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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Pediatr Res ; 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773295

RESUMEN

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.
J Neuroinflammation ; 16(1): 5, 2019 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-30621715

RESUMEN

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.


Asunto(s)
Citocinas/metabolismo , Encefalitis/etiología , Retardo del Crecimiento Fetal/patología , Retardo del Crecimiento Fetal/fisiopatología , Regulación del Desarrollo de la Expresión Génica/fisiología , Neuroglía/patología , Animales , Animales Recién Nacidos , Proteínas de Unión al Calcio , Caspasa 3/metabolismo , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Proteínas de Microfilamentos , Neuroglía/metabolismo , Embarazo , ARN Mensajero/metabolismo , Porcinos , Sustancia Blanca/patología
3.
Clin Exp Pharmacol Physiol ; 46(3): 274-279, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30347457

RESUMEN

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.


Asunto(s)
Fenómenos Fisiológicos Cardiovasculares , Recien Nacido Prematuro/fisiología , Volumen Sanguíneo , Humanos
4.
Dev Neurosci ; 39(5): 375-385, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28472809

RESUMEN

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.


Asunto(s)
Cerebelo/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Receptores de GABA-A/metabolismo , Animales , Animales Recién Nacidos , Edad Gestacional , Sus scrofa , Porcinos
5.
J Neurochem ; 139(3): 471-484, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27456541

RESUMEN

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.


Asunto(s)
Hipoxia-Isquemia Encefálica/metabolismo , Receptores de GABA-A/metabolismo , Convulsiones/metabolismo , Animales , Animales Recién Nacidos , Conducta Animal , Química Encefálica , Electroencefalografía/efectos de los fármacos , Femenino , Hipocampo/metabolismo , Inmunohistoquímica , Masculino , Respiración Artificial , Convulsiones/psicología , Porcinos
6.
J Am Chem Soc ; 135(39): 14568-73, 2013 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-23998633

RESUMEN

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.


Asunto(s)
Acetilación/efectos de los fármacos , Alquinos/química , Antiinflamatorios no Esteroideos/farmacología , Aspirina/análogos & derivados , Aspirina/farmacología , Proteínas/metabolismo , Animales , Azidas/química , Línea Celular , Línea Celular Tumoral , Colorantes Fluorescentes/química , Humanos
8.
Sci Rep ; 13(1): 282, 2023 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-36609414

RESUMEN

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.


Asunto(s)
Citocinas , Hipoxia-Isquemia Encefálica , Animales , Porcinos , Citocinas/metabolismo , Animales Recién Nacidos , Factor de Necrosis Tumoral alfa/metabolismo , Enfermedades Neuroinflamatorias , Neuroglía/metabolismo , Encéfalo/metabolismo , Hipoxia/metabolismo , Microglía/metabolismo , Hipoxia-Isquemia Encefálica/patología , Factor de Crecimiento Transformador beta/metabolismo , Inflamación/patología
9.
Neurochem Res ; 37(11): 2364-78, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22528834

RESUMEN

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.


Asunto(s)
Animales Recién Nacidos , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Animales , Western Blotting , Electroencefalografía , Hipoxia-Isquemia Encefálica/patología , Hipoxia-Isquemia Encefálica/fisiopatología , Inmunohistoquímica , Fosforilación , Porcinos
10.
Int J Sport Nutr Exerc Metab ; 22(2): 131-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22465866

RESUMEN

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.


Asunto(s)
Huesos/fisiología , Ejercicio Físico/fisiología , Síndrome de la Tríada de la Atleta Femenina , Fracturas por Estrés , Conocimientos, Actitudes y Práctica en Salud , Menstruación , Deportes/fisiología , Adolescente , Adulto , Amenorrea , Atletas , Trastornos de Alimentación y de la Ingestión de Alimentos , Femenino , Encuestas Epidemiológicas , Humanos , Osteoporosis , Adulto Joven
11.
Mol Neurobiol ; 59(2): 1018-1040, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34825315

RESUMEN

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.


Asunto(s)
Encéfalo , Ibuprofeno , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Humanos , Ibuprofeno/farmacología , Ibuprofeno/uso terapéutico , Microglía , Neuroglía , Porcinos
12.
Dev Neurosci ; 33(2): 99-109, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21613774

RESUMEN

The principal function of the γ-aminobutyric acid (GABA) system in the adult brain is inhibition; however, in the neonatal brain, GABA provides much of the excitatory drive. As the brain develops, transmembrane chloride gradients change and the inhibitory role of GABA is initiated and continues throughout juvenile and adult life. Previous studies have shown that GABA(A) receptor subunit expression is developmentally regulated, and it is thought that the change in GABA function from excitation to inhibition corresponds to the changeover in expression of 'immature' to 'mature' subunit isoforms. We examined the protein expression pattern and distribution of GABA type A (GABA(A)) receptor α1-, α3- and ß2-subunits in the parietal cortex and hippocampus of the developing piglet brain. Four perinatal ages were studied; 14 days preterm (P-14), 10 days preterm (P-10), day of birth (P0) and at postnatal day 7 (P7). Animals were obtained by either caesarean section or spontaneous birth. Protein expression levels and subunit localization were analysed by Western blotting and immunohistochemistry, respectively. In the cortex and hippocampus, GABA(A) receptor α1-subunit showed greatest expression at P7 when compared to all other age groups (p < 0.05). In contrast, α3 expression in the cortex was elevated in preterm brain, peaking at P0, followed by a significant reduction by P7 (p < 0.05); a similar trend was observed in the hippocampus. GABA(A) receptor ß2-subunit protein expression appeared relatively constant across all time points studied in both the cortex and hippocampus. Immunolabelling of the α1-, α3- and ß2-subunits was observed throughout all cortical layers at every age. GABA(A) receptor α3-subunit appeared to show specific localization to layers V and VI whilst labelling for the ß2-subunit was observed in layer IV. In the hippocampus of all animals, the α1- and ß2-subunits were shown to immunolabel various cells and processes in the dentate gyrus (DG), CA1 and CA3; the α3-subunit was barely observed except at the stratum moleculare of the DG. We report for the first time the ontogenesis of GABA(A) receptor subunits α1, α3 and ß2 in the perinatal pig brain.


Asunto(s)
Encéfalo/metabolismo , Hipocampo , Lóbulo Parietal , Subunidades de Proteína/metabolismo , Receptores de GABA-A , Animales , Animales Recién Nacidos/metabolismo , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Edad Gestacional , Hipocampo/embriología , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Humanos , Inmunohistoquímica , Lóbulo Parietal/embriología , Lóbulo Parietal/crecimiento & desarrollo , Lóbulo Parietal/metabolismo , Ratas , Receptores de GABA-A/análisis , Receptores de GABA-A/metabolismo , Sus scrofa , Factores de Tiempo , Distribución Tisular , Ácido gamma-Aminobutírico/metabolismo
13.
NPJ Regen Med ; 6(1): 75, 2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34795316

RESUMEN

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.

14.
Glia ; 58(2): 181-94, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19606499

RESUMEN

Astrocytes play a vital role in the brain; their structural integrity and sustained function are essential for neuronal viability, especially after injury or insult. In this study, we have examined the response of astrocytes to hypoxia/ischemia (H/I), employing multiple methods (immunohistochemistry, iontophoretic cell injection, Golgi-Kopsch staining, and D-aspartate uptake) in a neonatal pig model of H/I. We have identified morphological changes in cortical gray matter astrocytes in response to H/I. Initial astrocytic changes were evident as early as 8 h post-insult, before histological evidence for neuronal damage. By 72 h post-insult, astrocytes exhibited significantly fewer processes that were shorter, thicker, and had abnormal terminal swellings, compared with astrocytes from control brains that exhibited a complex structure with multiple fine branching processes. Quantification and image analysis of astrocytes at 72 h post-insult revealed significant decreases in the average astrocyte size, from 686 microm(2) in controls to 401 microm(2) in H/I brains. Sholl analysis revealed a significant decrease (>60%) in the complexity of astrocyte branching between 5 and 20 microm from the cell body. D-Aspartate uptake studies revealed that the H/I insult resulted in impaired astrocyte function, with significantly reduced clearance of the glutamate analog, D-aspartate. These results suggest that astrocytes may be involved in the pathophysiological events of H/I brain damage at a far earlier time point than first thought. Developing therapies that prevent or reverse these astrocytic changes may potentially improve neuronal survival and thus might be a useful strategy to minimize brain damage after an H/I insult.


Asunto(s)
Astrocitos/patología , Encéfalo/patología , Hipoxia-Isquemia Encefálica/patología , Fibras Nerviosas Amielínicas , Animales , Animales Recién Nacidos , Astrocitos/fisiología , Encéfalo/fisiopatología , Tamaño de la Célula , Ácido D-Aspártico/metabolismo , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Ácido Glutámico/metabolismo , Hipoxia-Isquemia Encefálica/fisiopatología , Inmunohistoquímica , Isoquinolinas , Masculino , Porcinos , Factores de Tiempo
15.
Mol Neurobiol ; 57(10): 4322-4344, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32720074

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/genética , Regulación de la Expresión Génica , Hipoxia-Isquemia Encefálica/genética , MicroARNs/genética , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Encéfalo/patología , Lesiones Encefálicas/sangre , Creatinina/metabolismo , Modelos Animales de Enfermedad , Sangre Fetal/metabolismo , Perfilación de la Expresión Génica , Humanos , Hipoxia-Isquemia Encefálica/sangre , Recién Nacido , Ácido Láctico/metabolismo , Modelos Lineales , Espectroscopía de Resonancia Magnética , MicroARNs/metabolismo , Neuritas/metabolismo , Especificidad de Órganos , Transducción de Señal/genética , Porcinos , Factores de Tiempo
16.
Brain Struct Funct ; 223(2): 1025-1033, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29282556

RESUMEN

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.


Asunto(s)
Encéfalo/citología , Neuronas/metabolismo , Receptores de GABA-A/metabolismo , Factores de Edad , Animales , Encéfalo/anatomía & histología , Fraccionamiento Celular , Humanos , Inmunoprecipitación , Modelos Moleculares , Porcinos
17.
Placenta ; 54: 111-116, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27939102

RESUMEN

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.


Asunto(s)
Barrera Hematoencefálica/crecimiento & desarrollo , Desarrollo Fetal , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Femenino , Humanos , Placenta/metabolismo , Embarazo , Proteínas Transportadoras de Solutos/metabolismo
18.
Int J Dev Neurosci ; 59: 1-9, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28219764

RESUMEN

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.


Asunto(s)
Encéfalo/patología , Retardo del Crecimiento Fetal/patología , Retardo del Crecimiento Fetal/fisiopatología , Receptores de GABA-A/metabolismo , Sustancia Blanca/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Peso Corporal/fisiología , Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Básica de Mielina/metabolismo , Tamaño de los Órganos/fisiología , Porcinos
19.
Behav Neurosci ; 119(4): 1072-83, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16187835

RESUMEN

The preoptic area (POA) is critical for maternal behavior in rats but little is known about what neurotransmitters released here influence maternal responding. POA infusion of 10 microg (but not 2 microg) of the dopamine D1 receptor antagonist SCH-23390 greatly impaired retrieval and licking of pups but not other maternal or nonmaternal behaviors in lactating rats. In contrast, POA infusion of 10 microg (but not 2 microg) of the D2 receptor antagonist raclopride facilitated nursing but did not affect oral maternal behaviors. SCH-23390 in the medial hypothalamus tended to impair licking but not retrieval. Raclopride in the medial hypothalamus had no effects. Therefore, D1 and D2 receptor activity, particularly in the POA, is important for regulating different maternal behaviors.


Asunto(s)
Benzazepinas/farmacología , Antagonistas de Dopamina/farmacología , Lactancia/efectos de los fármacos , Conducta Materna/efectos de los fármacos , Área Preóptica/efectos de los fármacos , Racloprida/farmacología , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Embarazo , Ratas , Ratas Long-Evans , Tiempo de Reacción/efectos de los fármacos , Factores de Tiempo
20.
J AOAC Int ; 85(5): 1037-44, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12374401

RESUMEN

AOAC Official Method 996.10, Assurance Enzyme Immunoassay (EIA) for Escherichia coli O157:H7 (EHEC), was modified to incorporate a new enrichment protocol using BioControl EHEC8 medium for testing raw and cooked beef. Foods were tested by EIA and the U.S. Department of Agriculture/Food Safety and Inspection Service (USDA/FSIS) enrichment conditions and the FDA Bacteriological Analytical Manual (BAM) isolation and confirmation techniques. A total of 14 collaborators participated. Raw and cooked ground beef were inoculated with E. coli O157:H7 at 2 different levels: a high level where predominantly positive results were expected, and a low level where fractional recovery was anticipated. Collaborators tested 378 test portions and controls by both the 8 h EIA and the USDA/FSIS enrichment methods, for a total of 756 test portions. Of the 378 paired test portions, 75 were positive and 212 were negative by both methods. Thirteen test portions were presumptively positive by EIA and could not be confirmed culturally; 30 were negative by EIA, but confirmed positive by culture; and 65 were negative by the culture method, but confirmed positive by the EIA method. There was no statistical difference between results obtained with the Assurance EIA for EHEC 8 h method and the culture method for raw ground beef. The Assurance EIA had a significantly higher recovery for cooked beef.


Asunto(s)
Escherichia coli O157/química , Contaminación de Alimentos/análisis , Carne/microbiología , Animales , Bovinos , Medios de Cultivo , Técnicas para Inmunoenzimas , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA