteres Sexuales
7.
Am J Primatol ; 81(9): e23057, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31566763

RESUMEN

Population hand preferences are rare in nonhuman primates, but individual hand preferences are consistent over a lifetime and considered to reflect an individual's preference to use a particular hemisphere when engaged in a specific task. Previous findings in marmosets have indicated that left-handed individuals tend to be more fearful than their right-handed counterparts. Based on these findings, we tested the hypotheses that left-handed marmosets are (a) more reactive to a social stressor and (b) are slower than right-handed marmosets in acquiring a reversal learning task. We examined the hand preference of 27 male and female marmosets (ages of 4-7 years old) previously tested in a social separation task and a reversal learning task. Hand preference was determined via a simple reaching task. In the social separation task, monkeys were separated from their partner and the colony for a single 7-hr session. Urinary cortisol levels and behavior were assessed at baseline, during the separation and 24 hr postseparation. Hand preferences were equally distributed between left (n = 10), right-handed (n = 10), and ambidextrous (n = 7) individuals. The separation phase was associated with an increase in cortisol levels and behavioral changes that were similar across handedness groups. However, cortisol levels at baseline were positively correlated with right-handedness, and this relationship was stronger in females than in males. In addition, the occurrence of social behaviors (pre- and postseparation) was positively correlated with right-handedness in both sexes. Baseline cortisol levels did not correlate significantly with social behavior. Acquisition of the reversals was poorer in females than males but did not differ as a function of handedness. We conclude that (a) both stress reactivity and cognitive flexibility are similar across handedness groups and (b) left-handers exhibit less social behavior and have lower basal cortisol levels than ambidextrous and right-handed subjects. The underlying causes for these differences remain to be established.


Asunto(s)
Callithrix/fisiología , Cognición , Lateralidad Funcional , Hidrocortisona/sangre , Conducta Social , Estrés Psicológico/etiología , Animales , Callithrix/psicología , Femenino , Masculino , Valores de Referencia
8.
Am J Primatol ; 81(2): e22924, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30281810

RESUMEN

The common marmoset (Callithrix jacchus) is uniquely suited for longitudinal studies of cognitive aging, due to a relatively short lifespan, sophisticated cognitive abilities, and patterns of brain aging that resemble those of humans. We examined cognitive function and fine motor skills in male and female marmosets (mean age ∼5 at study entry) followed longitudinally for 2 years. Each year, monkeys were tested on a reversal learning task with three pairs of stimuli (n = 18, 9 females) and a fine motor task requiring them to grasp small rewards from two staircases (Hill and Valley test, n = 12, 6 females). There was little evidence for a decline in cognitive flexibility between the two time points, in part because of practice effects. However, independent of year of testing, females took longer than males to reach criterion in the reversals, indicating impaired cognitive flexibility. Motivation was unlikely to contribute to this effect, as males refused a greater percentage of trials than females in the reversals. With regards to motor function, females were significantly faster than males in the Hill and Valley task. From Year 1 to Year 2, a slight slowing of motor function was observed in both sexes, but accuracy decreased significantly in males only. This study (1) demonstrates that marmosets exhibit sex differences in cognitive flexibility and fine motor function that resemble those described in humans; (2) that changes in fine motor function can already be detected at middle-age; and (3) that males may experience greater age-related changes in fine motor skills than females. Additional data points will determine whether these sex and age differences persist over time.


Asunto(s)
Envejecimiento , Callithrix/fisiología , Cognición/fisiología , Destreza Motora/fisiología , Animales , Callithrix/psicología , Femenino , Masculino , Aprendizaje Inverso/fisiología , Factores Sexuales
9.
Front Neuroendocrinol ; 47: 134-153, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28803147

RESUMEN

Loss of ovarian function in women is associated with sleep disturbances and cognitive decline, which suggest a key role for estrogens and/or progestins in modulating these symptoms. The effects of ovarian hormones on sleep and cognitive processes have been studied in separate research fields that seldom intersect. However, sleep has a considerable impact on cognitive function. Given the tight connections between sleep and cognition, ovarian hormones may influence selective aspects of cognition indirectly, via the modulation of sleep. In support of this hypothesis, a growing body of evidence indicates that the development of sleep disorders following menopause contributes to accelerated cognitive decline and dementia in older women. This paper draws from both the animal and human literature to present an integrated view of the effects of ovarian hormones on sleep and cognition across the adult female lifespan.


Asunto(s)
Cognición/fisiología , Estrógenos/sangre , Progesterona/sangre , Sueño/fisiología , Animales , Femenino , Humanos
10.
Anim Cogn ; 19(3): 619-30, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26909674

RESUMEN

This study examined sex differences in executive function in middle-aged gonadectomized marmosets (Callithrix jacchus) with or without hormonal replacement. We tested ten castrated male (mean age 5.5 years) marmosets treated with testosterone cypionate (T, n = 5) or vehicle (n = 5) on Reversal Learning, which contributes to cognitive flexibility, and the Delayed Response task, measuring working memory. Their performance was compared to that of 11 ovariectomized females (mean age = 3.7 years) treated with Silastic capsules filled with 17-ß estradiol (E2, n = 6) or empty capsules (n = 5), previously tested on the same tasks (Lacreuse et al. in J Neuroendocrinol 26:296-309, 2014. doi: 10.1111/jne.12147). Behavioral observations were conducted daily. Females exhibited more locomotor behaviors than males. Males and females did not differ in the number of trials taken to reach criterion on the reversals, but males had significantly longer response latencies, regardless of hormone replacement. They also had a greater number of refusals than females. Additionally, both control and T-treated males, but not females, had slower responses on incorrect trials, suggesting that males were making errors due to distraction, lack of motivation or uncertainty. Furthermore, although both males and females had slower responding following an incorrect compared to a correct trial, the sex difference in response latencies was disproportionally large following an incorrect trial. No sex difference was found in the Delayed Response task. Overall, slower response latencies in males than females during Reversal Learning, especially during and following an incorrect trial, may reflect greater sensitivity to punishment (omission of reward) and greater performance monitoring in males, compared to females. Because these differences occurred in gonadectomized animals and regardless of hormone replacement, they may be organized early in life.


Asunto(s)
Conducta Animal/efectos de los fármacos , Callithrix/fisiología , Cognición/efectos de los fármacos , Estradiol/farmacología , Aprendizaje Inverso/efectos de los fármacos , Caracteres Sexuales , Testosterona/análogos & derivados , Andrógenos/farmacología , Animales , Estrógenos/farmacología , Femenino , Masculino , Orquiectomía , Ovariectomía , Castigo , Testosterona/farmacología
11.
Horm Behav ; 74: 157-66, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25762288

RESUMEN

This article is part of a Special Issue "Estradiol and cognition". This review discusses the unique contribution of nonhuman primate research to our understanding of the neurocognitive effects of estrogens throughout the adult lifespan in females. Mounting evidence indicates that estrogens affect many aspects of hippocampal, prefrontal and cholinergic function in the primate brain and the underlying mechanisms are beginning to be elucidated. In addition, estrogens may also influence cognitive function indirectly, via the modulation of other systems that impact cognition. We will focus on the effects of estrogens on sleep and emphasize the need for primate models to better understand these complex interactions. Continued research with nonhuman primates is essential for the development of therapies that are optimal for the maintenance of women's cognitive health throughout the lifespan.


Asunto(s)
Cognición/fisiología , Estrógenos/fisiología , Longevidad/fisiología , Primates/fisiología , Adulto , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Cognición/efectos de los fármacos , Estrógenos/farmacología , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Humanos , Longevidad/efectos de los fármacos , Primates/psicología , Factores Sexuales
12.
Exp Brain Res ; 233(3): 829-37, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25466868

RESUMEN

Intermanual transfer refers to an effect, whereby training one hand to perform a motor task improves performance in the opposite untrained hand. We tested the hypothesis that handedness facilitates intermanual transfer in two nonhuman primate species: rhesus monkeys (N = 13) and chimpanzees (N = 52). Subjects were grouped into one of four conditions: (1) left-handers trained with the left (dominant) hand; (2) left-handers trained with the right (nondominant) hand; (3) right-handers trained with the left (nondominant) hand; and (4) right-handers trained with the right (dominant) hand. Intermanual transfer was measured using a task where subjects removed a Life Savers(®) candy (monkeys) or a washer (chimpanzees) from metal shapes. Transfer was measured with latency by comparing the average time taken to solve the task in the first session with the trained hand compared to the first session with the untrained hand. Hypotheses and predictions were derived from three models of transfer: access: benefit training with nondominant hand; proficiency: benefit training with dominant hand; and cross-activation: benefit irrespective of trained hand. Intermanual transfer (i.e., shorter latency in untrained hand) occurred regardless of whether monkeys trained with the dominant hand or nondominant hand, supporting the cross-activation model. However, transfer was only observed in chimpanzees that trained with the dominant hand. When handedness groups were examined separately, the transfer effect was only significant for right-handed chimpanzees, partially supporting the proficiency model. Findings may be related to neurophysiological differences in motor control as well as differences in handedness patterning between rhesus monkeys and chimpanzees.


Asunto(s)
Lateralidad Funcional/fisiología , Mano/fisiología , Desempeño Psicomotor/fisiología , Animales , Femenino , Macaca mulatta , Masculino , Destreza Motora/fisiología , Pan troglodytes
13.
Horm Behav ; 66(5): 731-42, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25308086

RESUMEN

Human aging is characterized by declines in cognition and fine motor function as well as improved emotional regulation. In men, declining levels of testosterone (T) with age have been implicated in the development of these age-related changes. However, studies examining the effects of T replacement on cognition, emotion and fine motor function in older men have not provided consistent results. Rhesus monkeys (Macaca mulatta) are excellent models for human cognitive aging and may provide novel insights on this issue. We tested 10 aged intact male rhesus monkeys (mean age=19, range 15-25) on a battery of cognitive, motor and emotional tasks at baseline and under low or high T experimental conditions. Their performance was compared to that of 6 young males previously tested in the same paradigm (Lacreuse et al., 2009; Lacreuse et al., 2010). Following a 4-week baseline testing period, monkeys were treated with a gonadotropin releasing hormone agonist (Depot Lupron, 200 µg/kg) to suppress endogenous T and were tested on the task battery under a 4-week high T condition (injection of Lupron+T enanthate, 20 mg/kg, n=8) or 4-week low T condition (injection of Lupron+oil vehicle, n=8) before crossing over to the opposite treatment. The cognitive tasks consisted of the Delayed Non-Matching-to-Sample (DNMS), the Delayed Response (DR), and the Delayed Recognition Span Test (spatial-DRST). The emotional tasks included an object Approach-Avoidance task and a task in which monkeys were played videos of unfamiliar conspecifics in different emotional context (Social Playbacks). The fine motor task was the Lifesaver task that required monkeys to remove a Lifesaver candy from rods of different complexity. T manipulations did not significantly affect visual recognition memory, working memory, reference memory or fine motor function at any age. In the Approach-Avoidance task, older monkeys, but not younger monkeys, spent more time in proximity of novel objects in the high T condition relative to the low T condition. In both age groups, high T increased watching time of threatening social stimuli in the Social Playbacks. These results suggest that T affects some aspects of emotional processing but has no effect on fine motor function or cognition in young or older male macaques. It is possible that the duration of T treatment was not long enough to affect cognition or fine motor function or that T levels were too high to improve these outcomes. An alternative explanation for the discrepancies of our findings with some of the cognitive and emotional effects of T reported in rodents and humans may be the use of a chemical castration, which reduced circulating gonadotropins in addition to T. Further studies will investigate whether the luteinizing hormone LH mediates the effects of T on brain function in male primates.


Asunto(s)
Envejecimiento/psicología , Cognición/efectos de los fármacos , Emociones/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Testosterona/análogos & derivados , Adolescente , Anciano , Animales , Cognición/fisiología , Esquema de Medicación , Emociones/fisiología , Hormona Liberadora de Gonadotropina/agonistas , Humanos , Leuprolida/administración & dosificación , Macaca mulatta , Masculino , Memoria/efectos de los fármacos , Testosterona/administración & dosificación
14.
Anim Cogn ; 16(6): 861-71, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23463380

RESUMEN

We examined attentional biases for social and non-social emotional stimuli in young adult men and compared the results to those of male rhesus monkeys (Macaca mulatta) previously tested in a similar dot-probe task (King et al. in Psychoneuroendocrinology 37(3):396-409, 2012). Recognition memory for these stimuli was also analyzed in each species, using a recognition memory task in humans and a delayed non-matching-to-sample task in monkeys. We found that both humans and monkeys displayed a similar pattern of attentional biases toward threatening facial expressions of conspecifics. The bias was significant in monkeys and of marginal significance in humans. In addition, humans, but not monkeys, exhibited an attentional bias away from negative non-social images. Attentional biases for social and non-social threat differed significantly, with both species showing a pattern of vigilance toward negative social images and avoidance of negative non-social images. Positive stimuli did not elicit significant attentional biases for either species. In humans, emotional content facilitated the recognition of non-social images, but no effect of emotion was found for the recognition of social images. Recognition accuracy was not affected by emotion in monkeys, but response times were faster for negative relative to positive images. Altogether, these results suggest shared mechanisms of social attention in humans and monkeys, with both species showing a pattern of selective attention toward threatening faces of conspecifics. These data are consistent with the view that selective vigilance to social threat is the result of evolutionary constraints. Yet, selective attention to threat was weaker in humans than in monkeys, suggesting that regulatory mechanisms enable non-anxious humans to reduce sensitivity to social threat in this paradigm, likely through enhanced prefrontal control and reduced amygdala activation. In addition, the findings emphasize important differences in attentional biases to social versus non-social threat in both species. Differences in the impact of emotional stimuli on recognition memory between monkeys and humans will require further study, as methodological differences in the recognition tasks may have affected the results.


Asunto(s)
Atención , Emociones , Macaca mulatta/psicología , Memoria , Adolescente , Animales , Expresión Facial , Humanos , Masculino , Estimulación Luminosa , Reconocimiento en Psicología , Adulto Joven
15.
Neurobiol Aging ; 123: 49-62, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36638681

RESUMEN

The investigation of neurobiological and neuropathological changes that affect synaptic integrity and function with aging is key to understanding why the aging brain is vulnerable to Alzheimer's disease. We investigated the cellular characteristics in the cerebral cortex of behaviorally characterized marmosets, based on their trajectories of cognitive learning as they transitioned to old age. We found increased astrogliosis, increased phagocytic activity of microglial cells and differences in resting and reactive microglial cell phenotypes in cognitively impaired compared to nonimpaired marmosets. Differences in amyloid beta deposition were not related to cognitive trajectory. However, we found age-related changes in density and morphology of dendritic spines in pyramidal neurons of layer 3 in the dorsolateral prefrontal cortex and the CA1 field of the hippocampus between cohorts. Overall, our data suggest that an accelerated aging process, accompanied by neurodegeneration, that takes place in cognitively impaired aged marmosets and affects the plasticity of dendritic spines in cortical areas involved in cognition and points to mechanisms of neuronal vulnerability to aging.


Asunto(s)
Péptidos beta-Amiloides , Callithrix , Animales , Encéfalo , Neuronas , Envejecimiento/fisiología
16.
Neurobiol Aging ; 109: 88-99, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34700200

RESUMEN

Longitudinal studies are essential to understand healthy and pathological neurocognitive aging such as Alzheimer's Disease, but longitudinal designs are rare in both humans and non-human primate models of aging because of the difficulty of tracking cognitive change in long-lived primates. Common marmosets (Callithrix jacchus) are uniquely suited for aging studies due to their naturally short lifespan (10-12 years), sophisticated cognitive and social abilities and Alzheimer Disease-like neuropathology. We report the first longitudinal study of cognitive aging in marmosets (N = 28) as they transitioned from middle- (∼5 years) to old age (∼9 years). We characterized aging trajectories using reversal learning with different stimuli each year. Marmosets initially improved on cognitive performance due to practice, but worsened in the final year, suggesting the onset of age-related decline. Cognitive impairment emerged earlier in females than males and was more prominent for discrimination than for reversal learning. Sex differences in cognitive aging could not be explained by differences in motivation or motor abilities, which improved or remained stable across aging. Likewise, males and females did not differ in aging trajectories of overall behavior or reactivity to a social stressor, with the exception of a progressive decline in the initiation of social behavior in females. Patterns of cognitive aging were highly variable across marmosets of both sexes, suggesting the potential for pathological aging for some individuals. Future work will link individual cognitive trajectories to neuropathology in order to better understand the relationships between neuropathologic burden and vulnerability to age-related cognitive decline in each sex.


