Network


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

Hotspot


Dive into the research topics where C. Shannon Weickert is active.

Publication


Featured researches published by C. Shannon Weickert.


Translational Psychiatry | 2014

Markers of inflammation and stress distinguish subsets of individuals with schizophrenia and bipolar disorder

S G Fillman; David A. Sinclair; Samantha J. Fung; Maree J. Webster; C. Shannon Weickert

Schizophrenia and bipolar disorder share a number of common features, both symptomatically and biologically. Abnormalities in the neuroimmune and the stress-signaling pathways have been previously identified in brains of individuals with both diseases. However, the possible relationship between abnormalities in stress and neuroimmune signaling within the cortex of people with psychotic illness has not been defined. To test the hypothesis that combined alterations in brain stress responsiveness and neuroimmune/inflammatory status are characteristic of some individuals suffering from major mental illness, we examined gene expression in the Stanley Array Cohort of 35 controls, 35 individuals with schizophrenia and 34 individuals with bipolar disorder. We used levels of 8 inflammatory-related transcripts, of which SERPINA3 was significantly elevated in individuals with schizophrenia (F(2,88)=4.137, P<0.05), and 12 glucocorticoid receptor signaling (stress) pathway transcripts previously examined, to identify two clusters of individuals: a high inflammation/stress group (n=32) and a low (n=68) inflammation/stress group. The high inflammation/stress group has a significantly greater number of individuals with schizophrenia (n=15), and a trend toward having more bipolar disorder individuals (n=11), when compared with controls (n=6). Using these subgroups, we tested which microarray-assessed transcriptional changes may be associated with high inflammatory/stress groups using ingenuity analysis and found that an extended network of gene expression changes involving immune, growth factors, inhibitory signaling and cell death factors also distinguished these groups. Our work demonstrates that some of the heterogeneity in schizophrenia and bipolar disorder may be partially explained by inflammation/stress interactions, and that this biological subtype cuts across Diagnostic and Statistical Manual of Mental Disorders (DSM)-defined categories.


Translational Psychiatry | 2014

Decreased BDNF and TrkB mRNA expression in multiple cortical areas of patients with schizophrenia and mood disorders.

M T Ray; C. Shannon Weickert; Maree J. Webster

Abnormalities in brain-derived neurotrophic factor (BDNF)/trkB signaling have been implicated in the etiology of schizophrenia and mood disorders. Patients with schizophrenia, bipolar disorder (BPD) and major depression (MDD) have reduced levels of neurotrophins in their brains when compared with normal unaffected individuals; however, only a few brain areas have been examined to date. Owing to the broad range of symptoms manifested in these disorders, we hypothesized that multiple associative areas of the neocortex may be implicated and that the degree of change in BDNF and trkB−TK+ mRNA expression and the cortical region or layers involved may vary according to Diagnostic and Statistical Manual of Mental Disorders (DSM) diagnosis. We compared BDNF and trkB−TK+ mRNA levels across all layers of the prefrontal cortex (dorsolateral prefrontal cortex, DLPFC), orbital frontal cortex (OFC), anterior cingulate cortex (ACC), inferior temporal gyrus (ITG) and superior temporal gyrus (STG) in four groups: schizophrenia, BPD, MDD and unaffected controls (n=60). BDNF mRNA levels were significantly decreased in layers IV and V of DLPFC in schizophrenia patients, in layer VI of ACC in schizophrenia and MDD and in layer VI of ITG in schizophrenia, BPD and MDD. BDNF mRNA levels were also significantly decreased in layer V and/or VI of STG in schizophrenia, BPD and MDD. TrkB−TK+ mRNA levels were only significantly decreased in the cortical layer VI of OFC in BPD. The shared and distinct patterns of neurotrophin transcript reductions, with some specific to each group, may compromise the function and plasticity of distinct cortical areas to various degrees in the different groups and contribute to the range and overlap of symptoms manifested across the diagnoses.


Psychoneuroendocrinology | 2013

Serum testosterone levels are related to cognitive function in men with schizophrenia.

Loretta Moore; M. Kyaw; Ans Vercammen; Rhoshel Lenroot; Jayashri Kulkarni; Jackie Curtis; Maryanne O’Donnell; Vaughan J. Carr; C. Shannon Weickert; Thomas W. Weickert

BACKGROUND Sex steroids such as oestrogen and testosterone are potent neurodevelopmental hormones that also play a role in neuromodulation and neuroprotection of the mature brain. Sex steroid hormones may also be involved in the pathophysiology of schizophrenia as reduced circulating sex steroid levels and changes in brain sex steroid receptors are found in people with schizophrenia compared to controls. In men with schizophrenia, recent studies have documented an inverse correlation between serum testosterone and negative symptoms. Our study sought to confirm whether men with schizophrenia had lower levels of testosterone relative to controls and to determine whether lower testosterone levels were related to higher symptom severity and impaired cognition. METHOD Circulating serum hormone levels (testosterone, oestrogen, and prolactin), cognitive function and symptoms were assessed in 29 chronically ill men with schizophrenia or schizoaffective disorder. Twenty healthy men were recruited as a comparison group. A series of regression analyses were performed to determine the extent to which circulating sex steroid hormone levels predict cognition and symptoms in men with schizophrenia. RESULTS We did not find a significant difference in serum testosterone levels between groups. However, circulating testosterone levels significantly predicted performance on verbal memory, processing speed, and working memory in men with schizophrenia. With the exception of an effect of oestrogen on verbal memory, circulating sex steroid levels did not predict cognitive function in healthy men. Testosterone levels were not related to positive or negative symptom severity, but testosterone influenced excitement/hostility levels in our schizophrenia sample. CONCLUSIONS The results suggest that circulating sex steroids may modulate cognitive deficits associated with schizophrenia.


Translational Psychiatry | 2016

Cortical grey matter volume reduction in people with schizophrenia is associated with neuro-inflammation

Yiru Zhang; Vibeke S. Catts; Donna Sheedy; T. McCrossin; Jillian J. Kril; C. Shannon Weickert

Cortical grey matter volume deficits and neuro-inflammation exist in patients with schizophrenia, although it is not clear whether elevated cytokines contribute to the cortical volume reduction. We quantified cortical and regional brain volumes in fixed postmortem brains from people with schizophrenia and matched controls using stereology. Interleukin (IL)-6, IL-1β, IL-8 and SERPINA3 messenger RNAs (mRNAs) were quantified in the contralateral fresh frozen orbitofrontal cortex. We found a small, but significant reduction in cortical grey matter (1.3%; F(1,85)=4.478, P=0.037) and superior frontal gyrus (6.5%; F(1,80)=5.700, P=0.019) volumes in individuals with schizophrenia compared with controls. Significantly reduced cortical grey matter (9.2%; F(1,24)=8.272, P=0.008) and superior frontal gyrus (13.9%; F(1,20)=5.374, P=0.031) volumes were found in cases with schizophrenia and ‘high inflammation’ status relative to schizophrenia cases with ‘low inflammation’ status in the prefrontal cortex. The expression of inflammatory mRNAs in the orbitofrontal cortex was significantly correlated with those in dorsolateral prefrontal cortex (all r>0.417, all P<0.022), except for IL-8. Moreover, average daily and lifetime antipsychotic intake negatively correlated with cortical grey matter and superior frontal gyrus volumes (all r<−0.362, all P<0.05). The results suggest that the reduction in cortical grey matter volume in people with schizophrenia is exaggerated in those who have high expression of inflammatory cytokines. Further, antipsychotic medication intake does not appear to ameliorate the reduction in brain volume.


Schizophrenia Research | 2015

Testosterone and reward prediction-errors in healthy men and men with schizophrenia

Richard W. Morris; Tertia D. Purves-Tyson; C. Shannon Weickert; D. Rothmond; Rhoshel Lenroot; Thomas W. Weickert

Sex hormones impact reward processing, which is dysfunctional in schizophrenia; however, the degree to which testosterone levels relate to reward-related brain activity in healthy men and the extent to which this relationship may be altered in men with schizophrenia has not been determined. We used functional magnetic resonance imaging (fMRI) to measure neural responses in the striatum during reward prediction-errors and hormone assays to measure testosterone and prolactin in serum. To determine if testosterone can have a direct effect on dopamine neurons, we also localized and measured androgen receptors in human midbrain with immunohistochemistry and quantitative PCR. We found correlations between testosterone and prediction-error related activity in the ventral striatum of healthy men, but not in men with schizophrenia, such that testosterone increased the size of positive and negative prediction-error related activity in a valence-specific manner. We also identified midbrain dopamine neurons that were androgen receptor immunoreactive, and found that androgen receptor (AR) mRNA was positively correlated with tyrosine hydroxylase (TH) mRNA in human male substantia nigra. The results suggest that sex steroid receptors can potentially influence midbrain dopamine biosynthesis, and higher levels of serum testosterone are linked to better discrimination of motivationally-relevant signals in the ventral striatum, putatively by modulation of the dopamine biosynthesis pathway via AR ligand binding. However, the normal relationship between serum testosterone and ventral striatum activity during reward learning appears to be disrupted in schizophrenia.


Hippocampus | 2014

Gonadectomy increases neurogenesis in the male adolescent rhesus macaque hippocampus.

Katherine M. Allen; Samantha J. Fung; Debora A. Rothmond; Pamela L. Noble; C. Shannon Weickert

New neurons are continuously produced in the subgranular zone of the adult hippocampus and can modulate hippocampal plasticity across life. Adolescence is characterized by dramatic changes in sex hormone levels, and social and emotional behaviors. It is also an age for increased risk of psychiatric disorders, including schizophrenia, which may involve altered hippocampal neurogenesis. The extent to which testosterone and other testicular hormones modulate hippocampal neurogenesis and adolescent behavioral development is unclear. This study aimed to determine if removal of testicular hormones during adolescence alters neurogenesis in the male rhesus macaque hippocampus. We used stereology to examine levels of cell proliferation, cell survival and neuronal differentiation in late adolescent male rhesus macaques (4.6‐yrs old) that had previously been gonadectomized or sham operated prior to puberty (2.4‐yrs old). While the absence of adolescent testicular hormones had no effect on cell proliferation, cell survival was increased by 65% and indices of immature neuronal differentiation were increased by 56% in gonadectomized monkeys compared to intact monkeys. We show for the first time that presence of circulating testicular hormones, including testosterone, may decrease neuronal survival in the primate hippocampus during adolescence. Our findings are in contrast to existing studies in adults where testosterone tends to be a pro‐survival factor and demonstrate that testicular hormones may reduce hippocampal neurogenesis during the age typical of schizophrenia onset.


Translational Psychiatry | 2017

Putative presynaptic dopamine dysregulation in schizophrenia is supported by molecular evidence from post-mortem human midbrain

Tertia D. Purves-Tyson; Samantha J. Owens; Debora A. Rothmond; Glenda M. Halliday; Kay L. Double; Julia Stevens; T. McCrossin; C. Shannon Weickert

The dopamine hypothesis of schizophrenia posits that increased subcortical dopamine underpins psychosis. In vivo imaging studies indicate an increased presynaptic dopamine synthesis capacity in striatal terminals and cell bodies in the midbrain in schizophrenia; however, measures of the dopamine-synthesising enzyme, tyrosine hydroxylase (TH), have not identified consistent changes. We hypothesise that dopamine dysregulation in schizophrenia could result from changes in expression of dopamine synthesis enzymes, receptors, transporters or catabolic enzymes. Gene expression of 12 dopamine-related molecules was examined in post-mortem midbrain (28 antipsychotic-treated schizophrenia cases/29 controls) using quantitative PCR. TH and the synaptic dopamine transporter (DAT) proteins were examined in post-mortem midbrain (26 antipsychotic-treated schizophrenia cases per 27 controls) using immunoblotting. TH and aromatic acid decarboxylase (AADC) mRNA and TH protein were unchanged in the midbrain in schizophrenia compared with controls. Dopamine receptor D2 short, vesicular monoamine transporter (VMAT2) and DAT mRNAs were significantly decreased in schizophrenia, with no change in DRD3 mRNA, DRD3nf mRNA and DAT protein between diagnostic groups. However, DAT protein was significantly increased in putatively treatment-resistant cases of schizophrenia compared to putatively treatment-responsive cases. Midbrain monoamine oxidase A (MAOA) mRNA was increased, whereas MAOB and catechol-O-methyl transferase mRNAs were unchanged in schizophrenia. We conclude that, whereas some mRNA changes are consistent with increased dopamine action (decreased DAT mRNA), others suggest reduced dopamine action (increased MAOA mRNA) in the midbrain in schizophrenia. Here, we identify a molecular signature of dopamine dysregulation in the midbrain in schizophrenia that mainly includes gene expression changes of molecules involved in dopamine synthesis and in regulating the time course of dopamine action.


Translational Psychiatry | 2017

Brain antibodies in the cortex and blood of people with schizophrenia and controls

Lj Glass; David A. Sinclair; Danny Boerrigter; K Naude; Samantha J. Fung; David A. Brown; Vibeke S. Catts; Paul A. Tooney; Maryanne O'Donnell; Rhoshel Lenroot; C Galletty; Dennis Liu; Thomas W. Weickert; C. Shannon Weickert

The immune system is implicated in the pathogenesis of schizophrenia, with elevated proinflammatory cytokine mRNAs found in the brains of ~40% of individuals with the disorder. However, it is not clear if antibodies (specifically immunoglobulin-γ (IgG)) can be found in the brain of people with schizophrenia and if their abundance relates to brain inflammatory cytokine mRNA levels. Therefore, we investigated the localization and abundance of IgG in the frontal cortex of people with schizophrenia and controls, and the impact of proinflammatory cytokine status on IgG abundance in these groups. Brain IgGs were detected surrounding blood vessels in the human and non-human primate frontal cortex by immunohistochemistry. IgG levels did not differ significantly between schizophrenia cases and controls, or between schizophrenia cases in ‘high’ and ‘low’ proinflammatory cytokine subgroups. Consistent with the existence of IgG in the parenchyma of human brain, mRNA and protein of the IgG transporter (FcGRT) were present in the brain, and did not differ according to diagnosis or inflammatory status. Finally, brain-reactive antibody presence and abundance was investigated in the blood of living people. The plasma of living schizophrenia patients and healthy controls contained antibodies that displayed positive binding to Rhesus macaque cerebellar tissue, and the abundance of these antibodies was significantly lower in patients than controls. These findings suggest that antibodies in the brain and brain-reactive antibodies in the blood are present under normal circumstances.


Brain Behavior and Immunity | 2015

Developmental influences on schizophrenia-like cognitive and neurophysiological outcomes in a rat model of maternal immune challenge

Lauren R. Harms; Crystal Meehan; Katerina Zavitsanou; R. Fulham; A. Wong; Juanita Todd; Ulrich Schall; C. Shannon Weickert; Patricia T. Michie; Deborah M. Hodgson

The developing brain is sensitive to early-life insults, which can increase the risk of neuropsychiatric disorders such as schizophrenia. Maternal immune system activation during gestation via infection is a known risk factor for schizophrenia. Here we examine the role of early-life immune activation on schizophrenia-related behavioural and electrophysiological outcomes. Pregnant Wistar rats were injected with the viral mimic, Poly(I:C) or vehicle at either gestational day (GD) 10 or GD19. Adult offspring were examined for behaviour (prepulse inhibition (PPI) and working memory) and electrophysiology (generation 50Hz auditory steady-state responses (aSSRs)).Brain tissue was examined for NMDA receptor (NMDAR) binding. Rats exposed to Poly(I:C) at GD10 were found to exhibit reduced PPI, whereas those exposed to Poly(I:C) during late gestation exhibited working memory impairments. In addition, late gestation-exposed rats exhibited reduced power of 50Hz aSSRs, indicating an impaired ability to generate neural oscillations in the high-frequency (gamma) range. Adult offspring exposed to Poly(I:C) on GD19 had increased binding to the NMDAR2A subunit and the NMDAR channel in the hippocampus. Maternal immune activation via a viral mimic was sufficient to alter the trajectory of brain development such that there were long-term impacts of several behavioural and electrophysiological outcomes in adulthood. Several of the behavioural and electrophysiological changes identified here are also found in patients with schizophrenia, indicating that the Maternal Immune Activated rat may model aspects of the disorder.


Schizophrenia Research | 2000

Decreased glucocorticoid receptor mRNA levels in individuals with depression, bipolar disorder and schizophrenia

Maree J. Webster; J. O'Grady; J. Orthmann; C. Shannon Weickert

Collaboration


Dive into the C. Shannon Weickert's collaboration.

Top Co-Authors

Avatar

Samantha J. Fung

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Thomas W. Weickert

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Maree J. Webster

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Rhoshel Lenroot

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Tertia D. Purves-Tyson

Neuroscience Research Australia

View shared research outputs
Top Co-Authors

Avatar

Danny Boerrigter

Neuroscience Research Australia

View shared research outputs
Top Co-Authors

Avatar

Debora A. Rothmond

Neuroscience Research Australia

View shared research outputs
Top Co-Authors

Avatar

Katherine M. Allen

Neuroscience Research Australia

View shared research outputs
Top Co-Authors

Avatar

M. Kyaw

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge