Dr  Julia Shaw

Dr Julia Shaw

Research Fellow

School of Biomedical Sciences and Pharmacy

Preventing the mental and behavioural risks of preterm birth

Dr Julia Shaw is a postdoctoral researcher working in the Pregnancy Compromise and Neuroprotection group with the Mothers & Babies Research Program. Her goal is to restore brain development trajectories in the immediate neonatal period.

Julia Shaw at a microscope in her lab

Julia's work is all about neurosteroid loss in preterm fetuses and how it can negatively impact the maturing brain.

"In the third trimester of pregnancy, the brain undergoes rapid development, and one of the key promoters of this is neurosteroids," she says.

Neurosteroids are a class of naturally occurring compounds synthesised in the brain and other parts of the nervous system. They play a significant role in regulating neuronal excitability, mood, stress responses and other physiological functions.

Neurosteroids are supplied by the placenta to the developing fetus and subsequently lost at birth once separation from the placenta occurs.

"When birth happens early, as in preterm birth, the neurosteroids are lost too soon, and brain development is significantly impacted, " she continues. "This can lead to behavioural, learning, and mental health impairments."

"If we can reinstate the supply of neurosteroids postnatally, then we could prevent this cascade of effects."

The preterm babies affected

Julia has always been interested in how the brain develops. But a big drawcard for her to this work was the potential to improve the quality of life for babies born preterm.

Globally, 10 per cent of births yearly are preterm, even in developed nations like Australia. That's 13.4 million babies annually.

This is an astounding number of babies whose lives could be drastically improved by a simple postnatal neurosteroid-based therapy administered between birth and full term.

"Even babies born in the 32-37 week range can appear well at the time of birth. However, they can go on to develop neurological impairments," says Julia.

This includes learning delays, behavioural disorders like ADHD, and mental health disorders including depression and anxiety. These can greatly affect their and their family's quality of life.

Clinical collaboration in New Zealand

In the first year of her PhD, Julia collaborated with a leading neonatologist at Wellington Hospital in New Zealand.

Wellington Hospital NICU services the central region of New Zealand and is, therefore, a major hub for neonatal intensive care. This collaboration also extended to researchers at the University of Otago.

She made numerous trips to Wellington to perform experiments with these researchers under the supervision of the neonatologist. This meant the research was directly relevant to the clinic, and the approaches used would be most transferrable to the clinic.

Restoring brain trajectory for better lives

To date, Julia and her team's work suggests that replacing the supply of neurosteroids after birth and until the equivalent of full-term birth, could restore brain development to a normal trajectory and prevent the negative impact.

The goal is to restore brain development trajectories in the immediate neonatal period and prevent these poor long-term neurodevelopmental outcomes.

This research has the potential to greatly improve the quality of life for babies born preterm and their families by mitigating the risk of developing neurodevelopmental disorders.

These disorders are often life-long and are associated with poor school performance, difficulty forming and maintaining relationships, and reduced income potential as an adult.

In addition, treatments are expensive and ongoing with a raft of side effects and don't target the cause but rather the symptoms of the disorders.

These individuals and their families are therefore burdened with additional costs and reduced ability to earn long-term.

The impact is significant.

Consuming work; relevant results

Julia shares that the biggest barrier to success in this type of research is the time-consuming nature of the models and approaches.

"We need our work performed in a way that gives the greatest clinical relevance. But unfortunately, this is very time-consuming in terms of the length of the experiment and time devotion during the experiment."

"However, this approach ultimately provides us with a model that gives repeatable results and, most importantly, clinically relevant results."

Excited by the potential

Julia continues to be propelled by the latent benefits of her work.

"I find it heartbreaking that an individual 's life could be affected in such a way by simply being born four weeks early. Before taking their first breaths, these babies are predisposed to developing an array of neurodevelopmental impairments with far-reaching impacts on their lives and that of their family,” she says.

The potential to give each baby the best possible start to life is what motivates her to perform her research. And the promising nature of the results is what excites her—that this actually may be feasible.

Doctor Julia Shaw at a microscope in her laboratory

Preventing the mental and behavioural risks of preterm birth

Julia's work is all about neurosteroid loss in preterm fetuses and how it can negatively impact the maturing brain.

Read more

Career Summary

Biography

Dr Julia Shaw is a postdoctoral researcher working in the Pregnancy Compromise and Neuroprotection group with the Mothers and Babies Research Centre. Her research examines the role of endocrine disruption following preterm birth on brain development of the offspring, and in particular, focuses on the long-term consequences into childhood and beyond.

She has recently been awarded her PhD in Experimental Pharmacology from the University of Newcastle in 2017. During her PhD, Dr Shaw focused on how a premature loss of placental neurosteroids results in diffuse white matter injury and impairments of the GABAergic system in the juvenile brain following preterm birth. She identified that the hippocampus and cerebellum are particularly vulnerable to these disturbances in brain development and may play a contributing role in the increased rate of behavioural disorders, such as ADHD and anxiety, that are more common following preterm birth.

Dr Shaw is currently investigating the use of a neurosteroid-replacement therapy to prevent these adverse outcomes of preterm birth on brain development.


Qualifications

  • Doctor of Philosophy, University of Newcastle
  • Bachelor of Biomedical Sciences, University of Newcastle
  • Bachelor of Biomedical Sciences (Hons), University of Newcastle

Keywords

  • Neurodevelopment
  • Neurosteroids
  • Oligodendrocytes
  • Preterm Birth

Fields of Research

Code Description Percentage
321501 Foetal development and medicine 100

Professional Experience

UON Appointment

Title Organisation / Department
Research Fellow University of Newcastle
School of Biomedical Sciences and Pharmacy
Australia

Awards

Prize

Year Award
2017 Best Poster Presentation (ASMR Newcastle Satellite Meeting)
Australian Society of Medical Research (ASMR) Newcastle Satellite Meeting
2014 Best Early PhD Oral Presentation (Fetal and Neonatal Workshop of Aus and NZ)
Fetal and Neonatal Workshop of Australia and New Zealand
Edit

Publications

For publications that are currently unpublished or in-press, details are shown in italics.


Journal article (21 outputs)

Year Citation Altmetrics Link
2024 Moloney RA, Palliser HK, Dyson RM, Pavy CL, Berry M, Hirst JJ, Shaw JC, 'Ongoing effects of preterm birth on the dopaminergic and noradrenergic pathways in the frontal cortex and hippocampus of guinea pigs.', Dev Neurobiol, (2024) [C1]
DOI 10.1002/dneu.22937
2024 Pavy CL, Shaw JC, Moloney RA, Palliser HK, Hirst JJ, 'Potential for a cerebellar role in moderate-late preterm associated behavioural disorders.', Front Pediatr, 12 1336137 (2024) [C1]
DOI 10.3389/fped.2024.1336137
Co-authors Hannah Palliser
2023 Moloney RA, Pavy CL, Kahl RGS, Palliser HK, Hirst JJ, Shaw JC, 'Dual isolation of primary neurons and oligodendrocytes from guinea pig frontal cortex.', Front Cell Neurosci, 17 1298685 (2023) [C1]
DOI 10.3389/fncel.2023.1298685
Co-authors Jon Hirst, Hannah Palliser
2023 Crombie GK, Palliser HK, Shaw JC, Hanley BA, Moloney RA, Hirst JJ, 'Prenatal Stress Induces Translational Disruption Associated with Myelination Deficits.', Developmental neuroscience, 45 290-308 (2023) [C1]
DOI 10.1159/000530282
Citations Scopus - 2
Co-authors Hannah Palliser, Jon Hirst
2022 Crombie GK, Palliser HK, Shaw JC, Hodgson DM, Walker DW, Hirst JJ, 'Evaluating changes in GABAergic and glutamatergic pathways in early life following prenatal stress and postnatal neurosteroid supplementation', PSYCHONEUROENDOCRINOLOGY, 139 (2022) [C1]
DOI 10.1016/j.psyneuen.2022.105705
Citations Scopus - 5
Co-authors Hannah Palliser, Deborah Hodgson, Jon Hirst
2022 Shaw JC, Dyson RM, Palliser HK, Sixtus RP, Barnes H, Pavy CL, et al., 'Examining Neurosteroid-Analogue Therapy in the Preterm Neonate For Promoting Hippocampal Neurodevelopment', FRONTIERS IN PHYSIOLOGY, 13 (2022) [C1]
DOI 10.3389/fphys.2022.871265
Citations Scopus - 2
Co-authors Hannah Palliser, Jon Hirst
2022 Shaw JC, Dyson RM, Palliser HK, Crombie GK, Berry MJ, Hirst JJ, 'Adaptations in the Hippocampus during the Fetal to Neonatal Transition in Guinea Pigs', Reproductive Medicine, 3 85-100 [C1]
DOI 10.3390/reprodmed3020008
Co-authors Hannah Palliser, Jon Hirst
2021 Crombie GK, Palliser HK, Shaw JC, Hodgson DM, Walker DW, Hirst JJ, 'Effects of prenatal stress on behavioural and neurodevelopmental outcomes are altered by maternal separation in the neonatal period', PSYCHONEUROENDOCRINOLOGY, 124 (2021) [C1]
DOI 10.1016/j.psyneuen.2020.105060
Citations Scopus - 17Web of Science - 9
Co-authors Deborah Hodgson, Hannah Palliser, Jon Hirst
2021 Crombie GK, Palliser HK, Shaw JC, Hodgson DM, Walker DW, Hirst JJ, 'Neurosteroid-based intervention using Ganaxolone and Emapunil for improving stress-induced myelination deficits and neurobehavioural disorders', PSYCHONEUROENDOCRINOLOGY, 133 (2021) [C1]
DOI 10.1016/j.psyneuen.2021.105423
Citations Scopus - 6Web of Science - 2
Co-authors Hannah Palliser, Deborah Hodgson, Jon Hirst
2021 Shaw JC, Crombie GK, Palliser HK, Hirst JJ, 'Impaired Oligodendrocyte Development Following Preterm Birth: Promoting GABAergic Action to Improve Outcomes', FRONTIERS IN PEDIATRICS, 9 (2021) [C1]
DOI 10.3389/fped.2021.618052
Citations Scopus - 15Web of Science - 4
Co-authors Hannah Palliser, Jon Hirst
2019 Shaw JC, Crombie GK, Zakar T, Palliser HK, Hirst JJ, 'Perinatal compromise contributes to programming of GABAergic and glutamatergic systems leading to long-term effects on offspring behaviour', JOURNAL OF NEUROENDOCRINOLOGY, 32 (2019) [C1]
DOI 10.1111/jne.12814
Citations Scopus - 12Web of Science - 9
Co-authors Hannah Palliser, Jon Hirst
2019 Shaw JC, Berry MJ, Dyson RM, Crombie GK, Hirst JJ, Palliser HK, 'Reduced Neurosteroid Exposure Following Preterm Birth and Its' Contribution to Neurological Impairment: A Novel Avenue for Preventative Therapies', Frontiers in Physiology, 10 (2019) [C1]
DOI 10.3389/fphys.2019.00599
Citations Scopus - 20Web of Science - 14
Co-authors Jon Hirst, Hannah Palliser
2019 Shaw JC, Dyson RM, Palliser HK, Gray C, Berry MJ, Hirst JJ, 'Neurosteroid replacement therapy using the allopregnanolone-analogue ganaxolone following preterm birth in male guinea pigs', Pediatric Research, 85 86-96 (2019) [C1]

Background: Children born preterm, especially boys, are at increased risk of developing attention deficit hyperactivity disorder (ADHD) and learning difficulties. We propose that ... [more]

Background: Children born preterm, especially boys, are at increased risk of developing attention deficit hyperactivity disorder (ADHD) and learning difficulties. We propose that neurosteroid-replacement therapy with ganaxolone (GNX) following preterm birth may mitigate preterm-associated neurodevelopmental impairment. Methods: Time-mated sows were delivered preterm (d62) or at term (d69). Male preterm pups were randomized to ganaxolone (Prem-GNX; 2.5 mg/kg subcutaneously twice daily until term equivalence), or preterm control (Prem-CON). Surviving male juvenile pups underwent behavioural testing at d25-corrected postnatal age (CPNA). Brain tissue was collected at CPNA28 and mature myelinating oligodendrocytes of the hippocampus and subcortical white matter were quantified by immunostaining of myelin basic protein (MBP). Results: Ganaxolone treatment returned the hyperactive behavioural phenotype of preterm-born juvenile males to a term-born phenotype. Deficits in MBP immunostaining of the preterm hippocampus and subcortical white matter were also ameliorated in animals receiving ganaxolone. However, during the treatment period weight gain was poor, and pups were sedated, ultimately increasing the neonatal mortality rate. Conclusion: Ganaxolone improved neurobehavioural outcomes in males suggesting that neonatal treatment may be an option for reducing preterm-associated neurodevelopmental impairment. However, dosing studies are required to reduce the burden of unwanted side effects.

DOI 10.1038/s41390-018-0185-7
Citations Scopus - 21Web of Science - 15
Co-authors Jon Hirst, Hannah Palliser
2018 Hirst JJ, Palliser HK, Shaw JC, Crombie G, Walker DW, Zakar T, 'Birth and Neonatal Transition in the Guinea Pig: Experimental Approaches to Prevent Preterm Birth and Protect the Premature Fetus', FRONTIERS IN PHYSIOLOGY, 9 (2018) [C1]
DOI 10.3389/fphys.2018.01802
Citations Scopus - 12Web of Science - 9
Co-authors Jon Hirst, Hannah Palliser
2018 Shaw JC, Palliser HK, Dyson RM, Berry MJ, Hirst JJ, 'Disruptions to the cerebellar GABAergic system in juvenile guinea pigs following preterm birth', International Journal of Developmental Neuroscience, 65 1-10 (2018) [C1]

Background Children that are born preterm are at an increased risk of developing cognitive problems and behavioural disorders, such as attention deficit hyperactivity disorder (AD... [more]

Background Children that are born preterm are at an increased risk of developing cognitive problems and behavioural disorders, such as attention deficit hyperactivity disorder (ADHD). There is increasing interest in the role of the cerebellum in these processes and the potential involvement of GABAergic pathways in neurodevelopmental disorders. We propose that preterm birth, and the associated loss of the trophic intrauterine environment, alters the development of the cerebellum, contributing to ongoing neurobehavioral disorders. Methods Guinea pigs were delivered preterm (GA62) or spontaneously at term (GA69), and tissues collected at corrected postnatal day (PND) 28. Neurodevelopmental and GABAergic markers myelin basic protein (MBP), neuronal nuclei (NeuN), calbindin (Purkinje cells), and GAD67 (GABA synthesis enzyme) were analysed in cerebellar lobules IX and X by immunohistochemistry. Protein expression of GAD67 and GAT1 (GABA transporter enzyme) were quantified by western blot, whilst neurosteroid-sensitive GABAA receptor subunits were measured by RT-PCR. Results MBP immunostaining was increased in lobule IX of preterm males, and reduced in lobule X of preterm females when compared to their term counterparts. GAD67 staining was decreased in lobule IX and X of the preterm males, but only in lobule X of the preterm females compared to term cohorts for each sex. Internal granule cell layer width of lobule X was decreased in preterm cohorts of both sexes compared to terms. There were no differences between gestational age groups for NeuN staining, GAD67 and GAT1 protein expression as measured by western blotting, or GABAA receptor subunits as measured by RT-PCR between preterm and term for either sex. Conclusions The present findings suggest that components of the cerebellar GABAergic system of the ex-preterm cerebellum are disrupted. The higher expression of myelin in the preterm males may be due to a deficit in axonal pruning, whereas females have a deficit in myelination at 28 corrected days of age. Together these ongoing alterations may contribute to the neurodevelopmental and behavioural disorders observed in those born preterm.

DOI 10.1016/j.ijdevneu.2017.10.002
Citations Scopus - 18Web of Science - 15
Co-authors Jon Hirst, Hannah Palliser
2018 Shaw JC, Palliser HK, Palazzi K, Hirst JJ, 'Administration of Progesterone Throughout Pregnancy Increases Maternal Steroids Without Adverse Effect on Mature Oligodendrocyte Immunostaining in the Guinea Pig', Reproductive Sciences, 25 395-405 (2018) [C1]

Progesterone is administered to pregnant women at risk of premature labor, despite systematic reviews showing conflicting outcomes regarding its use, highlighting doubt over the e... [more]

Progesterone is administered to pregnant women at risk of premature labor, despite systematic reviews showing conflicting outcomes regarding its use, highlighting doubt over the effectiveness of the therapy. Progesterone can be rapidly metabolized into a number of steroids, but to date, there has been a lack of investigation into the fetal steroid profiles following administration and whether this impacts fetal neurodevelopment. The objective of this study was to determine the effect of progesterone treatment on allopregnanolone and cortisol levels in the fetus and on a marker of myelination in the fetal brain. We used a guinea pig model where pregnant dams were administered vehicle (ß-cyclodextrin) or progesterone orally throughout pregnancy (GA29-61). Maternal and fetal fluids and tissues were collected at both preterm (GA61) and term (GA68) ages. Maternal and fetal progesterone and cortisol were analyzed by enzyme immunoassay and allopregnanolone by radioimmunoassay. Measurement of myelination of fetal brains (hippocampus, cingulum, and subcortical white matter) at preterm and term ages was performed by immunohistochemistry staining for myelin basic protein. We found that dams receiving progesterone had significantly elevated progesterone and cortisol concentrations, but there was no effect on allopregnanolone. Interestingly, the increased cortisol concentrations were not reflected in the fetuses, and there was no effect of progesterone treatment on myelination. Therefore, we conclude that in our guinea pig model, maternal administration of progesterone has no effect on cortisol levels or markers of mature oligodendrocytes in the fetus and suggest this is potentially due to the protective cortisol barrier in the placenta.

DOI 10.1177/1933719117715125
Citations Scopus - 4Web of Science - 2
Co-authors Hannah Palliser, Jon Hirst
2017 Bennett GA, Palliser HK, Shaw JC, Palazzi KL, Walker DW, Hirst JJ, 'Maternal stress in pregnancy affects myelination and neurosteroid regulatory pathways in the guinea pig cerebellum', Stress, 20 580-588 (2017) [C1]

Prenatal stress predisposes offspring to behavioral pathologies. These may be attributed to effects on cerebellar neurosteroids and GABAergic inhibitory signaling, which can be li... [more]

Prenatal stress predisposes offspring to behavioral pathologies. These may be attributed to effects on cerebellar neurosteroids and GABAergic inhibitory signaling, which can be linked to hyperactivity disorders. The aims were to determine the effect of prenatal stress on markers of cerebellar development, a key enzyme in neurosteroid synthesis and the expression of GABAA receptor (GABAAR) subunits involved in neurosteroid signaling. We used a model of prenatal stress (strobe light exposure, 2 h on gestational day 50, 55, 60 and 65) in guinea pigs, in which we have characterized anxiety and neophobic behavioral outcomes. The cerebellum and plasma were collected from control and prenatally stressed offspring at term (control fetus: n = 9 male, n = 7 female; stressed fetus: n = 7 male, n = 8 female) and postnatal day (PND) 21 (control: n = 8 male, n = 8 female; stressed: n = 9 male, n = 6 female). We found that term female offspring exposed to prenatal stress showed decreased expression of mature oligodendrocytes (~40% reduction) and these deficits improved to control levels by PND21. Reactive astrocyte expression was lower (~40% reduction) following prenatal stress. GABAAR subunit (d and a6) expression and circulating allopregnanolone concentrations were not affected by prenatal stress. Prenatal stress increased expression (~150¿250% increase) of 5a-reductase type-1 mRNA in the cerebellum, which may be a neuroprotective response to promote GABAergic inhibition and aid in repair. These observations indicate that prenatal stress exposure has marked effects on the development of the cerebellum. These findings suggest cerebellar changes after prenatal stress may contribute to adverse behavioral outcomes after exposure to these stresses.

DOI 10.1080/10253890.2017.1378637
Citations Scopus - 16Web of Science - 9
Co-authors Hannah Palliser, Greer Bennett, Jon Hirst
2016 Hirst JJ, Cumberland AL, Shaw JC, Bennett GA, Kelleher MA, Walker DW, Palliser HK, 'Loss of neurosteroid-mediated protection following stress during fetal life', Journal of Steroid Biochemistry and Molecular Biology, 160 181-188 (2016) [C1]

Elevated levels of neurosteroids during late gestation protect the fetal brain from hypoxia/ischaemia and promote neurodevelopment. Suppression of allopregnanolone production duri... [more]

Elevated levels of neurosteroids during late gestation protect the fetal brain from hypoxia/ischaemia and promote neurodevelopment. Suppression of allopregnanolone production during pregnancy leads to the onset of seizure-like activity and potentiates hypoxia-induced brain injury. Markers of myelination are reduced and astrocyte activation is increased. The placenta has a key role in maintaining allopregnanolone concentrations in the fetal circulation and brain during gestation and levels decline markedly after both normal and preterm birth. This leads to the preterm neonate developing in a neurosteroid deficient environment between delivery and term equivalence. The expression of 5a-reductases is also lower in the fetus prior to term. These deficiencies in neurosteroid exposure may contribute to the increase in incidence of the adverse patterns of behaviour seen in children that are born preterm. Repeated exposure to glucocorticoid stimulation suppresses 5a-reductase expression and allopregnanolone levels in the fetus and results in reduced myelination. Both fetal growth restriction and prenatal maternal stress lead to increased cortisol concentrations in the maternal and fetal circulation. Prenatal stress results in reduced expression of key GABAA receptor subunits that normally heighten neurosteroid sensitivity. These stressors also result in altered placental allopregnanolone metabolism pathways. These findings suggest that reduced neurosteroid production and action in the perinatal period may contribute to some of the adverse neurodevelopmental and behavioural outcomes that result from these pregnancy compromises. Studies examining perinatal steroid supplementation therapy with non-metabolisable neurosteroid analogues to improve these outcomes are warranted.

DOI 10.1016/j.jsbmb.2015.09.012
Citations Scopus - 28Web of Science - 21
Co-authors Hannah Palliser, Jon Hirst, Greer Bennett
2016 Shaw JC, Palliser HK, Dyson RM, Hirst JJ, Berry MJ, 'Long-term effects of preterm birth on behavior and neurosteroid sensitivity in the Guinea pig', Pediatric Research, 80 275-283 (2016) [C1]

Background: Ex-preterm children and adolescents are at risk of developing late-onset neurodevelopmental and behavioral disorders. The mechanisms by which this happens are poorly u... [more]

Background: Ex-preterm children and adolescents are at risk of developing late-onset neurodevelopmental and behavioral disorders. The mechanisms by which this happens are poorly understood and relevant animal models are required. Methods: Ex-preterm (delivered at 62 d gestation) and term (spontaneously delivered) juvenile Guinea pigs underwent behavioral testing at 25 d corrected postnatal age, with tissues collected at 28 d. Neurodevelopmental markers (myelin basic protein (MBP) and glial fibrillary acidic protein (GFAP)) were analyzed in the hippocampus and subcortical white matter by immunohistochemistry. Gamma-aminobutyric acid A (GABA A) receptor subunit mRNA levels were quantified by reverse transcription polymerase chain reaction (RT-PCR), and salivary cortisol measured by enzyme-linked immunosorbent assay. Results: Preterm males travelled greater distances, were mobile for longer, spent more time investigating objects, and approached or interacted with familiar animals more than controls. Myelination and reactive astrocyte coverage was lower in the hippocampus and the subcortical white matter in preterm males. Hippocampal levels of the a5 subunit were also lower in the preterm male brain. Baseline salivary cortisol was higher for preterm males compared to controls. Conclusion: We conclude that juvenile ex-preterm male Guinea pigs exhibit a hyperactive phenotype and feature impaired neurodevelopment, making this a suitable model for future therapeutic studies.

DOI 10.1038/pr.2016.63
Citations Scopus - 21Web of Science - 17
Co-authors Hannah Palliser, Jon Hirst
2015 Bennett GA, Palliser HK, Shaw JC, Walker D, Hirst JJ, 'Prenatal stress alters hippocampal neuroglia and increases anxiety in childhood', Developmental Neuroscience, 37 533-545 (2015) [C1]

Prenatal stress has been associated with detrimental outcomes of pregnancy, including altered brain development leading to behavioural pathologies. The neurosteroid allopregnanolo... [more]

Prenatal stress has been associated with detrimental outcomes of pregnancy, including altered brain development leading to behavioural pathologies. The neurosteroid allopregnanolone has been implicated in mediating some of these adverse outcomes following prenatal stress due to its potent inhibitory and anxiolytic effects on the brain. The aims of the current study were to characterise key markers for brain development as well as behavioural parameters, adrenocortical responses to handling and possible neurosteroid influences towards outcomes in Guinea pig offspring in childhood. Pregnant Guinea pig dams were exposed to strobe light for 2 h (9-11 a.m.) on gestational days 50, 55, 60, and 65 and were left to deliver spontaneously at term and care for their litter. Behavioural testing (open-field test, object exploration test) of the offspring was performed at postnatal day 18 (with salivary cortisol and DHEA measured), and brains were collected at post-mortem on day 21. Markers of brain development myelin basic protein (MBP) and glial fibrillary acidic protein (GFAP) were assessed via immunohistochemistry, and the neurosteroid allopregnanolone and its rate-limiting enzymes 5a-reductase types 1 and 2 (5aR1/2) were measured in neonatal brains by radioimmunoassay, reverse transcriptase polymerase chain reaction (RT-PCR), and Western blot, respectively. Brain-derived neurotrophic factor protein was measured as a marker of synaptic plasticity, and GABAA receptor subunit expression was also assessed using RT-PCR. Neonates born from mothers stressed during late pregnancy showed a reduction in both MBP (p < 0.01) and GFAP (p < 0.05) expression in the CA1 region of the hippocampus at 21 days of age. Pups of prenatally stressed pregnancies also showed higher levels of anxiety and neophobic behaviours at the equivalent of childhood (p < 0.05). There were no significant changes observed in allopregnanolone levels, 5aR1/2 expression, or GABAA receptor subunit expression in prenatally stressed neonates compared to controls. This study shows alterations in markers of myelination and reactive astrocytes in the hippocampus of offspring exposed to prenatal stress. These changes are also observed in offspring that show increased anxiety behaviours at the equivalent of childhood, which indicates ongoing structural and functional postnatal changes after prenatal stress exposure.

DOI 10.1159/000437302
Citations Scopus - 36Web of Science - 31
Co-authors Jon Hirst, Hannah Palliser, Greer Bennett
2015 Shaw JC, Palliser HK, Walker DW, Hirst JJ, 'Preterm birth affects GABA

Modulation of gamma-aminobutyric acid A (GABAA) receptor signalling by the neurosteroid allopregnanolone has a major role in late gestation neurodevelopment. The objective of this... [more]

Modulation of gamma-aminobutyric acid A (GABAA) receptor signalling by the neurosteroid allopregnanolone has a major role in late gestation neurodevelopment. The objective of this study was to characterize the mRNA levels of GABAA receptor subunits (a4, a5, a6 and d) that are key to neurosteroid binding in the brain, following preterm birth. Myelination, measured by the myelin basic protein immunostaining, was used to assess maturity of the preterm brains. Foetal guinea pig brains were obtained at 62 days' gestational age (GA, preterm) or at term (69 days). Neonates were delivered by caesarean section, at 62 days GA and term, and maintained until tissue collection at 24 h of age. Subunit mRNA levels were quantified by RT-PCR in the hippocampus and cerebellum of foetal and neonatal brains. Levels of the a6 and d subunits were markedly lower in the cerebellum of preterm guinea pigs compared with term animals. Importantly, there was an increase in mRNA levels of these subunits during the foetal-to-neonatal transition at term, which was not seen following preterm birth. Myelination was lower in preterm neonatal brains, consistent with marked immaturity. Salivary cortisol concentrations, measured by EIA, were also higher for the preterm neonates, suggesting greater stress. We conclude that there is an adaptive increase in the levels of mRNA of the key GABAA receptor subunits involved in neurosteroid action after term birth, which may compensate for declining allopregnanolone levels. The lower levels of these subunits in preterm neonates may heighten the adverse effect of the premature decline in neurosteroid exposure.

DOI 10.1017/S2040174415000069
Citations Scopus - 30Web of Science - 26
Co-authors Hannah Palliser, Jon Hirst
Show 18 more journal articles

Conference (4 outputs)

Year Citation Altmetrics Link
2023 Moloney RA, Kahl R, Shaw JC, Palliser HK, Hirst J, 'Protection from Oxygen-Glucose Deprivation (OGD) by the TSPO Agonist Etifoxine in Primary Cortical Guinea Pig GABAergic Neurons', REPRODUCTIVE SCIENCES, AUSTRALIA, Brisbane (2023)
Co-authors Hannah Palliser, Jon Hirst
2023 Hanley BA, Crombie GK, Palliser HK, Shaw JC, Hirst JJ, 'The Impact of Chronic Prenatal Stress on GABAergic Inhibitory Pathways in Guinea Pig Frontal Cortex', REPRODUCTIVE SCIENCES, AUSTRALIA, Brisbane (2023)
Co-authors Jon Hirst, Hannah Palliser
2018 Shaw J, Palliser H, Dyson R, Berry M, Hirst J, 'Neurosteroid Therapy Following Preterm Birth in the Guinea Pig Ameliorates Neurological Impairment.', REPRODUCTIVE SCIENCES, San Diego, CA (2018)
Co-authors Jon Hirst, Hannah Palliser
2017 Shaw JC, Palliser HK, Dyson RM, Berry MJ, Hirst JJ, 'Ongoing Defects in Cerebellar Development in a Guinea Pig Model of Preterm Birth.', REPRODUCTIVE SCIENCES, Orlando, FL (2017)
Co-authors Hannah Palliser, Jon Hirst
Show 1 more conference
Edit

Grants and Funding

Summary

Number of grants 14
Total funding $1,188,015

Click on a grant title below to expand the full details for that specific grant.


20231 grants / $14,767

Treating perinatal depression and anxiety (PNDA) to improve maternal well-being and ongoing childhood behavioural deficits$14,767

Funding body: Hunter New England Local Health District

Funding body Hunter New England Local Health District
Project Team Professor Jon Hirst, Doctor Rebecca Glover, Doctor Hannah Palliser, Doctor Julia Shaw
Scheme John Hunter Hospital Charitable Trust Grant
Role Investigator
Funding Start 2023
Funding Finish 2023
GNo G2300292
Type Of Funding C2400 – Aust StateTerritoryLocal – Other
Category 2400
UON Y

20211 grants / $822,403

Neonatal therapy for improving myelination and long term outcome following preterm birth$822,403

Funding body: NHMRC (National Health & Medical Research Council)

Funding body NHMRC (National Health & Medical Research Council)
Project Team Professor Jon Hirst, Doctor Julia Shaw, Doctor Hannah Palliser
Scheme Ideas Grants
Role Investigator
Funding Start 2021
Funding Finish 2023
GNo G2000614
Type Of Funding C1100 - Aust Competitive - NHMRC
Category 1100
UON Y

20202 grants / $59,662

Gut-Brain interactions in the development of Necrotising Enterocolitis$47,500

Funding body: Hunter Medical Research Institute

Funding body Hunter Medical Research Institute
Project Team Doctor Bridie Goggins, Doctor Julia Shaw, Professor Simon Keely, Doctor Aniruddh Deshpande, Doctor Peter Pockney
Scheme Project Grant
Role Investigator
Funding Start 2020
Funding Finish 2022
GNo G2000806
Type Of Funding C3300 – Aust Philanthropy
Category 3300
UON Y

Repurposing therapies to increase inhibitory GABAergic action in the preterm neonatal brain$12,162

Funding body: John Hunter Hospital Charitable Trust

Funding body John Hunter Hospital Charitable Trust
Project Team Doctor Julia Shaw, Doctor Rebecca Glover, Doctor Hannah Palliser
Scheme Research Grant
Role Lead
Funding Start 2020
Funding Finish 2020
GNo G2000308
Type Of Funding C3200 – Aust Not-for Profit
Category 3200
UON Y

20193 grants / $23,563

Restoring nutrients for the premature brain: Improving childhood outcomes following premature birth$20,063

Funding body: John Hunter Hospital Charitable Trust Grant

Funding body John Hunter Hospital Charitable Trust Grant
Project Team

Dr Julia Shaw, Dr Anil Lakkundi, Dr Hannah Palliser, Prof Jon Hirst

Scheme Research Grant
Role Lead
Funding Start 2019
Funding Finish 2019
GNo
Type Of Funding Internal
Category INTE
UON N

Society of Reproductive Investigation Conference Travel 2019$2,000

Funding body: Faculty of Health and Medicine Research Conference Travel Grant

Funding body Faculty of Health and Medicine Research Conference Travel Grant
Scheme Faculty of Health and Medicine Research Conference Travel Grant
Role Lead
Funding Start 2019
Funding Finish 2019
GNo
Type Of Funding Internal
Category INTE
UON N

PRC for Reproductive Science Travel Grant$1,500

Funding body: PRC for Reproductive Science, University of Newcastle

Funding body PRC for Reproductive Science, University of Newcastle
Scheme Travel support
Role Lead
Funding Start 2019
Funding Finish 2019
GNo
Type Of Funding Internal
Category INTE
UON N

20186 grants / $247,620

Neurosteroid analogue therapy to prevent behavioural and neurodevelopmental disorders in children and adolescents born preterm$180,120

Funding body: Neurological Foundation

Funding body Neurological Foundation
Project Team

Berry MJ, Dyson RM, Gray CL, Hirst JJ, Shaw JC

Scheme Neurological Foundation Project Grant
Role Investigator
Funding Start 2018
Funding Finish 2020
GNo
Type Of Funding External
Category EXTE
UON N

Neurosteroid therapy to prevent long-term behavioural deficits following prenatal stress exposure$23,750

Funding body: Hunter Medical Research Institute

Funding body Hunter Medical Research Institute
Project Team Doctor Hannah Palliser, Doctor Julia Shaw, Professor Jon Hirst, Professor Craig Pennell
Scheme Project Grant
Role Investigator
Funding Start 2018
Funding Finish 2018
GNo G1801352
Type Of Funding C3300 – Aust Philanthropy
Category 3300
UON Y

Neurosteroid therapy to prevent longterm behavioural deficits following prenatal stress exposure$23,750

Funding body: Hunter Medical Research Institute (HMRI)

Funding body Hunter Medical Research Institute (HMRI)
Project Team

Dr Hannah Palliser, Dr Julia Shaw, Prof Jon Hirst

Scheme Project Grant
Role Investigator
Funding Start 2018
Funding Finish 2020
GNo
Type Of Funding Internal
Category INTE
UON N

Neurosteroid replacement therapy to prevent neurological impairment following preterm birth$10,000

Funding body: Faculty of Health and Medicine, University of Newcastle

Funding body Faculty of Health and Medicine, University of Newcastle
Project Team

Shaw JC, Palliser HK, Hirst JJ

Scheme Faculty Pilot Grant
Role Lead
Funding Start 2018
Funding Finish 2018
GNo
Type Of Funding Internal
Category INTE
UON N

David-Henderson Smart Travel Scholarship$8,000

Funding body: Perinatal Society of Australia and New Zealand

Funding body Perinatal Society of Australia and New Zealand
Scheme David-Henderson Smart Travel Scholarship
Role Lead
Funding Start 2018
Funding Finish 2019
GNo
Type Of Funding External
Category EXTE
UON N

Society of Reproductive Investigation Conference Travel 2018$2,000

Funding body: Faculty of Health and Medicine, University of Newcastle

Funding body Faculty of Health and Medicine, University of Newcastle
Scheme Travel grant
Role Lead
Funding Start 2018
Funding Finish 2018
GNo
Type Of Funding Internal
Category INTE
UON N

20151 grants / $20,000

Stress in pregnancy and poor neurodevelopmental outcomes in children: the role of miRNA in regulating neurosteroid dysfunction$20,000

Funding body: Hunter Medical Research Institute (HMRI)

Funding body Hunter Medical Research Institute (HMRI)
Project Team

Palliser HK, Shaw JC, Hirst JJ

Scheme Bob and Terry Kennedy Childrens' Research Project Grant
Role Investigator
Funding Start 2015
Funding Finish 2015
GNo
Type Of Funding Internal
Category INTE
UON N
Edit

Research Supervision

Number of supervisions

Completed0
Current2

Current Supervision

Commenced Level of Study Research Title Program Supervisor Type
2020 PhD Neurosteroid Therapy Prevents Neurodevelopmental Deficits to the Dopamine Pathway and Myelination Following Preterm Birth PhD (Experimental Pharmacol), College of Health, Medicine and Wellbeing, The University of Newcastle Co-Supervisor
2020 PhD Neurodevelopmental Impacts of Neurosteroid and Excitatory/Inhibitory Modulation on the Cerebellum Following Preterm Birth Associated Insults PhD (Experimental Pharmacol), College of Health, Medicine and Wellbeing, The University of Newcastle Co-Supervisor
Edit

Dr Julia Shaw

Position

Research Fellow
Mothers and Babies Research Centre
School of Biomedical Sciences and Pharmacy
College of Health, Medicine and Wellbeing

Contact Details

Email julia.c.shaw@newcastle.edu.au
Phone (02) 4042 0485

Office

Room Level 3 East
Building HMRI Building
Location HMRI

,
Edit