Asunto(s)
Envejecimiento/fisiología , Envejecimiento/psicología , Callithrix , Envejecimiento Cognitivo/fisiología , Caracteres Sexuales , Animales , Conducta Animal , Cognición , Femenino , Estudios Longitudinales , Masculino , Modelos Animales , Aprendizaje Inverso , Conducta Social , Factores de Tiempo
17.
Horm Behav ; 58(5): 854-63, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20804760

RESUMEN

Animal studies indicate that sex hormones have widespread effects on the brain, cognition and emotion, but findings in humans are inconsistent. Well-controlled studies in nonhuman primates are crucial to resolve these discrepancies. In this study, we examined the effects of testosterone (T) on emotion in male rhesus monkeys. Six young adult males were tested on two emotional tasks during three hormonal conditions in a crossover design: when intact at baseline and when pharmacologically hypogonadal with add-back of T or placebo. The emotional tasks were the Approach-Avoidance task, which tested behavioral responses to three categories of objects (familiar, novel, and negative) and a Social Playback task which tested behavioral responses to scenes of unfamiliar conspecifics engaged in three types of social activities (neutral, positive, or negative). Following a 4-week baseline period, monkeys were treated with Depot Lupron, 200µg/kg before being randomly assigned to one of two treatment groups: Depot Lupron+Testosterone Enanthate (TE, 20mg/kg) or Depot Lupron+oil vehicle. In each treatment group, monkeys received one injection of Lupron and one injection of TE or one injection of Lupron and one injection of oil at the onset of a 4-week testing period, before crossing over to the alternate treatment for an additional 4weeks of testing. TE treatment had no effect on behavioral measures in the Approach-Avoidance task. For the Social Playback task, however, TE significantly increased watching time of video clips which depicted fights between unfamiliar conspecifics. The enhancing effect of T on watching time for negative social scenes is consistent with human data suggesting that T decreases aversion or facilitates approach to threatening social stimuli. Further studies are needed to understand the mechanisms by which T may mediate responsiveness to social threat in male primates.


Asunto(s)
Atención/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Miedo/efectos de los fármacos , Macaca mulatta/fisiología , Testosterona/farmacología , Algoritmos , Animales , Atención/fisiología , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Conducta Animal/fisiología , Conducta de Elección/efectos de los fármacos , Conducta de Elección/fisiología , Emociones/efectos de los fármacos , Emociones/fisiología , Miedo/fisiología , Hormona Liberadora de Gonadotropina/agonistas , Leuprolida/administración & dosificación , Leuprolida/farmacología , Masculino , Distribución Aleatoria , Conducta Social , Testosterona/administración & dosificación
18.
Philos Trans R Soc Lond B Biol Sci ; 375(1811): 20190618, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-32951543

RESUMEN

Executive function (EF) is a complex construct that reflects multiple higher-order cognitive processes such as planning, updating, inhibiting and set-shifting. Decline in these functions is a hallmark of cognitive ageing in humans, and age differences and changes in EF correlate with age-related differences and changes in association cortices, particularly the prefrontal areas. Here, we review evidence for age-related decline in EF and associated neurobiological changes in prosimians, New World and Old World monkeys, apes and humans. While EF declines with age in all primate species studied, the relationship of this decline with age-related alterations in the prefrontal cortex remains unclear, owing to the scarcity of neurobiological studies focusing on the ageing brain in most primate species. In addition, the influence of sex, vascular and metabolic risk, and hormonal status has rarely been considered. We outline several methodological limitations and challenges with the goal of producing a comprehensive integration of cognitive and neurobiological data across species and elucidating how ageing shapes neurocognitive trajectories in primates with different life histories, lifespans and brain architectures. Such comparative investigations are critical for fostering translational research and understanding healthy and pathological ageing in our own species. This article is part of the theme issue 'Evolution of the primate ageing process'.


Asunto(s)
Envejecimiento , Cognición/fisiología , Función Ejecutiva , Primates/fisiología , Animales , Femenino , Masculino , Neurobiología
19.
Sci Rep ; 10(1): 16647, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-33024242

RESUMEN

Nonhuman primates (NHPs) are an essential research model for gaining a comprehensive understanding of the neural mechanisms of neurocognitive aging in our own species. In the present study, we used resting state functional connectivity (rsFC) to investigate the relationship between prefrontal cortical and striatal neural interactions, and cognitive flexibility, in unanaesthetized common marmosets (Callithrix jacchus) at two time points during late middle age (8 months apart, similar to a span of 5-6 years in humans). Based on our previous findings, we also determine the reproducibility of connectivity measures over the course of 8 months, particularly previously observed sex differences in rsFC. Male marmosets exhibited remarkably similar patterns of stronger functional connectivity relative to females and greater cognitive flexibility between the two imaging time points. Network analysis revealed that the consistent sex differences in connectivity and related cognitive associations were characterized by greater node strength and/or degree values in several prefrontal, premotor and temporal regions, as well as stronger intra PFC connectivity, in males compared to females. The current study supports the existence of robust sex differences in prefrontal and striatal resting state networks that may contribute to differences in cognitive function and offers insight on the neural systems that may be compromised in cognitive aging and age-related conditions such as mild cognitive impairment and Alzheimer's disease.


Asunto(s)
Envejecimiento/psicología , Callithrix/psicología , Cognición/fisiología , Cuerpo Estriado/fisiología , Vías Nerviosas/fisiología , Corteza Prefrontal/fisiología , Caracteres Sexuales , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/psicología , Animales , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/psicología , Cuerpo Estriado/inmunología , Femenino , Masculino
20.
Physiol Behav ; 96(3): 448-56, 2009 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-19101578

RESUMEN

Many studies in women and animal models suggest that estrogens affect cognitive function. Yet, the mechanisms by which estrogens may impact cognition remain unclear. The goal of the present study was to assess the effects of different estrogen receptor (ER) ligands on cognitive function in adult ovariectomized female rhesus monkeys. The monkeys were tested for 6 weeks on a battery of memory and attentional tasks administered on a touchscreen: the object, face, and spatial versions of the Delayed Recognition Span Test (DRST) and a Visual Search task. Following a 2-week baseline period with oil vehicle treatment, monkeys were randomly assigned to one of 3 treatment groups: estradiol benzoate (EB), selective ERbeta agonist (diarylpropionitrile DPN) or selective ER modulator tamoxifen (TAM). In each treatment group, monkeys received oil vehicle for 2 weeks and the drug for 2 weeks, in a cross-over design. After a 4-week washout, a subset of monkeys was re-tested on the battery when treated with a selective ERalpha agonist (propyl-pyrazole-triol, PPT) or oil vehicle. Overall, drug treatments had no or negligible effects on cognitive performance. These results support the contention that exogenous estrogens and selective estrogen receptor modulators (SERMS) do not significantly affect cognition in young adult female macaques. Additional studies are needed to determine whether the cognitive effects of estrogens in monkeys of more advanced age are mediated by ERbeta, ERalpha or complex interactions between the two receptors.


Asunto(s)
Cognición/fisiología , Aprendizaje Discriminativo/fisiología , Moduladores de los Receptores de Estrógeno/farmacología , Estrógenos/fisiología , Reconocimiento en Psicología/fisiología , Factores de Edad , Animales , Atención/efectos de los fármacos , Atención/fisiología , Cognición/efectos de los fármacos , Estradiol/farmacología , Estradiol/fisiología , Receptor alfa de Estrógeno/efectos de los fármacos , Receptor alfa de Estrógeno/fisiología , Receptor beta de Estrógeno/efectos de los fármacos , Receptor beta de Estrógeno/fisiología , Estrógenos/farmacología , Femenino , Macaca mulatta , Nitrilos/farmacología , Ovariectomía , Fenoles , Propionatos/farmacología , Pirazoles/farmacología , Distribución Aleatoria , Reconocimiento en Psicología/efectos de los fármacos , Tamoxifeno/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA