Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Am Heart Assoc ; 8(15): e012711, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31331224

RESUMO

Background Reduced oxygen delivery in congenital heart disease causes delayed brain maturation and white matter abnormalities in utero. No treatment currently exists. Tetrahydrobiopterin (BH4) is a cofactor for neuronal nitric oxide synthase. BH4 availability is reduced upon NOS activation, such as during hypoxic conditions, and leads to toxin production. We hypothesize that BH4 levels are depleted in the hypoxic brain and that BH4 replacement therapy mitigates the toxic effects of hypoxia on white matter. Methods and Results Transgenic mice were used to visualize oligodendrocytes. Hypoxia was introduced during a period of white matter development equivalent to the human third trimester. BH4 was administered during hypoxia. BH4 levels were depleted in the hypoxic brain by direct quantification (n=7-12). The proliferation (n=3-6), apoptosis (n=3-6), and developmental stage (n=5-8) of oligodendrocytes were determined immunohistologically. Total oligodendrocytes increased after hypoxia, consistent with hypoxia-induced proliferation seen previously; however, mature oligodendrocytes were less prevalent in hypoxia, and there was accumulation of immature oligodendrocytes. BH4 treatment improved the mature oligodendrocyte number such that it did not differ from normoxia, and accumulation of immature oligodendrocytes was not observed. These results persisted beyond the initial period of hypoxia (n=3-4). Apoptosis increased with hypoxia but decreased with BH4 treatment to normoxic levels. White matter myelin levels decreased following hypoxia by western blot. BH4 treatment normalized myelination (n=6-10). Hypoxia worsened sensory-motor coordination on balance beam tasks, and BH4 therapy normalized performance (n=5-9). Conclusions Suboptimal BH4 levels influence hypoxic white matter abnormalities. Repurposing BH4 for use during fetal brain development may limit white matter dysmaturation in congenital heart disease.


Assuntos
Biopterinas/análogos & derivados , Doenças Fetais/fisiopatologia , Cardiopatias/congênito , Cardiopatias/fisiopatologia , Hipóxia/fisiopatologia , Substância Branca/efeitos dos fármacos , Substância Branca/crescimento & desenvolvimento , Animais , Biopterinas/farmacologia , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Transgênicos
2.
J Am Heart Assoc ; 6(8)2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28862938

RESUMO

BACKGROUND: Newly developed white matter (WM) injury is common after cardiopulmonary bypass (CPB) in severe/complex congenital heart disease. Fractional anisotropy (FA) allows measurement of macroscopic organization of WM pathology but has rarely been applied after CPB. The aims of our animal study were to define CPB-induced FA alterations and to determine correlations between these changes and cellular events after congenital heart disease surgery. METHODS AND RESULTS: Normal porcine WM development was first assessed between 3 and 7 weeks of age: 3-week-old piglets were randomly assigned to 1 of 3 CPB-induced insults. FA was analyzed in 31 WM structures. WM oligodendrocytes, astrocytes, and microglia were assessed immunohistologically. Normal porcine WM development resembles human WM development in early infancy. We found region-specific WM vulnerability to insults associated with CPB. FA changes after CPB were also insult dependent. Within various WM areas, WM within the frontal cortex was susceptible, suggesting that FA in the frontal cortex should be a biomarker for WM injury after CPB. FA increases occur parallel to cellular processes of WM maturation during normal development; however, they are altered following surgery. CPB-induced oligodendrocyte dysmaturation, astrogliosis, and microglial expansion affect these changes. FA enabled capturing CPB-induced cellular events 4 weeks postoperatively. Regions most resilient to CPB-induced FA reduction were those that maintained mature oligodendrocytes. CONCLUSIONS: Reducing alterations of oligodendrocyte development in the frontal cortex can be both a metric and a goal to improve neurodevelopmental impairment in the congenital heart disease population. Studies using this model can provide important data needed to better interpret human imaging studies.


Assuntos
Ponte Cardiopulmonar/efeitos adversos , Diferenciação Celular , Lobo Frontal/patologia , Leucoencefalopatias/etiologia , Oligodendroglia/patologia , Substância Branca/patologia , Fatores Etários , Animais , Anisotropia , Astrócitos/patologia , Biomarcadores/metabolismo , Imagem de Difusão por Ressonância Magnética , Imagem de Tensor de Difusão , Lobo Frontal/diagnóstico por imagem , Lobo Frontal/metabolismo , Imuno-Histoquímica , Leucoencefalopatias/diagnóstico por imagem , Leucoencefalopatias/metabolismo , Leucoencefalopatias/patologia , Microglia/patologia , Modelos Animais , Oligodendroglia/metabolismo , Sus scrofa , Fatores de Tempo , Substância Branca/diagnóstico por imagem , Substância Branca/metabolismo
3.
Sci Transl Med ; 9(374)2017 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-28123074

RESUMO

Long-term neurological deficits due to immature cortical development are emerging as a major challenge in congenital heart disease (CHD). However, cellular mechanisms underlying dysregulation of perinatal corticogenesis in CHD remain elusive. The subventricular zone (SVZ) represents the largest postnatal niche of neural stem/progenitor cells (NSPCs). We show that the piglet SVZ resembles its human counterpart and displays robust postnatal neurogenesis. We present evidence that SVZ NSPCs migrate to the frontal cortex and differentiate into interneurons in a region-specific manner. Hypoxic exposure of the gyrencephalic piglet brain recapitulates CHD-induced impaired cortical development. Hypoxia reduces proliferation and neurogenesis in the SVZ, which is accompanied by reduced cortical growth. We demonstrate a similar reduction in neuroblasts within the SVZ of human infants born with CHD. Our findings demonstrate that SVZ NSPCs contribute to perinatal corticogenesis and suggest that restoration of SVZ NSPCs' neurogenic potential is a candidate therapeutic target for improving cortical growth in CHD.


Assuntos
Lobo Frontal/patologia , Insuficiência Cardíaca/patologia , Neurogênese , Animais , Animais Recém-Nascidos , Movimento Celular , Lobo Frontal/crescimento & desenvolvimento , Insuficiência Cardíaca/congênito , Hipóxia/patologia , Interneurônios/citologia , Células-Tronco Neurais/citologia , Neuroglia/citologia , Neurônios/citologia , Nicho de Células-Tronco , Células-Tronco/citologia , Suínos
4.
J Thorac Cardiovasc Surg ; 151(1): 265-72.e1-3, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26412317

RESUMO

OBJECTIVES: White-matter injury after surgery is common in neonates with cerebral immaturity secondary to in utero hypoxia. Astrocytes play a central role in brain protection; however, the reaction of astrocytes to hypothermic circulatory arrest (HCA) remains unknown. We investigated the role of astrocytes in white-matter injury after HCA and determined the effects of preoperative hypoxia on this role, using a novel mouse model. METHODS: Mice were exposed to hypoxia from days 3 to 11, which is equivalent to the third trimester in humans (prehypoxia, n = 49). Brain slices were transferred to a chamber perfused by cerebrospinal fluid. Oxygen-glucose deprivation (OGD) was performed to simulate ischemia-reperfusion/reoxygenation resulting from circulatory arrest under hypothermia. Astrocyte reactions were compared with preoperative normoxia (prenormoxia; n = 45). RESULTS: We observed astrocyte activation after 25°C ischemia-reperfusion/reoxygenation in prenormoxia (P < .01). Astrocyte number after OGD correlated with caspase-3(+) cells (rho = .77, P = .001), confirming that astrogliosis is an important response after HCA. At 3 hours after OGD, astrocytes in prenormoxia had already proliferated and become activated (P < .05). Conversely, astrocytes that developed under hypoxia did not display these responses. At 20 hours after ischemia-reperfusion/reoxygenation, astrogliosis was not observed in prehypoxia, demonstrating that hypoxia altered the response of astrocytes to insult. In contrast to prenormoxia, caspase-3(+) cells in prehypoxia increased after ischemia reperfusion/reoxygenation, compared with control (P < .01). Caspase-3(+) cells were more common with prehypoxia than with prenormoxia (P < .001), suggesting that lack of astrogliosis permits increased white-matter injury. CONCLUSIONS: Preoperative hypoxia alters the neuroprotective function of astrocytes. Restoring this function before surgery may be a therapeutic option to reduce postoperative white-matter injury in the immature brain.


Assuntos
Astrócitos/patologia , Hipóxia Fetal/patologia , Hipóxia-Isquemia Encefálica/patologia , Leucoencefalopatias/patologia , Traumatismo por Reperfusão/patologia , Substância Branca/patologia , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Caspase 3/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Hipóxia Fetal/metabolismo , Proteína Glial Fibrilar Ácida/genética , Gliose , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Hipotermia Induzida , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/prevenção & controle , Técnicas In Vitro , Leucoencefalopatias/metabolismo , Leucoencefalopatias/prevenção & controle , Camundongos Transgênicos , Regiões Promotoras Genéticas , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Fatores de Tempo , Substância Branca/crescimento & desenvolvimento , Substância Branca/metabolismo
5.
Ann Thorac Surg ; 100(3): 1030-7, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26228605

RESUMO

BACKGROUND: White matter (WM) injury is common after neonatal cardiopulmonary bypass (CPB). We have demonstrated that the inflammatory response to CPB is an important mechanism of WM injury. Microglia are brain-specific immune cells that respond to inflammation and can exacerbate injury. We hypothesized that microglia activation contributes to WM injury caused by CPB. METHODS: Juvenile piglets were randomly assigned to 1 of 3 CPB-induced brain insults (1, no-CPB; 2, full-flow CPB; 3, CPB and circulatory arrest). Neurobehavioral tests were performed. Animals were sacrificed 3 days or 4 weeks postoperatively. Microglia and proliferating cells were immunohistologically identified. Seven analyzed WM regions were further categorized into 3 fiber connections (1, commissural; 2, projection; 3, association fibers). RESULTS: Microglia numbers significantly increased on day 3 after CPB and circulatory arrest, but not after full-flow CPB. Fiber categories did not affect these changes. On post-CPB week 4, proliferating cell number, blood leukocyte number, interleukin (IL)-6 levels, and neurologic scores had normalized. However, both full-flow CPB and CPB and circulatory arrest displayed significant increases in the microglia number compared with control. Thus brain-specific inflammation after CPB persists despite no changes in systemic biomarkers. Microglia number was significantly different among fiber categories, being highest in association and lowest in commissural connections. Thus there was a WM fiber-dependent microglia reaction to CPB. CONCLUSIONS: This study demonstrates prolonged microglia activation in WM after CPB. This brain-specific inflammatory response is systemically silent. It is connection fiber-dependent which may impact specific connectivity deficits observed after CPB. Controlling microglia activation after CPB is a potential therapeutic intervention to limit neurologic deficits after CPB.


Assuntos
Ponte Cardiopulmonar/efeitos adversos , Parada Circulatória Induzida por Hipotermia Profunda/efeitos adversos , Encefalite/etiologia , Substância Branca , Fatores Etários , Animais , Modelos Animais de Doenças , Feminino , Microglia/patologia , Distribuição Aleatória , Suínos , Fatores de Tempo
6.
Pediatr Res ; 75(5): 618-25, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24488087

RESUMO

BACKGROUND: White matter (WM) injury is common after cardiopulmonary bypass or deep hypothermic circulatory arrest in neonates who have cerebral immaturity secondary to in utero hypoxia. The mechanism remains unknown. We investigated effects of preoperative hypoxia on deep hypothermic circulatory arrest-induced WM injury using a combined experimental paradigm in rodents. METHODS: Mice were exposed to hypoxia (prehypoxia). Oxygen-glucose deprivation was performed under three temperatures to simulate brain conditions of deep hypothermic circulatory arrest including ischemia-reperfusion/reoxygenation under hypothermia. RESULTS: WM injury in prenormoxia was identified after 35 °C-oxygen-glucose deprivation. In prehypoxia, injury was displayed in all groups. Among oligodendrocyte stages, the preoligodendrocyte was the most susceptible, while the oligodendrocyte progenitor was resistant to insult. When effects of prehypoxia were assessed, injury of mature oligodendrocytes and oligodendrocyte progenitors in prehypoxia significantly increased as compared with prenormoxia, indicating that mature oligodendrocytes and progenitors that had developed under hypoxia had greater vulnerability. Conversely, damage of oligodendrocyte progenitors in prehypoxia were not identified after 15 °C-oxygen-glucose deprivation, suggesting that susceptible oligodendrocytes exposed to hypoxia are protected by deep hypothermia. CONCLUSION: Developmental alterations due to hypoxia result in an increased WM susceptibility to injury. Promoting WM regeneration by oligodendrocyte progenitors after earlier surgery using deep hypothermia is the most promising approach for successful WM development in congenital heart disease patients.


Assuntos
Lesões Encefálicas/fisiopatologia , Ponte Cardiopulmonar/efeitos adversos , Hipóxia , Substância Branca/patologia , Animais , Encéfalo/patologia , Linhagem da Célula , Modelos Animais de Doenças , Glucose/química , Proteínas de Fluorescência Verde/química , Hipotermia , Camundongos , Oligodendroglia/citologia , Oxigênio/química , Perfusão , Traumatismo por Reperfusão
7.
J Thorac Cardiovasc Surg ; 147(6): 1939-45, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24237885

RESUMO

OBJECTIVE: Lack of availability of aprotinin has resulted in increased clinical use of the alternative antifibrinolytic agents, ε-aminocaproic acid (EACA) and tranexamic acid (TXA), which are known to be associated with an increased risk of seizures. In contrast, aprotinin has previously been demonstrated to be neuroprotective through suppression of excitotoxicity-mediated neuronal degeneration via the extracellular plasminogen/plasmin system. This study compares the effect of antifibrinolytic agents on neuronal and mixed glial/neuronal cell cultures. METHODS: Mixed cortical cultures containing neuronal and glial cells were prepared from fetal mice and plated on a layer of confluent astrocytes from postnatal pups. A primary neuronal culture was obtained from the same gestational stage and plated in multiwall vessels. Slowly triggered excitotoxicity was induced by 24-hour exposure to 12.5 mM N-methyl-D-aspartate (NMDA). Apoptotic neuronal cell death was induced by exposure of primary neural cultures to 24 hours of serum deprivation. RESULTS: Compared with NMDA alone, no significant changes in cell death were observed for any dose of TXA or EACA in mixed cultures. Conversely, a clinical dose of aprotinin significantly reduced cell death by -31% on average. Aprotinin reduced apoptotic neuronal cell death from 75% to 37.3%, and to 34.1% at concentrations of 100 and 200 kIU/mL, respectively, and significantly decreased neuronal nuclear damage. These concentrations of aprotinin significantly inhibited caspase 9 and 3/7 activations; 250 kIU/mL aprotinin exerted maximal protection on primary cortical neurons. CONCLUSIONS: In contrast to aprotinin, EACA and TXA exert no protective effect against excitotoxic neuronal injury that can occur during cardiac surgery.


Assuntos
Ácido Aminocaproico/farmacologia , Antifibrinolíticos/farmacologia , Aprotinina/farmacologia , Córtex Cerebral/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/toxicidade , N-Metilaspartato/toxicidade , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ácido Tranexâmico/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Técnicas de Cocultura , Citoproteção , Relação Dose-Resposta a Droga , Ativação Enzimática , Células Alimentadoras , Idade Gestacional , Camundongos , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
8.
J Thorac Cardiovasc Surg ; 146(6): 1526-1533.e1, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23540655

RESUMO

OBJECTIVE: Cerebral white matter (WM) injury is common after cardiac surgery in neonates and young infants who have brain immaturity and genetic abnormalities. To understand better the mechanisms associated with WM injury, we tested the adequacy of a novel ex vivo brain slice model, with a particular focus on how the maturational stage modulates the injury. METHODS: To replicate conditions of cardiopulmonary bypass, we transferred living brain slices to a closed chamber perfused by artificial cerebrospinal fluid under controlled temperature and oxygenation. Oxygen-glucose deprivation (OGD) simulated circulatory arrest. The effects of maturation were investigated in 7- and 21-day-old mice (P7, P21) that are equivalent in maturation stage to the human fetus and young adult. RESULTS: There were no morphologic changes in axons after 60 minutes of OGD at 15°C in both P7 WM and P21 WM. Higher temperature and longer duration of OGD were associated with significantly greater WM axonal damage, suggesting that the model replicates the injury seen after hypothermic circulatory arrest. The axonal damage at P7 was significantly less than at P21, demonstrating that immature axons are more resistant than mature axons. Conversely, a significant increase in caspase3(+) oligodendrocytes in P7 mice was identified relative to P21, indicating that oligodendrocytes in immature WM are more vulnerable than oligodendrocytes in mature WM. CONCLUSIONS: Neuroprotective strategies for immature WM may need to focus on reducing oligodendrocyte injury. The brain slice model will be helpful in understanding the effects of cardiac surgery on the immature brain and the brain with genetic abnormalities.


Assuntos
Encéfalo/patologia , Ponte Cardiopulmonar/efeitos adversos , Parada Circulatória Induzida por Hipotermia Profunda/efeitos adversos , Leucoencefalopatias/patologia , Fatores Etários , Animais , Axônios/metabolismo , Axônios/patologia , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Caspase 3/metabolismo , Hipóxia Celular , Glucose/deficiência , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Técnicas In Vitro , Leucoencefalopatias/etiologia , Leucoencefalopatias/metabolismo , Leucoencefalopatias/prevenção & controle , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Perfusão , Temperatura , Fatores de Tempo
9.
Circulation ; 125(7): 859-71, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-22247493

RESUMO

BACKGROUND: Neurodevelopmental delays in motor skills and white matter (WM) injury have been documented in congenital heart disease and after pediatric cardiac surgery. The lack of a suitable animal model has hampered our understanding of the cellular mechanisms underlying WM injury in these patients. Our aim is to identify an optimal surgical strategy for WM protection to reduce neurological injury in congenital heart disease patients. METHODS AND RESULTS: We developed a porcine cardiopulmonary bypass model that displays area-dependent WM maturation. In this model, WM injury was identified after cardiopulmonary bypass-induced ischemia-reperfusion injury. The degree of injury was inversely correlated with the maturation stage, which indicates maturation-dependent vulnerability of WM. Within different oligodendrocyte developmental stages, we show selective vulnerability of O4+ preoligodendrocytes, whereas oligodendrocyte progenitor cells were resistant to insults. This indicates that immature WM is vulnerable to cardiopulmonary bypass-induced injury but has an intrinsic potential for recovery mediated by endogenous oligodendrocyte progenitor cells. Oligodendrocyte progenitor cell number decreased with age, which suggests that earlier repair allows successful WM development. Oligodendrocyte progenitor cell proliferation was observed within a few days after cardiopulmonary bypass-induced ischemia-reperfusion injury; however, by 4 weeks, arrested oligodendrocyte maturation and delayed myelination were detected. Logistic model confirmed that maintenance of higher oxygenation and reduction of inflammation were effective in minimizing the risk of injury at immature stages of WM development. CONCLUSIONS: Primary repair in neonates and young infants potentially provides successful WM development in congenital heart disease patients. Cardiac surgery during this susceptible period should avoid ischemia-reperfusion injury and minimize inflammation to prevent long-term WM-related neurological impairment.


Assuntos
Encéfalo/patologia , Ponte Cardiopulmonar/efeitos adversos , Cardiopatias Congênitas/cirurgia , Animais , Axônios/patologia , Caspase 3/análise , Proliferação de Células , Feminino , Bainha de Mielina/fisiologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Oligodendroglia/fisiologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...