Network


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

Hotspot


Dive into the research topics where Rachel Anne Hill is active.

Publication


Featured researches published by Rachel Anne Hill.


Biological Psychiatry | 2007

Estrogen deficient male mice develop compulsive behavior

Rachel Anne Hill; Kerry J. McInnes; Emily C.H. Gong; Margaret E. E. Jones; Evan R. Simpson; Wah Chin Boon

BACKGROUND Aromatase converts androgen to estrogen. Thus, the aromatase knockout (ArKO) mouse is estrogen deficient. We investigated the compulsive behaviors of these animals and the protein levels of catechol-O-methyltransferase (COMT) in frontal cortex, hypothalamus and liver. METHODS Grooming was analyzed during the 20-min period immediately following a water-mist spray. Running wheel activity over two consecutive nights and barbering were analyzed. COMT protein levels were measured by Western analysis. RESULTS Six-month old male but not female ArKO mice develop compulsive behaviors such as excessive barbering, grooming and wheel-running. Excessive activities were reversed by 3 weeks of 17beta-estradiol replacement. Interestingly, the presentation of compulsive behaviors is accompanied by concomitant decreases (p < .05) in hypothalamic COMT protein levels in male ArKO mice. These values returned to normal upon 17beta-estradiol treatment. In contrast, hepatic and frontal cortex COMT levels were not affected by the estrogen status, indicating region- and tissue-specific regulation of COMT levels by estrogen. No differences in COMT levels were detectable between female animals of both genotypes. CONCLUSIONS This study describes the novel observation of a possible link between estrogen, COMT and development of compulsive behaviors in male animals which may have therapeutic implications in obsessive compulsive disorder (OCD) patients.


Molecular Psychiatry | 2015

The BDNF gene Val66Met polymorphism as a modifier of psychiatric disorder susceptibility: progress and controversy

Rachel Anne Hill; M. van den Buuse

Brain-derived neurotrophic factor (BDNF) has a primary role in neuronal development, differentiation and plasticity in both the developing and adult brain. A single-nucleotide polymorphism in the proregion of BDNF, termed the Val66Met polymorphism, results in deficient subcellular translocation and activity-dependent secretion of BDNF, and has been associated with impaired neurocognitive function in healthy adults and in the incidence and clinical features of several psychiatric disorders. Research investigating the Val66Met polymorphism has increased markedly in the past decade, and a gap in integration exists between and within academic subfields interested in the effects of this variant. Here we comprehensively review the role and relevance of the Val66Met polymorphism in psychiatric disorders, with emphasis on suicidal behavior and anxiety, eating, mood and psychotic disorders. The cognitive and molecular neuroscience of the Val66Met polymorphism is also concisely reviewed to illustrate the effects of this genetic variant in healthy controls, and is complemented by a commentary on the behavioral neuroscience of BDNF and the Val66Met polymorphism where relevant to specific disorders. Lastly, a number of controversies and unresolved issues, including small effect sizes, sampling of allele inheritance but not genotype and putative ethnicity-specific effects of the Val66Met polymorphism, are also discussed to direct future research.


Neuroscience & Biobehavioral Reviews | 2015

A role for the BDNF gene Val66Met polymorphism in schizophrenia? A comprehensive review

Rachel Anne Hill; Maarten van den Buuse

Schizophrenia is believed to arise from complex gene-environment interactions. Brain-derived neurotrophic factor (BDNF) is involved in neuronal development, differentiation and plasticity. A functional single nucleotide polymorphism that results in a valine (Val) to methionine (Met) substitution at codon 66 (Val66Met) results in the aberrant sorting and release of mature BDNF through the activity-dependent secretion pathway. The Val66Met polymorphism has been linked to impaired neurocognitive function in healthy adults, and identified as a locus of risk for a range of neuropsychiatric disorders including schizophrenia. Here we provide a comprehensive review of the relationship between the BDNF Val66Met polymorphism and schizophrenia, integrating evidence from the fields of genetic epidemiology, clinical psychiatry, behavioral neuroscience and neuroimaging. We argue that while the Val66Met polymorphism may not be a major risk-conferring agent for the development of schizophrenia per se, there is mounting evidence that the polymorphism modulates a range of clinical features of the illness, including age of onset, symptoms, therapeutic responsiveness, neurocognitive function and brain morphology.


Molecular and Cellular Neuroscience | 2004

Estrogen deficiency leads to apoptosis in dopaminergic neurons in the medial preoptic area and arcuate nucleus of male mice.

