Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Carmen Chow is active.

Publication


Featured researches published by Carmen Chow.


Journal of Neuroendocrinology | 2013

Sex, hormones and neurogenesis in the hippocampus: hormonal modulation of neurogenesis and potential functional implications.

Liisa A.M. Galea; Steven R. Wainwright; Meighen M. Roes; Paula Duarte-Guterman; Carmen Chow; Dwayne K. Hamson

The hippocampus is an area of the brain that undergoes dramatic plasticity in response to experience and hormone exposure. The hippocampus retains the ability to produce new neurones in most mammalian species and is a structure that is targeted in a number of neurodegenerative and neuropsychiatric diseases, many of which are influenced by both sex and sex hormone exposure. Intriguingly, gonadal and adrenal hormones affect the structure and function of the hippocampus differently in males and females. Adult neurogenesis in the hippocampus is regulated by both gonadal and adrenal hormones in a sex‐ and experience‐dependent way. Sex differences in the effects of steroid hormones to modulate hippocampal plasticity should not be completely unexpected because the physiology of males and females is different, with the most notable difference being that females gestate and nurse the offspring. Furthermore, reproductive experience (i.e. pregnancy and mothering) results in permanent changes to the maternal brain, including the hippocampus. This review outlines the ability of gonadal and stress hormones to modulate multiple aspects of neurogenesis (cell proliferation and cell survival) in both male and female rodents. The function of adult neurogenesis in the hippocampus is linked to spatial memory and depression, and the present review provides early evidence of the functional links between the hormonal modulation of neurogenesis that may contribute to the regulation of cognition and stress.


Frontiers in Neuroscience | 2013

Hippocampus-dependent learning influences hippocampal neurogenesis

Jonathan R. Epp; Carmen Chow; Liisa A.M. Galea

The structure of the mammalian hippocampus continues to be modified throughout life by continuous addition of neurons in the dentate gyrus. Although the existence of adult neurogenesis is now widely accepted the function that adult generated granule cells play is a topic of intense debate. Many studies have argued that adult generated neurons, due to unique physiological characteristics, play a unique role in hippocampus-dependent learning and memory. However, it is not currently clear whether this is the case or what specific capability adult generated neurons may confer that developmentally generated neurons do not. These questions have been addressed in numerous ways, from examining the effects of increasing or decreasing neurogenesis to computational modeling. One particular area of research has examined the effects of hippocampus dependent learning on proliferation, survival, integration and activation of immature neurons in response to memory retrieval. Within this subfield there remains a range of data showing that hippocampus dependent learning may increase, decrease or alternatively may not alter these components of neurogenesis in the hippocampus. Determining how and when hippocampus-dependent learning alters adult neurogenesis will help to further clarify the role of adult generated neurons. There are many variables (such as age of immature neurons, species, strain, sex, stress, task difficulty, and type of learning) as well as numerous methodological differences (such as marker type, quantification techniques, apparatus size etc.) that could all be crucial for a clear understanding of the interaction between learning and neurogenesis. Here, we review these findings and discuss the different conditions under which hippocampus-dependent learning impacts adult neurogenesis in the dentate gyrus.


Psychoneuroendocrinology | 2013

Sex differences in neurogenesis and activation of new neurons in response to spatial learning and memory.

Carmen Chow; Jonathan R. Epp; Stephanie E. Lieblich; Cindy K. Barha; Liisa A.M. Galea

Adult hippocampal neurogenesis is often associated with hippocampus-dependent learning and memory. Throughout a new neurons development, it is differentially sensitive to factors that can influence its survival and functionality. Previous research shows that spatial training that occurred 6-10 days after an injection of the DNA synthesis marker, bromodeoxyuridine (BrdU), increased cell survival in male rats. Because sex differences in spatial cognition and hippocampal neurogenesis have been reported, it is unclear whether spatial training would influence hippocampal neurogenesis in the same way in males and females. Therefore, this study examined sex differences in hippocampal neurogenesis following training in a spatial task. Male and female rats were trained in the spatial or cued version of the Morris water maze 6-10 days after one injection of BrdU (200mg/kg). Twenty days following BrdU injection, all animals were given a probe trial and perfused. Males performed better in the spatial, but not cue, task than females. Spatial training increased BrdU-labeled cells relative to cue training only in males, but both males and females showed greater activation of new cells (BrdU co-labeled with immediate early gene product zif268) after spatial training compared to cue training. Furthermore, performance during spatial training was positively correlated with cell activation in females but not males. This study shows that while spatial training differentially regulates hippocampal neurogenesis in males and females, the activity of new neurons in response to spatial memory retrieval is similar. These findings highlight the importance of sex on neural plasticity and cognition.


Journal of Neuroendocrinology | 2014

Effects of Chronic Oestradiol, Progesterone and Medroxyprogesterone Acetate on Hippocampal Neurogenesis and Adrenal Mass in Adult Female Rats

Melissa Y.T. Chan; Carmen Chow; Dwayne K. Hamson; Stephanie E. Lieblich; Liisa A.M. Galea

Both natural oestrogens and progesterone influence synaptic plasticity and neurogenesis within the female hippocampus. However, less is known of the impact of synthetic hormones on hippocampal structure and function. There is some evidence that the administration of the synthetic progestin, medroxyprogesterone acetate (MPA) is not as beneficial as natural progesterone and can attenuate oestrogen‐induced neuroprotection. Although the effects of oestradiol have been well studied, little is known about the effects of natural and synthetic progestins alone and in combination with oestradiol on adult neurogenesis in females. In the present study, we investigated the effects of chronic oestradiol, progesterone, MPA and the co‐administration of each progestin with oestradiol on neurogenesis within the dentate gyrus of adult ovariectomised female rats. Twenty‐four hours after a bromodeoxyuridine (BrdU; 200 mg/kg) injection, female rats were repeatedly administered either progesterone (1 or 4 mg), MPA (1 or 4 mg), oestradiol benzoate (EB), progesterone or MPA in combination with EB (10 μg), or vehicle for 21 days. Rats were perfused on day 22 and brain tissue was analysed for the number of BrdU‐labelled and Ki67 (an endogenous marker of cell proliferation)‐expressing cells. EB alone and MPA + EB significantly decreased neurogenesis and the number of surviving BrdU‐labelled cells in the dorsal region of the dentate gyrus, independent of any effects on cell proliferation. Furthermore, MPA (1 and 4 mg) and MPA + EB treated animals had significantly lower adrenal/body mass ratios and reduced serum corticosterone (CORT) levels. By contrast, progesterone + EB treated animals had significantly higher adrenal/body mass ratios and 1 mg of progesterone, progesterone + EB, and EB significantly increased CORT levels. The results of the present study demonstrate that different progestins alone and in combination with oestradiol can differentially affect neurogenesis (via cell survival) and regulation of the hypothalamic‐pituitary‐adrenal axis. These findings have implications for women using hormone replacement therapies with MPA for both neuroprotection and stress‐related disorders.


Hippocampus | 2016

Sex and strategy use matters for pattern separation, adult neurogenesis, and immediate early gene expression in the hippocampus

Shunya Yagi; Carmen Chow; Stephanie E. Lieblich; Liisa A.M. Galea

Adult neurogenesis in the dentate gyrus (DG) plays a crucial role for pattern separation, and there are sex differences in the regulation of neurogenesis. Although sex differences, favoring males, in spatial navigation have been reported, it is not known whether there are sex differences in pattern separation. The current study was designed to determine whether there are sex differences in the ability for separating similar or distinct patterns, learning strategy choice, adult neurogenesis, and immediate early gene (IEG) expression in the DG in response to pattern separation training. Male and female Sprague‐Dawley rats received a single injection of the DNA synthesis marker, bromodeoxyuridine (BrdU), and were tested for the ability of separating spatial patterns in a spatial pattern separation version of delayed nonmatching to place task using the eight‐arm radial arm maze. Twenty‐seven days following BrdU injection, rats received a probe trial to determine whether they were idiothetic or spatial strategy users. We found that male spatial strategy users outperformed female spatial strategy users only when separating similar, but not distinct, patterns. Furthermore, male spatial strategy users had greater neurogenesis in response to pattern separation training than all other groups. Interestingly, neurogenesis was positively correlated with performance on similar pattern trials during pattern separation in female spatial strategy users but negatively correlated with performance in male idiothetic strategy users. These results suggest that the survival of new neurons may play an important positive role for pattern separation of similar patterns in females. Furthermore, we found sex and strategy differences in IEG expression in the CA1 and CA3 regions in response to pattern separation. These findings emphasize the importance of studying biological sex on hippocampal function and neural plasticity.


Neurobiology of Aging | 2015

Multiparity-induced enhancement of hippocampal neurogenesis and spatial memory depends on ovarian hormone status in middle age

Cindy K. Barha; Stephanie E. Lieblich; Carmen Chow; Liisa A.M. Galea

Menopause is associated with cognitive decline, and previous parity can increase or delay the trajectory of cognitive aging. Furthermore, parity enables the hippocampus to respond to estrogens in middle age. The present study investigated how previous parity and estrogens influence cognition, neurogenesis, and neuronal activation in response to memory retrieval in the hippocampus of middle-aged females. Multiparous and nulliparous rats were ovariectomized (OVX) or received sham surgery and were treated with vehicle, 17β-estradiol, 17α-estradiol, or estrone. Rats were trained on the spatial working and reference memory versions of the Morris water maze. Multiparous rats had a significantly greater density of immature neurons in the hippocampus, enhanced acquisition of working memory, but poorer reference memory compared with nulliparous rats. Furthermore, OVX increased, while treatment with estrogens reduced, the density of immature neurons, regardless of parity. OVX improved reference memory only in nulliparous rats. Thus, motherhood has long-lasting effects on the neuroplasticity and function of the hippocampus. These findings have wide-ranging implications for the treatment of age-associated decline in women.


Neuropharmacology | 2016

Maternal postpartum corticosterone and fluoxetine differentially affect adult male and female offspring on anxiety-like behavior, stress reactivity, and hippocampal neurogenesis.

Aarthi R. Gobinath; Joanna L. Workman; Carmen Chow; Stephanie E. Lieblich; Liisa A.M. Galea

Postpartum depression (PPD) affects approximately 15% of mothers, disrupts maternal care, and can represent a form of early life adversity for the developing offspring. Intriguingly, male and female offspring are differentially vulnerable to the effects of PPD. Antidepressants, such as fluoxetine, are commonly prescribed for treating PPD. However, fluoxetine can reach offspring via breast milk, raising serious concerns regarding the long-term consequences of infant exposure to fluoxetine. The goal of this study was to examine the long-term effects of maternal postpartum corticosterone (CORT, a model of postpartum stress/depression) and concurrent maternal postpartum fluoxetine on behavioral, endocrine, and neural measures in adult male and female offspring. Female Sprague-Dawley dams were treated daily with either CORT or oil and fluoxetine or saline from postnatal days 2-23, and offspring were weaned and left undisturbed until adulthood. Here we show that maternal postpartum fluoxetine increased anxiety-like behavior and impaired hypothalamic-pituitary-adrenal (HPA) axis negative feedback in adult male, but not female, offspring. Furthermore, maternal postpartum fluoxetine increased the density of immature neurons (doublecortin-expressing) in the hippocampus of adult male offspring but decreased the density of immature neurons in adult female offspring. Maternal postpartum CORT blunted HPA axis negative feedback in males and tended to increase density of immature neurons in males but decreased it in females. These results indicate that maternal postpartum CORT and fluoxetine can have long-lasting effects on anxiety-like behavior, HPA axis negative feedback, and adult hippocampal neurogenesis and that adult male and female offspring are differentially affected by these maternal manipulations.


PLOS ONE | 2015

Estradiol and GPER Activation Differentially Affect Cell Proliferation but Not GPER Expression in the Hippocampus of Adult Female Rats.

Paula Duarte-Guterman; Stephanie E. Lieblich; Carmen Chow; Liisa A.M. Galea

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: 17β-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.


Hormones and Behavior | 2016

Testosterone has antidepressant-like efficacy and facilitates imipramine-induced neuroplasticity in male rats exposed to chronic unpredictable stress

Steven R. Wainwright; Joanna L. Workman; Amir Tehrani; Dwayne K. Hamson; Carmen Chow; Stephanie E. Lieblich; Liisa A.M. Galea

Hypogonadal men are more likely to develop depression, while testosterone supplementation shows antidepressant-like effects in hypogonadal men and facilitates antidepressant efficacy. Depression is associated with hypothalamic-pituitary-adrenal (HPA) axis hyperactivity and testosterone exerts suppressive effects on the HPA axis. The hippocampus also plays a role in the feedback regulation of the HPA axis, and depressed patients show reduced hippocampal neuroplasticity. We assessed the antidepressant-like effects of testosterone with, or without, imipramine on behavioral and neural endophenotypes of depression in a chronic unpredictable stress (CUS) model of depression. A 21-day CUS protocol was used on gonadectomized male Sprague-Dawley rats treated with vehicle, 1mg of testosterone propionate, 10mg/kg of imipramine, or testosterone and imipramine in tandem. Testosterone treatment reduced novelty-induced hypophagia following CUS exposure, but not under non-stress conditions, representing state-dependent effects. Further, testosterone increased the latency to immobility in the forced swim test (FST), reduced basal corticosterone, and reduced adrenal mass in CUS-exposed rats. Testosterone also facilitated the effects of imipramine by reducing the latency to immobility in the FST and increasing sucrose preference. Testosterone treatment had no significant effect on neurogenesis, though the combination of testosterone and imipramine increased PSA-NCAM expression in the ventral dentate gyrus. These findings demonstrate the antidepressant- and anxiolytic-like effects of testosterone within a CUS model of depression, and provide insight into the mechanism of action, which appears to be independent of enhanced hippocampal neurogenesis.


Neuropharmacology | 2018

Voluntary running influences the efficacy of fluoxetine in a model of postpartum depression

Aarthi R. Gobinath; Robin Richardson; Carmen Chow; Joanna L. Workman; Stephanie E. Lieblich; Alasdair M. Barr; Liisa A.M. Galea

ABSTRACT Postpartum depression affects approximately 15% of mothers. Unfortunately, treatment options for postpartum depression are limited. Pharmacological antidepressants such as fluoxetine (FLX) can be controversial due to inconclusive evidence of efficacy during the postpartum and concerns of neonatal exposure to antidepressants. Alternatively, non‐pharmacological antidepressants such as exercise may be less controversial but its efficacy in postpartum depression is unclear. To investigate this, we treated rat dams daily with high levels of corticosterone (CORT; 40 mg/kg), to induce a depressive‐like phenotype, or oil (vehicle for CORT) during the postpartum period. Within the oil and CORT conditions, four additional antidepressant conditions were created: 1. FLX (10 mg/kg) + exercise (voluntary access to running wheel); 2. FLX + no exercise; 3. Saline (vehicle for FLX) + exercise; 4. Saline + No exercise. We examined maternal care, depressive‐like and anxiety‐like behavior, stress reactivity, and hippocampal neurogenesis and dams were categorized as “high running” or “low running.” FLX treatment, alone or with high running, prevented CORT‐induced disruptions in maternal care. As expected, CORT increased depressive‐like behavior but exercise, regardless of running amount, reduced depressive‐like behavior. Intriguingly, FLX, but not CORT, increased anxiety‐like behavior, which was not mitigated by concurrent exercise. FLX treatment slightly but significantly facilitated serum CORT recovery after forced swim stress. CORT and FLX alone reduced neurogenesis, while exercise coupled with FLX increased density of doublecortin‐expressing cells. High running increased density of doublecortin‐expressing cells (immature neurons) in comparison to controls. Collectively, these findings indicate that FLX and exercise reverse different endophenotypes of depression in dams, which has translational implications for surveying treatment options of postpartum depression. HIGHLIGHTSPostpartum CORT (corticosterone) reduced maternal care and hippocampal neurogenesis.Postpartum fluoxetine (FLX) prevented CORT‐induced reductions in maternal care.Exercise, but not FLX, reduced CORT‐induced maternal depressive‐like behavior.FLX increased neurogenesis in dams only in combination with exercise.

Collaboration


Dive into the Carmen Chow's collaboration.

Top Co-Authors

Avatar

Liisa A.M. Galea

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Stephanie E. Lieblich

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Aarthi R. Gobinath

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Paula Duarte-Guterman

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Cindy K. Barha

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Dwayne K. Hamson

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Joanna L. Workman

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Jonathan R. Epp

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Shunya Yagi

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Steven R. Wainwright

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge