Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pediatr Res ; 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773295

RESUMO

BACKGROUND: Understanding changes in blood volume after preterm birth is critical to preventing cardiovascular deterioration in preterm infants. The aims were to determine if blood volume is higher in preterm than term piglets and if blood volume changes in the hours after birth. METHODS: Paired blood volume measurements were conducted in preterm piglets (98/115d gestation, ~28wk gestation infant) at 0.5-5 h (n = 12), 0.5-9 h (n = 44) and 5-11 h (n = 7) after birth, and in a term cohort at 0.5-9 h (n = 40) while under intensive care. RESULTS: At 30 min after birth, blood volume was significantly lower in preterm piglets compared to term piglets. By 9 h after birth, blood volume had reduced by 18% in preterm piglets and 13% in term piglets. By 5-9 h after birth, preterm piglets had significantly lower blood volumes than at term (61 ± 10 vs. 76 ± 11 mL/kg). CONCLUSIONS: In contrast to clinical resources, preterm piglets have a lower blood volume than at term. Substantial reductions in blood volume after birth leave some preterm piglets hypovolemic. If this also occurs in preterm infants, this may have important clinical consequences. Modern studies of blood volume changes after birth are essential for improving preterm outcomes. IMPACT: Preterm piglets do not have a higher blood volume than their term counterparts, in contrast to current clinical estimates. Rapid reduction in blood volume after birth leads to hypovolemia in some preterm piglets. There is a critical need to understand blood volume changes after birth in preterm infants in order to improve clinical management of blood volume.

2.
Sci Rep ; 13(1): 282, 2023 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-36609414

RESUMO

Neuroinflammation is a hallmark of hypoxic-ischemic injury and can be characterized by the activation of glial cells and the expression of inflammatory cytokines and chemokines. Interleukin (IL)-1ß and tumor necrosis factor (TNF)α are among the best-characterized early response cytokines and are often expressed concurrently. Several types of central nervous system cells secrete IL-1ß and TNFα, including microglia, astrocytes, and neurons, and these cytokines convey potent pro-inflammatory actions. Chemokines also play a central role in neuroinflammation by controlling inflammatory cell trafficking. Our aim was to characterise the evolution of early neuroinflammation in the neonatal piglet model of hypoxic-ischemic encephalopathy (HIE). Piglets (< 24 h old) were exposed to HI insult, and recovered to 2, 4, 8, 12 or 24H post-insult. Brain tissue from the frontal cortex and basal ganglia was harvested for assessment of glial cell activation profiles and transcription levels of inflammatory markers in HI piglets with comparison to a control group of newborn piglets. Fluorescence microscopy was used to observe microglia, astrocytes, neurons, degenerating neurons and possibly apoptotic cells, and quantitative polymerase chain reaction was used to measure gene expression of several cytokines and chemokines. HI injury was associated with microglial activation and morphological changes to astrocytes at all time points examined. Gene expression analyses of inflammation-related markers revealed significantly higher expression of pro-inflammatory cytokines tumor necrosis factor-α (TNFα) and interleukin 1 beta (IL-1ß), chemokines cxc-chemokine motif ligand (CXCL)8 and CXCL10, and anti-inflammatory cytokine transforming growth factor (TGF)ß in every HI group, with some region-specific differences noted. No significant difference was observed in the level of C-X-C chemokine receptor (CCR)5 over time. This high degree of neuroinflammation was associated with a reduction in the number of neurons in piglets at 12H and 24H in the frontal cortex, and the putamen at 12H. This reduction of neurons was not associated with increased numbers of degenerating neurons or potentially apoptotic cells. HI injury triggered a robust early neuroinflammatory response associated with a reduction in neurons in cortical and subcortical regions in our piglet model of HIE. This neuroinflammatory response may be targeted using novel therapeutics to reduce neuropathology in our piglet model of neonatal HIE.


Assuntos
Citocinas , Hipóxia-Isquemia Encefálica , Animais , Suínos , Citocinas/metabolismo , Animais Recém-Nascidos , Fator de Necrose Tumoral alfa/metabolismo , Doenças Neuroinflamatórias , Neuroglia/metabolismo , Encéfalo/metabolismo , Hipóxia/metabolismo , Microglia/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Fator de Crescimento Transformador beta/metabolismo , Inflamação/patologia
3.
Mol Neurobiol ; 59(2): 1018-1040, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34825315

RESUMO

The developing brain is particularly vulnerable to foetal growth restriction (FGR) and abnormal neurodevelopment is common in the FGR infant ranging from behavioural and learning disorders to cerebral palsy. No treatment exists to protect the FGR newborn brain. Recent evidence suggests inflammation may play a key role in the mechanism responsible for the progression of brain impairment in the FGR newborn, including disruption to the neurovascular unit (NVU). We explored whether ibuprofen, an anti-inflammatory drug, could reduce NVU disruption and brain impairment in the FGR newborn. Using a preclinical FGR piglet model, ibuprofen was orally administered for 3 days from birth. FGR brains demonstrated a proinflammatory state, with changes to glial morphology (astrocytes and microglia), and blood-brain barrier disruption, assessed by IgG and albumin leakage into the brain parenchyma and a decrease in blood vessel density. Loss of interaction between astrocytic end-feet and blood vessels was evident where plasma protein leakage was present, suggestive of structural deficits to the NVU. T-cell infiltration was also evident in the parenchyma of FGR piglet brains. Ibuprofen treatment reduced the pro-inflammatory response in FGR piglets, reducing the number of activated microglia and enhancing astrocyte interaction with blood vessels. Ibuprofen also attenuated plasma protein leakage, regained astrocytic end-feet interaction around vessels, and decreased T-cell infiltration into the FGR brain. These findings suggest postnatal administration of ibuprofen modulates the inflammatory state, allowing for stronger interaction between vasculature and astrocytic end-feet to restore NVU integrity. Modulation of the NVU improves the FGR brain microenvironment and may be key to neuroprotection.


Assuntos
Encéfalo , Ibuprofeno , Animais , Astrócitos/metabolismo , Encéfalo/metabolismo , Humanos , Ibuprofeno/farmacologia , Ibuprofeno/uso terapêutico , Microglia , Neuroglia , Suínos
4.
NPJ Regen Med ; 6(1): 75, 2021 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-34795316

RESUMO

The foetal brain is particularly vulnerable to the detrimental effects of foetal growth restriction (FGR) with subsequent abnormal neurodevelopment being common. There are no current treatments to protect the FGR newborn from lifelong neurological disorders. This study examines whether pure foetal mesenchymal stromal cells (MSC) and endothelial colony-forming cells (ECFC) from the human term placenta are neuroprotective through modulating neuroinflammation and supporting the brain vasculature. We determined that one dose of combined MSC-ECFCs (cECFC; 106 ECFC 106 MSC) on the first day of life to the newborn FGR piglet improved damaged vasculature, restored the neurovascular unit, reduced brain inflammation and improved adverse neuronal and white matter changes present in the FGR newborn piglet brain. These findings could not be reproduced using MSCs alone. These results demonstrate cECFC treatment exerts beneficial effects on multiple cellular components in the FGR brain and may act as a neuroprotectant.

5.
Mol Neurobiol ; 57(10): 4322-4344, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32720074

RESUMO

Hypoxic ischemic encephalopathy (HIE) is the most frequent cause of acquired infant brain injury. Early, clinically relevant biomarkers are required to allow timely application of therapeutic interventions. We previously reported early alterations in several microRNAs (miRNA) in umbilical cord blood at birth in infants with HIE. However, the exact timing of these alterations is unknown. Here, we report serial changes in six circulating, cross-species/bridging biomarkers in a clinically relevant porcine model of neonatal HIE with functional analysis. Six miRNAs-miR-374a, miR-181b, miR-181a, miR-151a, miR-148a and miR-128-were significantly and rapidly upregulated 1-h post-HI. Changes in miR-374a, miR-181b and miR-181a appeared specific to moderate-severe HI. Histopathological injury and five miRNAs displayed positive correlations and were predictive of MRS Lac/Cr ratios. Bioinformatic analysis identified that components of the bone morphogenic protein (BMP) family may be targets of miR-181a. Inhibition of miR-181a increased neurite length in both SH-SY5Y cells at 1 DIV (days in vitro) and in primary cultures of rat neuronal midbrain at 3 DIV. In agreement, inhibition of miR-181a increased expression of BMPR2 in differentiating SH-SY5Y cells. These miRNAs may therefore act as early biomarkers of HIE, thereby allowing for rapid diagnosis and timely therapeutic intervention and may regulate expression of signalling pathways vital to neuronal survival.


Assuntos
Lesões Encefálicas/genética , Regulação da Expressão Gênica , Hipóxia-Isquemia Encefálica/genética , MicroRNAs/genética , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Encéfalo/patologia , Lesões Encefálicas/sangue , Creatinina/metabolismo , Modelos Animais de Doenças , Sangue Fetal/metabolismo , Perfilação da Expressão Gênica , Humanos , Hipóxia-Isquemia Encefálica/sangue , Recém-Nascido , Ácido Láctico/metabolismo , Modelos Lineares , Espectroscopia de Ressonância Magnética , MicroRNAs/metabolismo , Neuritos/metabolismo , Especificidade de Órgãos , Transdução de Sinais/genética , Suínos , Fatores de Tempo
6.
J Neuroinflammation ; 16(1): 5, 2019 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-30621715

RESUMO

BACKGROUND: The fetal brain is particularly vulnerable to intrauterine growth restriction (IUGR) conditions evidenced by neuronal and white matter abnormalities and altered neurodevelopment in the IUGR infant. To further our understanding of neurodevelopment in the newborn IUGR brain, clinically relevant models of IUGR are required. This information is critical for the design and implementation of successful therapeutic interventions to reduce aberrant brain development in the IUGR newborn. We utilise the piglet as a model of IUGR as growth restriction occurs spontaneously in the pig as a result of placental insufficiency, making it a highly relevant model of human IUGR. The purpose of this study was to characterise neuropathology and neuroinflammation in the neonatal IUGR piglet brain. METHODS: Newborn IUGR (< 5th centile) and normally grown (NG) piglets were euthanased on postnatal day 1 (P1; < 18 h) or P4. Immunohistochemistry was utilised to examine neuronal, white matter and inflammatory responses, and PCR for cytokine analysis in parietal cortex of IUGR and NG piglets. RESULTS: The IUGR piglet brain displayed less NeuN-positive cells and reduced myelination at both P1 and P4 in the parietal cortex, indicating neuronal and white matter disruption. A concurrent decrease in Ki67-positive proliferative cells and increase in cell death (caspase-3) in the IUGR piglet brain was also apparent on P4. We observed significant increases in the number of both Iba-1-positive microglia and GFAP-positive astrocytes in the white matter in IUGR piglet brain on both P1 and P4 compared with NG piglets. These increases were associated with a change in activation state, as noted by altered glial morphology. This inflammatory state was further evident with increased expression levels of proinflammatory cytokines (interleukin-1ß, tumour necrosis factor-α) and decreased levels of anti-inflammatory cytokines (interleukin-4 and -10) observed in the IUGR piglet brains. CONCLUSIONS: These findings suggest that the piglet model of IUGR displays the characteristic neuropathological outcomes of neuronal and white matter impairment similar to those reported in the IUGR human brain. The activated glial morphology and elevated proinflammatory cytokines is indicative of an inflammatory response that may be associated with neuronal damage and white matter disruption. These findings support the use of the piglet as a pre-clinical model for studying mechanisms of altered neurodevelopment in the IUGR newborn.


Assuntos
Citocinas/metabolismo , Encefalite/etiologia , Retardo do Crescimento Fetal/patologia , Retardo do Crescimento Fetal/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neuroglia/patologia , Animais , Animais Recém-Nascidos , Proteínas de Ligação ao Cálcio , Caspase 3/metabolismo , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Proteínas dos Microfilamentos , Neuroglia/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Suínos , Substância Branca/patologia
7.
Clin Exp Pharmacol Physiol ; 46(3): 274-279, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30347457

RESUMO

Preterm infants are at higher risk of adverse neurodevelopmental outcomes. Inadequate cerebral oxygen delivery resulting from poor cardiovascular function is likely to be a significant contributor to preterm brain injury. In this context, improved support of cardiovascular function is integral to improving preterm outcomes. Many of the treatments used to support preterm cardiovascular function are based on adult physiology and may not be appropriate for the unique physiology of the preterm infant. The preterm heart is structurally immature with reduced contractility and low cardiac output. However, there is limited evidence that inotropic support with dopamine and/or dobutamine is effective in preterm babies. Hypovolemia may also contribute to poor preterm cardiovascular function; there is evidence that capillary leakage results in considerable loss of plasma from the circulation of newborn preterm babies. In addition, the vasoconstrictor response to acute stimuli does not develop until quite late in gestation and is limited in the preterm infant. This may lead to inappropriate vasodilatation adding to functional hypovolemia. The first line treatment for hypotension in preterm infants is volume expansion with crystalloid solutions, but this has limited efficacy in the preterm infant. More effective methods of volume expansion are required. Effective support of preterm cardiovascular function requires better understanding of preterm cardiovascular physiology so that treatments can target mechanisms that are sufficiently mature to respond.


Assuntos
Fenômenos Fisiológicos Cardiovasculares , Recém-Nascido Prematuro/fisiologia , Volume Sanguíneo , Humanos
9.
Brain Struct Funct ; 223(2): 1025-1033, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29282556

RESUMO

The GABAA receptor provides the majority of inhibitory neurotransmission in the adult central nervous system but in immature brain is responsible for much of the excitatory drive, a requirement for normal brain development. It is well established that GABAA receptor subunit expression changes across the course of brain development. In the present study, we have identified a splice variant of the GABAA receptor α3 subunit which appears unique to the developing brain, referred to here as the GABAA receptor α3 subunit neonatal variant (GABAA receptor α3N). RT-PCR and sequence analysis revealed splicing of exon 8 of the α3 subunit. Western blot analysis showed expression of GABAA receptor α3N in the cortex of several neonatal species and significantly reduced expression of this splice variant in the corresponding adult brains. Expression was evident in multiple brain regions and decreased across development in the pig. Fractionation revealed differential cellular localisation in the parietal cortex, hippocampus and thalamus of the full-length GABAA receptor α3 and GABAA receptor α3N. Immunoprecipitation showed direct interaction with the GABAA receptor subunits α1 and γ2 but not with gephyrin.


Assuntos
Encéfalo/citologia , Neurônios/metabolismo , Receptores de GABA-A/metabolismo , Fatores Etários , Animais , Encéfalo/anatomia & histologia , Fracionamento Celular , Humanos , Imunoprecipitação , Modelos Moleculares , Suínos
10.
Dev Neurosci ; 39(5): 375-385, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28472809

RESUMO

GABA is a major neurotransmitter in the mammalian brain. In the mature brain GABA exerts inhibitory actions via the GABAA receptor (GABAAR); however, in the immature brain GABA provides much of the excitatory drive. We examined the expression of 3 predominant GABAA α-subunit proteins in the pig brain at various pre- and postnatal ages. Brain tissue was collected from piglets born via caesarean section at preterm ages 91, 97, 100, and 104 days' gestational age (GA), at term equivalent (114 days' GA, caesarean section) and at term, postnatal day 0 (P0) (spontaneous delivery, term = 115 days). Tissue was obtained from piglets at P4 and P7. Adult tissue from sows was collected postmortem after caesarean section. In all cortical regions and basal ganglia (1) α3 exhibited a significant increase in protein expression at 100 days' GA, (2) α3 expression decreased with age after 100 days' GA, (3) α1 increased with age, with peak expression at P7 in cortices, hippocampus, and thalamus, and (4) α2 protein expression remained relatively constant across the ages examined. The subunit expression of α3 was most abundant at preterm ages, with α1 the predominant subunit expressed postnatally. Immunofluorescent labelling revealed α1 expression on the somatic membranes of pyramidal cells in the cortex and hippocampus, and in the cerebellar Purkinje cells. Positive α3 labelling was apparent on interneurones in the cortex and hippocampus. The switch between dominant α-subunits may coincide with the functional change in GABAergic neurotransmission from excitation to inhibition. Brain growth in the pig closely reflects that in the term human, making the pig a valuable non-primate model for studying development and the effects of insults on the perinatal brain.


Assuntos
Cerebelo/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Receptores de GABA-A/metabolismo , Animais , Animais Recém-Nascidos , Idade Gestacional , Sus scrofa , Suínos
12.
Int J Dev Neurosci ; 59: 1-9, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28219764

RESUMO

Intrauterine growth restriction (IUGR) is one of the most common causes of perinatal mortality and morbidity. White matter and neuronal injury are major pathophysiological features of the IUGR neonatal brain. GABAA (γ-aminobutyric acid type A) receptors have been shown to play a role in oligodendrocyte differentiation and proliferation in the neonatal brain and may be a key factor in white matter injury and myelination in IUGR neonates. Whether there are impairments to the GABAergic system and neuronal cytoskeleton in IUGR brain has yet to be elucidated. This study aims to examine GABAA receptor α1 and α3 subunit protein expression and distribution in parietal cortex and hippocampus of the IUGR piglet at four different ages (term=115d - days gestational age), 100d, 104d, birth (postnatal day 0-P0) and P7 and to examine neuronal and myelination patterns. Significant alterations to GABAA receptor α1 and α3 protein expression levels were observed in the IUGR piglet brain of P7 IUGR piglets with significantly greater α3 expression compared to α1 expression in the hippocampus while there was virtually no difference between the two subunits in the parietal cortex. However a significantly lower α1/α3 ratio was evident in P7 IUGR cortex when compared with P7 NG cortex. Neuronal somatodendrites studied using MAP2 immunohistochemistry showed reduced and disrupted somatodendrites while MBP immunolabelling showed loss of axonal fibres from gestational day 104d through to P7. These findings provide insights into the effects of IUGR on the development of the GABA system, altered developmental maturation of GABAA receptor subunit expression in the IUGR brain may influence myelination and may partly explain the cognitive disabilities observed in IUGR. Understanding the mechanisms behind grey and white matter injury in the IUGR infant is essential to identifying targets for treatments to improve long-term outcomes for IUGR infants.


Assuntos
Encéfalo/patologia , Retardo do Crescimento Fetal/patologia , Retardo do Crescimento Fetal/fisiopatologia , Receptores de GABA-A/metabolismo , Substância Branca/metabolismo , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Peso Corporal/fisiologia , Encéfalo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteína Básica da Mielina/metabolismo , Tamanho do Órgão/fisiologia , Suínos
13.
Placenta ; 54: 111-116, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27939102

RESUMO

While placental function is fundamental to normal fetal development, the blood-brain barrier provides a second checkpoint critical to protecting the fetal brain and ensuring healthy brain development. The placenta is considered the key barrier between the mother and fetus, regulating delivery of essential nutrients, removing waste as well as protecting the fetus from potentially noxious substances. However, disturbances to the maternal environment and subsequent adaptations to placental function may render the placenta ineffective for providing a suitable environment for the developing fetus and to providing sufficient protection from harmful substances. The developing brain is particularly vulnerable to changes in the maternal/fetal environment. Development of the blood-brain barrier and maturation of barrier transporter systems work to protect the fetal brain from exposure to drugs, excluding them from the fetal CNS. This review will focus on the role of the 'other' key barrier during gestation - the blood-brain barrier - which has been shown to be functional as early as 8 weeks' gestation.


Assuntos
Barreira Hematoencefálica/crescimento & desenvolvimento , Desenvolvimento Fetal , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Feminino , Humanos , Placenta/metabolismo , Gravidez , Proteínas Carreadoras de Solutos/metabolismo
14.
J Neurochem ; 139(3): 471-484, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27456541