Rachel Anne Hill; Sueli Pompolo; Margaret E. E. Jones; Evan R. Simpson; Wah Chin Boon

The aromatase knockout (ArKO) mouse is unable to synthesize estrogens. Immunohistochemical studies on active caspase-3 and tyrosine hydroxylase (TH) revealed apoptosis of dopaminergic neurons in the medial preoptic area (MPO) and arcuate nucleus (Arc) of the hypothalamus of 1-year-old (1yo) male ArKO mice while no active caspase-3 was detected in wild type (WT). Furthermore, the number of TH-positive cells in the MPO and caudal Arc was significantly decreased in 1yo ArKO compared to WT. RNase protection assays support the presence of apoptosis in 1yo ArKO hypothalamus, revealing an up-regulation of pro-apoptotic genes: FASL, FADD, and caspase-8. Concomitantly, the ratio of bcl-2-related anti-apoptotic genes to pro-apoptotic genes in the hypothalamus of 1yo ArKO mice was significantly down-regulated. Previously, we have reported that no such changes were observed in the hypothalamus of female ArKO mice. Thus, we have provided direct evidence that estrogen is required to maintain the survival and functional integrity of dopaminergic neurons in the MPO and Arc of male, but not female mice.


Journal of Neuroendocrinology | 2012

Modulatory Effects of Sex Steroid Hormones on Brain-Derived Neurotrophic Factor-Tyrosine Kinase B Expression during Adolescent Development in C57Bl/6 Mice

Rachel Anne Hill; YeeWen Candace Wu; Perrin Kwek; M. van den Buuse

Sex steroid hormones and neurotrophic factors are involved in pruning and shaping the adolescent brain and have been implicated in the pathogenesis of neurodevelopmental disorders, including mental illness. We aimed to determine the association between altered levels of sex steroid hormones during adolescent development and neurotrophic signalling in the C57Bl/6 mouse. We first performed a week by week analysis from pre‐pubescence to adulthood in male and female C57Bl/6 mice, measuring serum levels of testosterone and oestradiol in conjunction with western blot analysis of neurotrophin expression in the forebrain and hippocampal regions. Second, we manipulated adolescent sex steroid hormone levels by gonadectomy and hormone replacement at the pre‐pubescent age of 5 weeks. Young‐adult forebrain and hippocampal neurotrophin expression was then determined. Male mice showed significant changes in brain‐derived neurotrophic factor (BDNF) expression in the forebrain regions during weeks 7–10, which corresponded significantly with a surge in serum testosterone. Castration and testosterone or di‐hydrotestosterone replacement experiments revealed an androgen receptor‐dependent effect on BDNF‐tyrosine kinase (Trk) B signalling in the forebrain and hippocampal regions during adolescence. Female mice showed changes in BDNF‐TrkB signalling at a much earlier time point (weeks 4–8) in the forebrain and hippocampal regions and these did not correspond with changes in serum oestradiol. Ovariectomy actually increased BDNF expression but decreased TrkB phosphorylation in the forebrain regions. 17β‐Oestradiol replacement had no effect, suggesting a role for other ovarian hormones in regulating BDNF‐TrkB signalling in the adolescent female mouse brain. These results suggest the differential actions of sex steroid hormones in modulating BDNF‐TrkB signalling during adolescence. These data provide insight into how the male and female brain changes in response to altered levels of circulating sex steroid hormones and could help to explain some of the developmental sex differences in the pathogenesis of neurodevelopmental disorders, including mental illness.


Neuroscience | 2013

Sex differences and the role of estrogen in animal models of schizophrenia: interaction with BDNF

YeeWen Candace Wu; Rachel Anne Hill; Andrea Gogos; M. van den Buuse

Schizophrenia is a severe psychiatric disorder with a complex and variable set of symptoms. Both genetic and environmental mechanisms are involved in the development of the illness and lead to structural and neurochemical abnormalities in the brain. An intriguing facet of schizophrenia is sex differences, which have been described for nearly all features of the illness, including the peak age of onset, symptoms and treatment response. The ovarian hormone, estrogen, may be protective against schizophrenia and evidence is accumulating that estrogen may exert this effect via an interaction with brain-derived neurotrophic factor (BDNF). Both estrogen and BDNF have trophic effects on the developing brain and promote synaptic plasticity and maintain neurons well into adulthood. Major neurotransmitter systems including dopaminergic, serotonergic and glutamatergic pathways are modulated and supported by estrogen and BDNF. Despite their commonalities, estrogen and BDNF have mostly been examined independently but increasing evidence suggests an interaction between the two in brain regions pertinent to schizophrenia. This review will focus on the role of estrogen and BDNF in clinical and animal studies of schizophrenia. We include animal models of neurotransmitter dysfunction and genetic manipulation to show how estrogen may provide a protective effect in schizophrenia, including through mediating BDNF expression and activity. This posited estrogen-BDNF interaction could play a key role in modulating sex-dependent results reported in animal work as well as sex differences in clinical aspects of schizophrenia.


Hippocampus | 2014

Sex-Specific Disruptions in Spatial Memory and Anhedonia in a "Two Hit" Rat Model Correspond With Alterations in Hippocampal Brain-Derived Neurotrophic Factor Expression and Signaling

Rachel Anne Hill; Maren Klug; Szerenke Kiss Von Soly; Michele D. Binder; Anthony J. Hannan; Maarten van den Buuse

Post‐mortem studies have demonstrated reduced expression of brain‐derived neurotrophic factor (BDNF) in the hippocampus of schizophrenia and major depression patients. The “two hit” hypothesis proposes that two or more major disruptions at specific time points during development are involved in the pathophysiology of these mental illnesses. However, the role of BDNF in these “two hit” effects is unclear. Our aim was to behaviorally characterize a “two hit” rat model of developmental stress accompanied by an in‐depth assessment of BDNF expression and signalling. Wistar rats were exposed to neonatal maternal separation (MS) stress and/or adolescent/young‐adult corticosterone (CORT) treatment. In adulthood, models of cognitive and negative symptoms of mental illness were analyzed. The hippocampus was then dissected into dorsal (DHP) and ventral (VHP) regions and analyzed by qPCR for exon‐specific BDNF gene expression or by Western blot for BDNF protein expression and downstream signaling. Male “two hit” rats showed marked disruptions in short‐term spatial memory (Y‐maze) which were absent in females. However, female “two hit” rats showed signs of anhedonia (sucrose preference test), which were absent in males. Novel object recognition and anxiety (elevated plus maze) were unchanged by either of the two “hits”. In the DHP, MS caused a male‐specific increase in BDNF Exons I, II, IV, VII, and IX mRNA but a decrease in mature BDNF and phosphorylated TrkB (pTrkB) protein expression in adulthood. In the VHP, BDNF transcript expression was unchanged; however, in female rats only, MS significantly decreased mature BDNF and pTrkB protein expression in adulthood. These data demonstrate that MS causes region‐specific and sex‐specific long‐term effects on BDNF expression and signaling and, importantly, mRNA expression does not always infer protein expression. Alterations to BDNF signaling may mediate the sex‐specific effects of developmental stress on anhedonic behaviors.


Brain Research | 2011

Sex-dependent and region-specific changes in TrkB signaling in BDNF heterozygous mice

Rachel Anne Hill; Maarten van den Buuse

Altered expression of neurotrophins may contribute to the pathogenesis of schizophrenia. Several studies suggest sex steroid hormones may be involved in the regulation of brain-derived neurotrophic factor (BDNF) signaling, as well as the symptoms of schizophrenia. This study aimed to identify sex differences in BDNF-TrkB signaling in the forebrain of wild type (WT) and BDNF heterozygous (+/-) mice. Protein expression of neurotrophins and TrkB were measured by Western blot analysis in brain regions pertinent to schizophrenia, namely the frontal cortex, striatum, and dorsal (DHP) and ventral hippocampus (VHP). In both the frontal cortex and striatum, protein expression levels of phosphorylated TrkB (pTrkB) over total TrkB (pTrkB/TrkB) was significantly increased in male, but not female BDNF(+/-) mice, suggesting sex-specific changes in TrkB signaling. pTrkB/TrkB levels were also elevated in the DHP of both male and female BDNF(+/-), while levels remained unchanged in the VHP, indicating region-specific changes in TrkB signaling. Sex-specific phosphorylation of TrkB corresponded with downstream changes in ERK2 phosphorylation in the frontal cortex and striatum. No sex-specific effects of genotype were found in the expression of TrkB ligands, BDNF and NT-4. However, a marked, region-specific increase in NT-4 expression was found in the striatum of both male and female BDNF(+/-) mice. In conclusion, there are complex sex- and region-specific changes in BDNF-TrkB signaling in BDNF(+/-) mice. These results provide new insight into sex-dependent BDNF signaling in forebrain regions and assist in understanding the role of neurotrophins in schizophrenia.


Neurobiology of Disease | 2012

Long-term behavioral and NMDA receptor effects of young-adult corticosterone treatment in BDNF heterozygous mice

Maren Klug; Rachel Anne Hill; Kwok Ho Christopher Choy; Michael Kyrios; Anthony J. Hannan; Maarten van den Buuse

Psychiatric illnesses, such as schizophrenia, are most likely caused by an interaction between genetic predisposition and environmental factors, including stress during development. The neurotrophin, brain-derived neurotrophic factor (BDNF) has been implicated in this illness as BDNF levels are decreased in the brain of patients with schizophrenia. The aim of the present study was to assess the combined effect of reduced BDNF levels and postnatal stress, simulated by chronic young-adult treatment with the stress hormone, corticosterone. From 6 weeks of age, female and male BDNF heterozygous mice and their wild-type controls were chronically treated with corticosterone in their drinking water for 3 weeks. At 11 weeks of age, male, but not female BDNF heterozygous mice treated with corticosterone exhibited a profound memory deficit in the Y-maze. There were no differences between the groups in baseline prepulse inhibition (PPI), a measure of sensorimotor gating, or its disruption by treatment with MK-801. However, an increase in startle caused by MK-801 treatment was absent in male, but not female BDNF heterozygous mice, irrespective of corticosterone treatment. Analysis of protein levels of the NMDA receptor subunits NR1, NR2A, NR2B and NR2C, showed a marked increase of NR2B levels in the dorsal hippocampus of male BDNF heterozygous mice treated with corticosterone. In the ventral hippocampus, significantly reduced levels of NR2A, NR2B and NR2C were observed in male BDNF heterozygous mice. The NMDA receptor effects in hippocampal sub-regions could be related to the spatial memory deficits and the loss of the effect of MK-801 on startle in these mice, respectively. No significant changes in NMDA receptor subunit levels were observed in any of the female groups. Similarly, no significant changes in levels of BDNF or its receptor, TrkB, were found other than the expected reduced levels of BDNF in heterozygous mice. In conclusion, the data show differential interactive effects of reduced levels of BDNF expression and corticosterone treatment on spatial memory and startle in male and female mice, accompanied by significant, but region-specific changes in NMDA receptor subunit levels in the dorsal and ventral hippocampus. These results could be important for our understanding of the interaction of neurodevelopmental stress and BDNF deficiency in cognitive and anxiety-related symptoms of psychiatric illnesses, such as schizophrenia.


Hippocampus | 2011

Brain-derived neurotrophic factor expression is increased in the hippocampus of 5-HT2C receptor knockout mice

Rachel Anne Hill; Simon S. Murray; Paul Halley; Michele D. Binder; Sally Martin; Maarten van den Buuse

Several studies have suggested a close interaction between serotonin (5‐HT) and BDNF; however, little is known of the specific relationship between BDNF and the 5‐HT2C receptor. Therefore, in this study we investigated BDNF expression in 5‐HT2C receptor knockout mice (5‐HT2CKO). We also assessed functional consequences of any changes in BDNF using a behavioral test battery. Western blot analysis demonstrated a significant 2.2‐fold increase in the expression of the mature form of BDNF in 5‐HT2CKO mice when compared with wild‐type controls (WT) in the hippocampus (P = 0.008), but not frontal cortex or striatum. No differences in the expression of the pro‐BDNF isoform were found, and the ratio of mature/pro BDNF was significantly increased in 5‐HT2CKO (P = 0.003). BDNF mRNA expression in the hippocampus was not different between the genotypes. Hence, increased mature BDNF levels in 5‐HT2CKO hippocampus are most likely due to increased extracellular cleavage rates of pro‐BDNF to its mature form. Protein expression of the BDNF receptor, tropomycin‐related receptor B (TrkB), was also unchanged in the hippocampus, frontal cortex and striatum. With repeated training in a 10‐day win‐shift radial arm maze task, 5‐HT2CKO and WT showed similar decreases of the number of working memory and reference memory errors. In addition, no genotype specific differences were observed for passive or active avoidance learning. 5‐HT2CKO showed modest locomotor hyperactivity but no differences in tests for anxiety, sensorimotor gating, or depressive‐like behaviors; however, in the tail suspension test 5‐HT2CKO showed significantly reduced climbing (P < 0.05). In conclusion, loss of 5‐HT2C receptor expression leads to a marked and selective increase in levels of the mature form of BDNF in the hippocampus. Despite this marked increase, 5‐HT2CKO show only subtle behavioral changes.

Collaboration


Dive into the Rachel Anne Hill's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wah Chin Boon

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Evan R. Simpson

Hudson Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

M. van den Buuse

Mental Health Research Institute

View shared research outputs
Top Co-Authors

Avatar

Maren Klug

Mental Health Research Institute

View shared research outputs
Top Co-Authors

Avatar

Anthony J. Hannan

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Top Co-Authors

Avatar

Margaret E. E. Jones

Prince Henry's Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Anna Schroeder

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge