Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 6645, 2024 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-38503820

RESUMO

Neonatal Encephalopathy (NE) is a major cause of lifelong disability and neurological complications in affected infants. Identifying novel diagnostic biomarkers in this population may assist in predicting MRI injury and differentiate neonates with NE from those with low-cord pH or healthy neonates and may help clinicians make real-time decisions. To compare the microRNA (miRNA) profiles between neonates with NE, healthy controls, and neonates with low cord pH. Moreover, miRNA concentrations were compared to brain injury severity in neonates with NE. This is a retrospective analysis of miRNA profiles from select samples in the biorepository and data registry at the University of Florida Health Gainesville. The Firefly miRNA assay was used to screen a total of 65 neurological miRNA targets in neonates with NE (n = 36), low cord pH (n = 18) and healthy controls (n = 37). Multivariate statistical techniques, including principal component analysis and orthogonal partial least squares discriminant analysis, and miRNA Enrichment Analysis and Annotation were used to identify miRNA markers and their pathobiological relevance. A set of 10 highly influential miRNAs were identified, which were significantly upregulated in the NE group compared to healthy controls. Of these, miR-323a-3p and mir-30e-5p displayed the highest fold change in expression levels. Moreover, miR-34c-5p, miR-491-5p, and miR-346 were significantly higher in the NE group compared to the low cord pH group. Furthermore, several miRNAs were identified that can differentiate between no/mild and moderate/severe injury in the NE group as measured by MRI. MiRNAs represent promising diagnostic and prognostic tools for improving the management of NE.


Assuntos
Lesões Encefálicas , Doenças do Recém-Nascido , MicroRNAs , Recém-Nascido , Lactente , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Estudos Retrospectivos , Biomarcadores , Estudos de Coortes , Lesões Encefálicas/diagnóstico , Lesões Encefálicas/genética , Perfilação da Expressão Gênica/métodos
2.
Pediatr Res ; 93(5): 1199-1207, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-35273370

RESUMO

BACKGROUND: Neuroprognostication in neonates with neonatal encephalopathy (NE) may be enhanced by early serial measurement of a panel of four brain-specific biomarkers. METHODS: To evaluate serum biomarkers, 40 NE samples and 37 healthy neonates from a biorepository were analyzed. Blood samples were collected at 0-6, 12, 24, 48, and 96 h of life. MRI provided a short-term measure of injury. Long-term outcomes included death or a Bayley III score at 17-24 months of age. RESULTS: Glial fibrillary acidic protein (GFAP), ubiquitin c-terminal hydrolase-L1 (UCH-L1), and Tau peaked at 0-6 h of life, while neurofilament light chain (NFL) peaked at 96 h of life. These four marker concentrations at 96 h of life differentiated moderate/severe from none/mild brain injury by MRI, while GFAP and Tau showed early discrimination. For long-term outcomes, GFAP, NFL, Tau, and UCH-L1 could differentiate a poor outcome vs good outcome as early as 0-6 h of life, depending on the Bayley domain, and a combination of the four markers enhanced the sensitivity and specificity. Machine learning trajectory analyses identified upward trajectory patients with a high concordance to poor outcomes. CONCLUSION: GFAP, NFL, Tau, and UCH-L1 may be of neuroprognostic significance after NE. IMPACT: Serial measurements of GFAP, NFL, Tau, and UCH-L1 show promise in aiding the bedside clinician in making treatment decisions in neonatal encephalopathy. The panel of four neuroproteins increased the ability to predict neurodevelopmental outcomes. The study utilized a trajectory analysis that enabled predictive modeling. A panel approach provides the bedside clinician with objective data to individualize care. This study provides the foundation to develop a point of care device in the future.


Assuntos
Lesões Encefálicas , Filamentos Intermediários , Recém-Nascido , Humanos , Proteína Glial Fibrilar Ácida , Ubiquitina Tiolesterase , Biomarcadores
3.
Front Neurol ; 13: 934755, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35873777

RESUMO

Objective: To determine the concentrations of four neuroprotein biomarkers and 68 miRNAs in neonates with low cord pH and/or mild hypoxic-ischemic encephalopathy (HIE). Study Design: A prospective cohort study enrolled neonates with low cord pH (n = 18), moderate-severe HIE (n = 40), and healthy controls (n = 38). Groups provided serum samples at 0-6 h of life. The concentrations of biomarkers and miRNAs were compared between cohorts. Result: The low cord pH and moderate-severe HIE groups had increased concentrations of GFAP, NFL and Tau compared to controls (P < 0.05, P < 0.001, respectively). NFL concentrations in mild HIE was higher than controls (P < 0.05) but less than moderate-severe HIE (P < 0.001). Of 68 miRNAs, 36 in low cord pH group and 40 in moderate-severe HIE were upregulated compared to controls (P < 0.05). Five miRNAs in low cord pH group (P < 0.05) and 3 in moderate-severe HIE were downregulated compared to controls (P < 0.05). Conclusion: A biomarker panel in neonates with low cord pH may help clinicians make real-time decisions.

4.
Arch Dis Child Fetal Neonatal Ed ; 107(1): 60-64, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34021027

RESUMO

OBJECTIVE: Central nervous system (CNS) derived exosomes can be purified from peripheral blood and have been used widely in adult neurological disease. Application to neonatal neurological disease deserves investigation in the setting of hypoxic-ischaemic encephalopathy (HIE). DESIGN: Observational cohort. SETTING: Level III neonatal intensive care unit. PARTICIPANTS: Term/near-term neonates undergoing therapeutic hypothermia (TH) for HIE. INTERVENTIONS: Blood samples were collected at 0-6, 12, 24, 48 and 96 hours of life. MAIN OUTCOMES AND MEASURES: CNS exosomes were purified from serum using previously described methods. Biomarker protein levels were quantified using standard ELISA methods and normalised to exosome marker CD-81. The slope of change for biomarker levels was calculated for each time interval. Our primary outcome was MRI basal ganglia/watershed score of ≥3. RESULTS: 26 subjects were included (umbilical artery pH range 6.6-7.29; 35% seizures). An increasing MRI injury score was significantly associated with decreasing levels of synaptopodin between 0-6 and 12 hours (p=0.03) and increasing levels of lipocalin-2 (NGAL) between 12 and 48 hours (p<0.0001). Neuronal pentraxin was not significant. The negative predictive values for increasing synaptopodin and decreasing NGAL was 70.0% and 90.9%, respectively. CONCLUSIONS AND RELEVANCE: Our results indicate that CNS exosome cargo has the potential to act as biomarkers of the severity of brain injury and response to TH as well as quantify pharmacological response to neuroactive therapeutic/adjuvant agents. Rigorous prospective trials are critical to evaluate potential clinical use of exosome biomarkers.


Assuntos
Exossomos/metabolismo , Hipotermia Induzida , Hipóxia-Isquemia Encefálica/sangue , Hipóxia-Isquemia Encefálica/terapia , Lipocalina-2/sangue , Proteínas dos Microfilamentos/sangue , Biomarcadores , Proteína C-Reativa , Sistema Nervoso Central/citologia , Imagem de Difusão por Ressonância Magnética , Feminino , Humanos , Hipóxia-Isquemia Encefálica/diagnóstico por imagem , Recém-Nascido , Unidades de Terapia Intensiva Neonatal , Masculino , Proteínas do Tecido Nervoso/sangue , Projetos Piloto , Estudos Retrospectivos
5.
J Pineal Res ; 66(4): e12565, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30734962

RESUMO

INTRODUCTION: Neonates with hypoxic-ischemic encephalopathy (HIE) undergoing hypothermia may benefit from adjunctive therapy with melatonin. However, melatonin safety, pharmacokinetics (PK), and dosage in this sensitive population are still unknown. METHODS AND RESULTS: This study assessed the PK and safety of melatonin enteral administration to neonates with HIE undergoing hypothermia. Melatonin was infused at 0.5 mg/kg in five neonates with HIE undergoing hypothermia. Infusion started 1 hour after the neonates reached the target temperature of 33.5°C. Blood samples were collected before and at selective times after melatonin infusion. Abdominal complications or clinically significant changes in patients' vital signs were not found during or after melatonin. The peak plasma concentration reached 0.25 µg/mL. The area under the curve in 24 hours was 4.35 µg/mL*h. DISCUSSION: Melatonin half-life and clearance were prolonged, and the distribution volume decreased compared to adults. In silico simulation estimated that the steady state can be reached after four infusions. Hypothermia does not affect melatonin PK. In humans high blood concentrations with lower doses can be achieved compared to animal experimentation, although intravenous administration is advised in the neonate population. Our study is a preparatory step for future clinical studies aimed at assessing melatonin efficacy in HIE.


Assuntos
Hipotermia Induzida , Melatonina/farmacocinética , Feminino , Humanos , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/terapia , Recém-Nascido , Masculino , Melatonina/uso terapêutico
6.
Cytokine ; 111: 119-124, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30142532

RESUMO

PURPOSE: Inflammation is a crucial but understudied mechanism of neuronal injury after hypoxia-ischemia. The aim was to identify a panel of cytokines involved in brain injury in neonates with hypoxic ischemic encephalopathy (HIE). METHODS: Ten newborns with HIE undergoing to therapeutic hypothermia (TH, HIE Group) and 8 healthy newborns (CTRL Group) were enrolled. For the HIE group, 5 samples were collected: between 0 and 6 h of life (time 1), 12 h (time 2), 24 h (time 3), 48 h (time 4) and 96 h of life (time 5). For the CTRL group, one sample was collected. A panel of 48 inflammatory cytokines was determined in all samples. Data were analyzed using multivariate statistical analysis (Principal component analysis, PCA) RESULTS: 17 cytokines, among 48 analyzed, were found to be significantly different, initially, between the CTRL and HIE groups: 12 with reported pro-inflammatory effects and 5 with reported anti-inflammatory effects. In the HIE group cytokines showed a decreasing trend during the TH and at the end of treatment comparable to the CTRL group. IL-18 did demonstrate a slight increase at time 3 during HT but decreased steadily at sampling times, 4 and 5. CONCLUSIONS: Our data demonstrates that many pathways of the inflammatory cascade are activated following hypoxic-ischemic injury. This information will increase our understanding of changes in cytokines over time in neonates with HIE undergoing TH.


Assuntos
Citocinas , Hipotermia Induzida , Hipóxia-Isquemia Encefálica , Citocinas/biossíntese , Citocinas/imunologia , Feminino , Humanos , Hipóxia-Isquemia Encefálica/sangue , Hipóxia-Isquemia Encefálica/imunologia , Hipóxia-Isquemia Encefálica/terapia , Recém-Nascido , Masculino , Estudos Retrospectivos
7.
Front Pediatr ; 6: 120, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29765933

RESUMO

Objective: The objective of this pilot study was to start evaluating the efficacy and the safety (i.e., carboxyhemoglobin concentration of carbon monoxide (CO)) as a putative neuroprotective therapy in neonates. Study Design: Neonatal C57BL/6 mice were exposed to CO at a concentration of either 200 or 250 ppm for a period of 1 h. The pups were then sacrificed at 0, 10, 20, 60, 120, 180, and 240 min after exposure to either concentration of CO, and blood was collected for analysis of carboxyhemoglobin. Following the safety study, 7-day-old pups underwent a unilateral carotid ligation. After recovery, the pups were exposed to a humidified gas mixture of 8% oxygen and 92% nitrogen for 20 min in a hypoxia chamber. One hour after the hypoxia exposure, the pups were randomized to one of two groups: air (HI+A) or carbon monoxide (HI+CO). An inhaled dose of 250 ppm of CO was administered to the pups for 1 h per day for a period of 3 days. At 7 days post-injury, the pups were sacrificed and the brains analyzed for cortical and hippocampal volumes. Results: CO exposure at 200 and 250 ppm produced a peak carboxyhemoglobin concentration of 21.52 ± 1.18% and 27.55 ± 3.58%, respectively. The carboxyhemoglobin concentrations decreased rapidly, reaching control concentrations by 60 min post exposure. At 14 days of age (7 days post injury), the HI+CO (treated with 1 h per day of 250 ppm of CO for 3 days post injury) had significant preservation of the ratio of ipsilateral to contralateral cortex (median 1.07, 25% 0.97, 75% 1.23, n = 10) compared the HI+A group (p < 0.05). Conclusion: CO exposure of 250 ppm did not reach carboxyhemoglobin concentrations which would induce acute neurologic abnormalities and was effective in preserving cortical volumes following hypoxic-ischemic injury.

8.
Neonatology ; 113(4): 347-352, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29510382

RESUMO

BACKGROUND: Withholding enteral feedings during hypothermia lacks supporting evidence. OBJECTIVES: We aimed to determine if minimal enteral nutrition (MEN) during hypothermia in patients with hypoxic-ischemic encephalopathy was associated with a reduced duration of parenteral nutrition, time to full oral feeds, and length of stay, but would not be associated with increased systemic inflammation or feeding complications. METHODS: We performed a pilot, retrospective, matched case-control study within the Florida Neonatal Neurologic Network from December 2012 to May 2016 of patients who received MEN during hypothermia (n = 17) versus those who were not fed (n = 17). Length of stay, feeding-related outcomes, and brain injury identified by MRI were compared. Serum inflammatory mediators were measured at 0-6, 24, and 96 h of life by multiplex assay. MRI were scored using the Barkovich system. RESULTS: MEN subjects had a reduced length of hospital stay (mean 15 ± 11 vs. 24 ± 19 days, p < 0.05), days receiving parenteral nutrition (7 ± 2 vs. 11 ± 6, p < 0.05), and time to full oral feeds (8 ± 5 vs. 18 ± 18, p < 0.05). MEN was associated with a significantly reduced serum IL-12p70 at 24 and 96 h (p < 0.05). Brain MRI scores were not significantly different between groups. CONCLUSION: MEN during hypothermia was associated with a reduced length of stay and time to full feeds, but did not increase feeding complications or systemic inflammation.


Assuntos
Nutrição Enteral , Hipotermia Induzida , Hipóxia-Isquemia Encefálica/terapia , Interleucina-12/sangue , Feminino , Florida , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Tempo de Internação , Imageamento por Ressonância Magnética , Masculino , Nutrição Parenteral , Projetos Piloto , Estudos Retrospectivos
9.
PLoS One ; 7(12): e51706, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251608

RESUMO

Hypoxic ischemic encephalopathy (HIE) affects 2-3 per 1000 full-term neonates. Up to 75% of newborns with severe HIE die or have severe neurological handicaps. Stem cell therapy offers the potential to replace HIE-damaged cells and enhances the autoregeneration process. Our laboratory implanted Multipotent Astrocytic Stem Cells (MASCs) into a neonatal rat model of hypoxia-ischemia (HI) and demonstrated that MASCs move to areas of injury in the cortex and hippocampus. However, only a small proportion of the implanted MASCs differentiated into neurons. MASCs injected into control pups did not move into the cortex or differentiate into neurons. We do not know the mechanism by which the MASCs moved from the site of injection to the injured cortex. We found neurotrophins present after the hypoxic-ischemic milieu and hypothesized that neurotrophins could enhance the migration and differentiation of MASCs. Using a Boyden chamber device, we demonstrated that neurotrophins potentiate the in vitro migration of stem cells. NGF, GDNF, BDNF and NT-3 increased stem cell migration when compared to a chemokinesis control. Also, MASCs had increased differentiation toward neuronal phenotypes when these neurotrophins were added to MASC culture tissue. Due to this finding, we believed neurotrophins could guide migration and differentiation of stem cell transplants after brain injury.


Assuntos
Astrócitos/citologia , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células-Tronco Multipotentes/citologia , Fatores de Crescimento Neural/farmacologia , Neurônios/citologia , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Bovinos , Ensaios de Migração Celular , Humanos , Camundongos , Células-Tronco Multipotentes/efeitos dos fármacos , Células-Tronco Multipotentes/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Tubulina (Proteína)/metabolismo
10.
Acta Neurobiol Exp (Wars) ; 72(3): 253-63, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23093012

RESUMO

Hypoxic-Ischemic Encephalopathy (HIE) is one of the most recognized causes of neurological deficits in children. Cerebral blood flow (CBF) reductions, as seen with HIE, resulting in neuronal injury have not been evaluated in real-time. Photoacoustic Tomography (PAT) is a form of optical imaging which can detect cerebral hemodynamic alterations in a noninvasive, non-ionizing fashion via changes in hemoglobin optical absorption. Further, this technology has the potential to capture cerebral blood volume (CBV) fluctuations and perhaps CBF changes in real-time. We hypothesized that PAT can detect a reduction in cerebral hemoglobin optical absorption, and therefore CBF, in a neonatal model of hypoxia-ischemia. To investigate, P7 rats underwent right carotid artery ligation and exposure to 8 percent oxygen for 60 minutes while imaged with PAT every 20 minutes. Cerebral hemodynamic alterations, as measured by mean optical absorption (MOA), were calculated as a change from baseline. Global and regional MOA was analyzed using a linear mixed model. Global MOA was reduced within the right hemisphere as compared to the left during hypoxia. Regional differences in MOA were detected between the left and right sides for the middle and posterior cortical regions. Injury was confirmed using immunohistochemistry. We conclude that a reduction in global and regional MOA, and hence CBF, could be identified by PAT in a neonatal rat model of HIE. This is the first study described in the literature utilizing a neonatal rat model of HIE to demonstrate in vivo alterations in cerebral hemodynamics in a non-invasive and near real-time fashion.


Assuntos
Circulação Cerebrovascular/fisiologia , Hemodinâmica/fisiologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Técnicas Fotoacústicas , Algoritmos , Animais , Animais Recém-Nascidos , Volume Sanguíneo , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos da radiação , Proteína Glial Fibrilar Ácida , Lasers de Estado Sólido , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Tomografia Computadorizada por Raios X , Tubulina (Proteína)/metabolismo
11.
J Biomater Sci Polym Ed ; 22(12): 1621-37, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20699061

RESUMO

In severe hypoxic-ischemic brain injury, cellular components such as neurons and astrocytes are injured or destroyed along with the supporting extracellular matrix. This presents a challenge to the field of regenerative medicine since the lack of extracellular matrix and supporting structures makes the transplant milieu inhospitable to the transplanted cells. A potential solution to this problem is the use of a biomaterial to provide the extracellular components needed to keep cells localized in cystic brain regions, allowing the cells to form connections and repair lost brain tissue. Ideally, this biomaterial would be combined with stem cells, which have been proven to have therapeutic potentials, and could be delivered via an injection. To study this approach, we derived a hydrogel biomaterial tissue scaffold from oligomeric gelatin and copper-capillary alginate gel (GCCAG). We then demonstrated that our multipotent astrocytic stem cells (MASCs) could be maintained in GCCAG scaffolds for up to 2 weeks in vitro and that the cells retained their multipotency. We next performed a pilot transplant study in which GCCAG was mixed with MASCs and injected into the brain of a neonatal rat pup. After a week in vivo, our results showed that: the GCCAG biomaterial did not cause a significant reactive gliosis; viable cells were retained within the injected scaffolds; and some delivered cells migrated into the surrounding brain tissue. Therefore, GCCAG tissue scaffolds are a promising, novel injectable system for transplantation of stem cells to the brain.


Assuntos
Alginatos/química , Cobre/química , Gelatina/química , Transplante de Células-Tronco/métodos , Alicerces Teciduais/química , Animais , Astrócitos/citologia , Sobrevivência Celular/efeitos dos fármacos , Géis , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Humanos , Injeções , Camundongos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/transplante , Multimerização Proteica , Estrutura Quaternária de Proteína , Ratos , Fatores de Tempo
12.
Pediatr Res ; 68(6): 531-6, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20736881

RESUMO

Severe hypoxic-ischemic encephalopathy (HIE) is a devastating condition that can lead to mortality and long-term disabilities in term newborns. No rapid and reliable laboratory test exists to assess the degree of neuronal injury in these patients. We propose two possible biomarkers: 1) phosphorylated axonal neurofilament heavy chain (pNF-H) protein, one of the major subunits of neurofilaments, found only in axonal cytoskeleton of neurons and 2) Ubiquitin C-terminal hydrolase 1 (UCHL1 protein) that is heavily and specifically concentrated in neuronal perikarya and dendrites. High-serum pNF-H and UCHL1 levels are reported in subarachnoid hemorrhage and traumatic brain injury, suggesting that they are released into blood following neuronal injury. We hypothesized that serum pNF-H and UCHL1 were higher in neonates with moderate-to-severe HIE than in healthy neonates. A time-limited enrollment of 14 consecutive patients with HIE and 14 healthy controls was performed. UCHL1 and pNF-H were correlated with clinical data and brain MRI. UCHL1 and pNF-H serum levels were higher in HIE versus controls. UCHL1 showed correlation with the 10-min Apgar score, and pNF-H showed correlation with abnormal brain MRI. Our findings suggest that serum UCHL1 and pNF-H could be explored as diagnostic and prognostic tools in neonatal HIE.


Assuntos
Biomarcadores/sangue , Hipóxia-Isquemia Encefálica/sangue , Recém-Nascido/sangue , Animais , Feminino , Humanos , Masculino , Proteínas de Neurofilamentos/metabolismo , Projetos Piloto , Ubiquitina Tiolesterase/metabolismo
13.
Dev Neurosci ; 29(3): 268-74, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17124376

RESUMO

The neutral amino acid transporters SNAT1-3 and ASCT1 play critical roles in the recycling of glutamine, and subsequently glutamate, via the glutamine-glutamate cycle. Hypoxia-ischemia was induced in rat pups using the Rice-Vannucci model. Brains were harvested at 1 h, 24 h and 7 days after ischemia. The expression of NAATs was evaluated using immunoblotting, real-time PCR, and immunohistochemistry. Results were compared with age-matched controls and shams. SNAT1 mRNA decreased at 1 h after injury in both hemispheres when compared with the control animals and correlated with a decrease in protein expression at 24 h in the hippocampus and cortex. SNAT1 protein expression increased globally at 7 days after injury and specifically in the hippocampus. Finally, SNAT2 and 3 demonstrated subtle changes in various brain regions after injury. These data suggest that neutral amino acid transporters remain largely intact after hypoxia-ischemia.


Assuntos
Sistema A de Transporte de Aminoácidos/metabolismo , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/fisiopatologia , Sistema A de Transporte de Aminoácidos/genética , Sistema ASC de Transporte de Aminoácidos/genética , Sistema ASC de Transporte de Aminoácidos/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Aminoácidos Neutros/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Immunoblotting , Imuno-Histoquímica , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Brain Res ; 1112(1): 99-105, 2006 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-16919606

RESUMO

Hypoxic-ischemic encephalopathy (HIE) in neonates results in long-term disabilities. Stem cell therapy may offer an attractive treatment for HIE. Multipotent astrocytic stem cells (MASCs) from mice transplanted into a rat model of hypoxia-ischemia (HI) survived the transplantation and showed signs of migration towards the injured cortex. Some MASCs around the injured cortex differentiated into neuronal and astrocytic phenotypes. MASCs transplanted into non-ischemic pups survived but retained their astrocytic phenotype. These data suggest that transplanted MASCs can survive and differentiate into neurons and astrocytes in the post-injury milieu of the neonatal brain injured by HI.


Assuntos
Astrócitos/fisiologia , Hipóxia-Isquemia Encefálica/cirurgia , Transplante de Células-Tronco/métodos , Células-Tronco/fisiologia , Animais , Animais Recém-Nascidos , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Modelos Animais de Doenças , Imunofluorescência/métodos , Proteína Glial Fibrilar Ácida/metabolismo , Ratos , Tubulina (Proteína)/metabolismo
15.
J Mol Histol ; 36(4): 301-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-16200463

RESUMO

System A is a highly regulated, Na+-dependent transporter that accepts neutral amino acids containing short, polar side chains. System A plays an important role during rat development as decreased pup weights are observed in dams infused during gestation with a non-metabolizable System A substrate. Given the potential importance of SNAT1 during development in the rat brain, we examined whether SNAT1 would be present at an earlier gestation during organogenesis in multiple organs by immunohistochemistry and immunoblotting. SNAT1 protein was observed in the developing lungs, intestines, kidneys, heart, pancreas, and skeletal muscle of rats at prenatal days 14, 17, 19, 21, and postnatal day 2 rats. SNAT1 protein expression decreased in the liver and intestine shortly after birth and as the rat matured. SNAT1 expression was constant throughout development in the lungs and kidney and increased in the heart from prenatal day 19 to postnatal day 2. Highest levels of expression in older animals were seen in organs undergoing rapid cell division.


Assuntos
Sistema A de Transporte de Aminoácidos/metabolismo , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Sistema A de Transporte de Aminoácidos/imunologia , Animais , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Feminino , Immunoblotting , Imuno-Histoquímica , Ratos , Ratos Sprague-Dawley
16.
Brain Res ; 1056(2): 105-12, 2005 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-16122709

RESUMO

Efflux of glutamate from intracellular pools during hypoxia-ischemia has been postulated to be mediated by amino acid transporters and can lead to excitotoxicity. In addition, a decrease in pH seen during global hypoxia-ischemia may influence which transporter is responsible for this glutamate efflux. For example, the neutral amino acid transporter ASCT1 is an effective transporter of glutamate at low pH. We have examined the effects of pH, pH and temperature, and hypoxia on glutamate efflux in a rat primary neuronal cell culture model. We observed a marked increase of glutamate efflux as pH was decreased from 7.4 to 5.5. This pH-dependent efflux is likely due to a transporter-mediated process because it was seen in the presence of tetrodotoxin and was blunted by decreasing the temperature to either 35 degrees C or 33 degrees C. In addition, no increase in LDH was seen at pH 5.5 suggesting that increased glutamate levels were not due to cellular death. No change in glutamate levels was seen when the oxygen tension of the medium was lowered from 150 mm Hg to either 30 or 15 mm Hg. Given that EAAT transporters are inhibited by low pH, other transporters, such as ASCT1, may be responsible for this pH-dependent efflux of glutamate.


Assuntos
Sistema ASC de Transporte de Aminoácidos/fisiologia , Ácido Glutâmico/metabolismo , Concentração de Íons de Hidrogênio , Hipóxia/fisiopatologia , Neurônios/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Células Cultivadas , Córtex Cerebral/citologia , L-Lactato Desidrogenase/metabolismo , Ratos , Ratos Sprague-Dawley , Temperatura , Fatores de Tempo
17.
Brain Res Dev Brain Res ; 143(2): 151-9, 2003 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-12855186

RESUMO

The glutamine-glutamate/GABA cycle is critical for the developing brain as glutamatergic neurotransmission is important for neuronal survival and drives synaptogenesis and activity-dependent synaptic plasticity. GABAergic transmission may be essential for the formation of neural circuits. Recently a cDNA encoding a brain-enriched System A transporter (SAT1/ATA1), has been identified which may provide glutamine to neurons for the biosynthesis of neurotransmitters glutamate and gamma-aminobutyric acid (GABA). In this study, we have examined the developmental expression pattern of SAT1/ATA1 protein in rat brain by immunohistochemistry. We find that SAT1/ATA1 was present in the developing rat brain at all gestational ages examined including prenatal days 17 and 19 and postnatal days 2, 10, 14, and adult. SAT1/ATA1 immunoreactivity was seen in the neocortex, hippocampus, and neuroepithelium at the earliest time point examined, prenatal day 17. SAT1/ATA1 was prominent in the striatum, the hippocampus and the cortex in the postnatal animals. In adults, SAT1/ATA1 was limited to the cell body region while in developing animals SAT1/ATA1 protein was found in neuronal processes. These results contribute to our understanding of the relationship between the cycling of glutamate and glutamine between astrocytes and glia and the pathophysiological conditions that occur in hypoxic ischemic encephalopathy.


Assuntos
Sistema A de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Neutros , Encéfalo/embriologia , Encéfalo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Sistema A de Transporte de Aminoácidos/biossíntese , Animais , Animais Recém-Nascidos , Embrião de Mamíferos , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Immunoblotting , Imuno-Histoquímica , Proteínas de Membrana Transportadoras/biossíntese , Proteínas de Membrana Transportadoras/genética , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...