RESUMO

Seizures are a common manifestation of hypoxic-ischaemic brain injury in the neonate. In status epilepticus models alterations to GABAA R subunit expression have been suggested to contribute to (i) abnormal development of the GABAergic system, (ii) why seizures become self-sustaining and (iii) the development of pharmacoresistance. Detailed investigation of GABAA R subunit protein expression after neonatal hypoxia-ischaemia (HI) is currently insufficient. Using our pig model of HI and subsequent spontaneous neonatal seizures, we investigated changes in protein expression of the three predominant α-subunits of the GABAA R; α1 , α2 and α3 . Anaesthetized, ventilated newborn pigs (< 24 h old) were subjected to 30 min HI and subsequently recovered to 24 or 72 h. Amplitude-integrated electroencephalography was used to monitor brain activity and identify seizure activity. Brain tissue was collected post-mortem and GABAA R α-subunit protein expression was analysed using western blot and immunohistochemistry. GABAA R α1 and α3 protein expression was significantly reduced in animals that developed seizures after HI; HI animals that did not develop seizures did not exhibit the same reductions. Immunohistochemistry revealed decreased α1 and α3 expression, and α1 redistribution from the cell membrane to the cytosol, in the hippocampus of seizure animals. Multivariate analyses, controlling for HI severity and neuronal injury, revealed that seizures were independently associated with significant GABAA R α3 reduction. This is the first study to show loss and redistribution of GABAA R α-subunits in a neonatal brain experiencing seizures. Our findings are similar to those reported in models of SE and in chronic epilepsy.


Assuntos
Hipóxia-Isquemia Encefálica/metabolismo , Receptores de GABA-A/metabolismo , Convulsões/metabolismo , Animais , Animais Recém-Nascidos , Comportamento Animal , Química Encefálica , Eletroencefalografia/efeitos dos fármacos , Feminino , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Respiração Artificial , Convulsões/psicologia , Suínos
15.
J Am Chem Soc ; 135(39): 14568-73, 2013 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-23998633

RESUMO

Aspirin (acetylsalicylic acid) is widely used for the acute treatment of inflammation and the management of cardiovascular disease. More recently, it has also been shown to reduce the risk of a variety of cancers. The anti-inflammatory properties of aspirin in pain-relief, cardio-protection, and chemoprevention are well-known to result from the covalent inhibition of cyclooxygenase enzymes through nonenzymatic acetylation of key serine residues. However, any additional molecular mechanisms that may contribute to the beneficial effects of aspirin remain poorly defined. Interestingly, studies over the past 50 years using radiolabeled aspirin demonstrated that other proteins are acetylated by aspirin and enrichment with antiacetyl-lysine antibodies identified 33 potential targets of aspirin-dependent acetylation. Herein we describe the development of an alkyne-modified aspirin analogue (AspAlk) as a chemical reporters of aspirin-dependent acetylation in living cells. When combined with the Cu(I)-catalyzed [3 + 2] azide-alkyne cycloaddition, this chemical reporter allowed for the robust in-gel fluorescent detection of acetylation and the subsequent enrichment and identification of 120 proteins, 112 of which have not been previously reported to be acetylated by aspirin in cellular or in vivo contexts. Finally, AspAlk was shown to modify the core histone proteins, implicating aspirin as a potential chemical-regulator of transcription.


Assuntos
Acetilação/efeitos dos fármacos , Alcinos/química , Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/análogos & derivados , Aspirina/farmacologia , Proteínas/metabolismo , Animais , Azidas/química , Linhagem Celular , Linhagem Celular Tumoral , Corantes Fluorescentes/química , Humanos
16.
PLoS One ; 8(7): e68763, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874755

RESUMO

BACKGROUND: Large animal models are an essential tool in the development of rationally-based new clinical therapies for preterm infants. We provide a description of the newborn pig as a model of the preterm neonate in terms of growth parameters, physiology and the requirement for intensive care over a range of gestational ages. METHODS: Twenty-nine litters of piglets (n = 298) were delivered by caesarean section at six timepoints during gestation from 91d to 113d (term = 115d). Two groups, at 91 and 97d gestation, also received maternal glucocorticoid treatment. At four of these timepoints, piglets (n = 79) were ventilated, sedated and monitored using standard neonatal intensive care techniques for up to 8 h in various experimental protocols. RESULTS: Body weight increased from mean 697 g (SD 193) at 91d gestation to 1331 g (SD 368) at 113d gestation. Piglets delivered at 97d gestation were able to be resuscitated and kept alive for at least 8 h on respiratory support after surfactant administration. Maternal glucocorticoid treatment 48 h and 24 h hours prior to delivery reduced the requirement for ventilator support and improved cardiovascular stability. CONCLUSION: The pig provides a relevant model for the study of human preterm physiology and for investigation of novel therapies to improve outcomes.


Assuntos
Animais Recém-Nascidos , Antropometria , Recém-Nascido Prematuro , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Pressão Sanguínea , Cesárea , Feminino , Idade Gestacional , Glucocorticoides/administração & dosagem , Humanos , Recém-Nascido , Masculino , Exposição Materna , Suínos
17.
Int J Sport Nutr Exerc Metab ; 22(2): 131-8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22465866

RESUMO

PURPOSE: Prevention of the female athlete triad is essential to protect female athletes' health. The aim of this study was to investigate the knowledge, attitudes, and behaviors of regularly exercising adult women in Australia toward eating patterns, menstrual cycles, and bone health. METHODS: A total of 191 female exercisers, age 18-40 yr, engaging in ≥2 hr/wk of strenuous activity, completed a survey. After 11 surveys were excluded (due to incomplete answers), the 180 participants were categorized into lean-build sports (n = 82; running/athletics, triathlon, swimming, cycling, dancing, rowing), non-lean-build sports (n = 94; basketball, netball, soccer, hockey, volleyball, tennis, trampoline, squash, Australian football), or gym/fitness activities (n = 4). RESULTS: Mean (± SD) training volume was 9.0 ± 5.5 hr/wk, with participants competing from local up to international level. Only 10% of respondents could name the 3 components of the female athlete triad. Regardless of reported history of stress fracture, 45% of the respondents did not think that amenorrhea (absence of menses for ≥3 months) could affect bone health, and 22% of those involved in lean-build sports would do nothing if experiencing amenorrhea (vs. 3.2% in non-lean-build sports, p = .005). Lean-build sports, history of amenorrhea, and history of stress fracture were all significantly associated with not taking action in the presence of amenorrhea (all p < .005). CONCLUSIONS: Few active Australian women are aware of the detrimental effects of menstrual dysfunction on bone health. Education programs are needed to prevent the female athlete triad and ensure that appropriate actions are taken by athletes when experiencing amenorrhea.


Assuntos
Osso e Ossos/fisiologia , Exercício Físico/fisiologia , Síndrome da Tríade da Mulher Atleta , Fraturas de Estresse , Conhecimentos, Atitudes e Prática em Saúde , Menstruação , Esportes/fisiologia , Adolescente , Adulto , Amenorreia , Atletas , Transtornos da Alimentação e da Ingestão de Alimentos , Feminino , Inquéritos Epidemiológicos , Humanos , Osteoporose , Adulto Jovem
18.
Neurochem Res ; 37(11): 2364-78, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22528834

RESUMO

Glial fibrillary acidic protein (GFAP) is an intermediate filament protein expressed in the astrocyte cytoskeleton that plays an important role in the structure and function of the cell. GFAP can be phosphorylated at six serine (Ser) or threonine (Thr) residues but little is known about the role of GFAP phosphorylation in physiological and pathophysiological states. We have generated antibodies against two phosphorylated GFAP (pGFAP) proteins: p8GFAP, where GFAP is phosphorylated at Ser-8 and p13GFAP, where GFAP is phosphorylated at Ser-13. We examined p8GFAP and p13GFAP expression in the control neonatal pig brain and at 24 and 72 h after an hypoxic-ischemic (HI) insult. Immunohistochemistry demonstrated pGFAP expression in astrocytes with an atypical cytoskeletal morphology, even in control brains. Semi-quantitative western blotting revealed that p8GFAP expression was significantly increased at 24 h post-insult in HI animals with seizures in frontal, parietal, temporal and occipital cortices. At 72 h post-insult, p8GFAP and p13GFAP expression were significantly increased in HI animals with seizures in brain regions that are vulnerable to cellular damage (cortex and basal ganglia), but no changes were observed in brain regions that are relatively spared following an HI insult (brain stem and cerebellum). Increased pGFAP expression was associated with poor neurological outcomes such as abnormal encephalography and neurobehaviour, and increased histological brain damage. Phosphorylation of GFAP may play an important role in astrocyte remodelling during development and disease and could potentially contribute to the plasticity of the central nervous system.


Assuntos
Animais Recém-Nascidos , Proteína Glial Fibrilar Ácida/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Animais , Western Blotting , Eletroencefalografia , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Imuno-Histoquímica , Fosforilação , Suínos
19.
Pharmacol Biochem Behav ; 100(1): 130-7, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21851834

RESUMO

Postpartum female rodents are less anxious than diestrous virgins and this difference contributes to dams' ability to adequately care for pups and defend the nest. Low postpartum anxiety has been observed in many behavioral paradigms but the results of previous studies using the light-dark box have been inconsistent. We here reexamined the usefulness of the light-dark box to assess differences between postpartum and diestrous virgin female rats in their anxiety-related behavior. We found a significant effect of reproductive state, such that dams spent more time in the light chamber than did diestrous virgins. This difference required recent physical contact with pups because a four-hour separation from pups reduced dams' time spent in the light chamber by half, similar to what we previously found for litter-separated dams tested in an elevated plus maze. We then examined if dams' low-anxiety behavior in the light-dark box depends on high GABA(A) receptor activity by inhibiting the receptor at different binding sites using (+)-Bicuculline to target the GABA site, FG-7142 to target the benzodiazepine site, and pentylenetetrazol to target the picrotoxin site. Only pentylenetetrazol was consistently anxiogenic in dams, while having little effect in diestrous virgins. Thus, the light-dark box can be a useful paradigm to study differences between postpartum and diestrous virgin female rats in their anxiety-related behaviors, and this difference is influenced by dams' recent contact with pups and GABA(A) receptor neurotransmission particularly affected by activity at the picrotoxin site.


Assuntos
Ansiedade/diagnóstico , Ansiedade/psicologia , Escuridão , Luz , Período Pós-Parto/psicologia , Comportamento Sexual Animal , Animais , Escuridão/efeitos adversos , Feminino , Luz/efeitos adversos , Masculino , Atividade Motora/fisiologia , Período Pós-Parto/fisiologia , Gravidez , Ratos , Ratos Long-Evans , Comportamento Sexual Animal/fisiologia
20.
Brain Res Bull ; 86(1-2): 60-4, 2011 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-21664440

RESUMO

Postpartum female rats exhibit a suppression of anxiety-related behaviors when compared to diestrous virgin females, pregnant females, and males. This blunted anxiety promotes optimal maternal care and involves elevated GABA neurotransmission, possibly including greater density of GABA(A) and benzodiazepine receptors in the postpartum brain. We here examined autoradiographic binding of [(3)H]muscimol to measure the total population of GABA(A) receptors and [(3)H]flunitrazepam to assess density of benzodiazepine sites in the medial prefrontal cortex, bed nucleus of the stria terminalis, amygdala, hippocampus, and periaqueductal gray of female rats sacrificed on day 7 postpartum, day 10 of pregnancy, or as diestrous virgins. A group of sexually naïve male rats was also included. We found that [(3)H]muscimol binding did not differ among groups in any site but that diestrous virgin females had greater [(3)H]flunitrazepam binding in the CA1 and dentate gyrus of the hippocampus compared to mid-pregnant females and males. Notably, postpartum and diestrous virgin females did not significantly differ in binding of either ligand in any site examined. This is the first study to evaluate the densities of GABA(A) and benzodiazepine binding sites simultaneously across three female reproductive states and sex with a focus on brain sites influencing anxiety-related behaviors. The results suggest that changes in other GABA(A) receptor characteristics such as subunit composition, or increased presynaptic GABA release during interactions with offspring, must instead play a greater role in the postpartum suppression of anxiety in laboratory rats.


Assuntos
Ansiedade/fisiopatologia , Rede Nervosa/fisiologia , Período Pós-Parto/metabolismo , Gravidez/metabolismo , Receptores de GABA-A/metabolismo , Animais , Ansiolíticos/química , Ansiolíticos/metabolismo , Autorradiografia , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Feminino , Flunitrazepam/química , Flunitrazepam/metabolismo , Agonistas de Receptores de GABA-A/química , Agonistas de Receptores de GABA-A/metabolismo , Humanos , Masculino , Muscimol/química , Muscimol/metabolismo , Rede Nervosa/anatomia & histologia , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...