Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Int J Mol Sci ; 22(12)2021 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-34205464

RESUMEN

BACKGROUND: Toll-like receptor (TLR) agonists are key immunomodulatory factors that can markedly ameliorate or exacerbate hypoxic-ischemic brain injury. We recently demonstrated that central infusion of the TLR7 agonist Gardiquimod (GDQ) following asphyxia was highly neuroprotective after 3 days but not 7 days of recovery. We hypothesize that this apparent transient neuroprotection is associated with modulation of seizure-genic processes and hemodynamic control. METHODS: Fetuses received sham asphyxia or asphyxia induced by umbilical cord occlusion (20.9 ± 0.5 min) and were monitored continuously for 7 days. GDQ 3.34 mg or vehicle were infused intracerebroventricularly from 1 to 4 h after asphyxia. RESULTS: GDQ infusion was associated with sustained moderate hypertension that resolved after 72 h recovery. Electrophysiologically, GDQ infusion was associated with reduced number and burden of postasphyxial seizures in the first 18 h of recovery (p < 0.05). Subsequently, GDQ was associated with induction of slow rhythmic epileptiform discharges (EDs) from 72 to 96 h of recovery (p < 0.05 vs asphyxia + vehicle). The total burden of EDs was associated with reduced numbers of neurons in the caudate nucleus (r2 = 0.61, p < 0.05) and CA1/2 hippocampal region (r2 = 0.66, p < 0.05). CONCLUSION: These data demonstrate that TLR7 activation by GDQ modulated blood pressure and suppressed seizures in the early phase of postasphyxial recovery, with subsequent prolonged induction of epileptiform activity. Speculatively, this may reflect delayed loss of early protection or contribute to differential neuronal survival in subcortical regions.


Asunto(s)
Aminoquinolinas/uso terapéutico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Imidazoles/uso terapéutico , Convulsiones/prevención & control , Receptor Toll-Like 7/agonistas , Aminoquinolinas/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Electroencefalografía , Femenino , Terapias Fetales/métodos , Hipoxia-Isquemia Encefálica/complicaciones , Imidazoles/farmacología , Embarazo , Nacimiento Prematuro , Convulsiones/etiología , Ovinos
2.
J Physiol ; 598(5): 999-1015, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31912503

RESUMEN

KEY POINTS: Recombinant human erythropoietin (rEpo) is neuroprotective in immature animals, but it is unclear whether the combination of high-dose rEpo therapy with therapeutic hypothermia can further improve outcomes. Hypothermia and rEpo independently improved neuronal survival, with greater improvement with hypothermia, and similarly reduced numbers of caspase-3 positive cells and reactive microglia after 7 days recovery. Hypothermia, but not rEpo, was associated with markedly improved EEG power, whereas both interventions improved recovery of EEG frequency. There was no significant improvement in any outcome after combined rEpo and hypothermia compared with hypothermia alone, and of concern, the combination was associated with increased numbers of cortical caspase-3-positive cells compared with ischaemia-hypothermia. These data suggest that the mechanisms of neuroprotection with hypothermia and rEpo overlap and, thus, high-dose rEpo infusion does not appear to be an effective adjunct therapy for therapeutic hypothermia. ABSTRACT: Therapeutic hypothermia for hypoxic-ischaemic encephalopathy (HIE) provides incomplete neuroprotection. Recombinant human erythropoietin (rEpo) is neuroprotective in immature animals, but it is unclear whether adjunct rEpo therapy with therapeutic hypothermia can further improve outcomes. Near-term fetal sheep received sham-ischaemia (n = 9) or global cerebral ischaemia for 30 min (ischaemia-vehicle, n = 8), followed by intravenous infusion of rEpo (ischaemia-Epo, n = 8; 5000 U/kg loading dose, then 833.3 U/kg/h), cerebral hypothermia (ischaemia-hypothermia, n = 8), or rEpo plus hypothermia (ischaemia-Epo-hypothermia, n = 8), from 3 to 72 h post ischaemia. Fetal brains were collected 7 days after cerebral ischaemia. Cerebral ischaemia was associated with severe neuronal loss and microglial induction in the parasagittal cortex and subcortical regions. Hypothermia reduced overall neuronal loss, cortical caspase-3 and reactive microglia in the striatum and cortex, with greater recovery of electroencephalographic (EEG) power and spectral edge (SEF) from 48 h onwards. rEpo independently improved neuronal survival in the parasagittal cortex, hippocampal CA4 and thalamus, and reduced cortical caspase-3 and activated microglia in striatal and cortical areas, with greater SEF from 120 h onwards. However, ischaemia-Epo-hypothermia did not further improve outcomes compared with ischaemia-hypothermia and was associated with increased numbers of cortical caspase-3-positive cells. These findings suggest that although delayed, prolonged treatment with both hypothermia and rEpo are independently neuroprotective, they have overlapping anti-inflammatory and anti-apoptotic mechanisms, such that the delayed, high-dose rEpo infusion for 3 days did not materially augment neuroprotection with therapeutic hypothermia.


Asunto(s)
Eritropoyetina , Hipotermia Inducida , Hipoxia-Isquemia Encefálica , Animales , Electroencefalografía , Feto , Hipoxia-Isquemia Encefálica/terapia , Ovinos
3.
J Neuroinflammation ; 17(1): 92, 2020 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-32293473

RESUMEN

BACKGROUND: Increased circulating levels of tumor necrosis factor (TNF) are associated with greater risk of impaired neurodevelopment after preterm birth. In this study, we tested the hypothesis that systemic TNF inhibition, using the soluble TNF receptor Etanercept, would attenuate neuroinflammation in preterm fetal sheep exposed to lipopolysaccharide (LPS). METHODS: Chronically instrumented preterm fetal sheep at 0.7 of gestation were randomly assigned to receive saline (control; n = 7), LPS infusion (100 ng/kg i.v. over 24 h then 250 ng/kg/24 h for 96 h plus 1 µg LPS boluses at 48, 72, and 96 h, to induce inflammation; n = 8) or LPS plus two i.v. infusions of Etanercept (2 doses, 5 mg/kg infused over 30 min, 48 h apart) started immediately before LPS-exposure (n = 8). Sheep were killed 10 days after starting infusions, for histology. RESULTS: LPS boluses were associated with increased circulating TNF, interleukin (IL)-6 and IL-10, electroencephalogram (EEG) suppression, hypotension, tachycardia, and increased carotid artery perfusion (P < 0.05 vs. control). In the periventricular and intragyral white matter, LPS exposure increased gliosis, TNF-positive cells, total oligodendrocytes, and cell proliferation (P < 0.05 vs control), but did not affect myelin expression or numbers of neurons in the cortex and subcortical regions. Etanercept delayed the rise in circulating IL-6, prolonged the increase in IL-10 (P < 0.05 vs. LPS), and attenuated EEG suppression, hypotension, and tachycardia after LPS boluses. Histologically, Etanercept normalized LPS-induced gliosis, and increase in TNF-positive cells, proliferation, and total oligodendrocytes. CONCLUSION: TNF inhibition markedly attenuated white matter gliosis but did not affect mature oligodendrocytes after prolonged systemic inflammation in preterm fetal sheep. Further studies of long-term brain maturation are now needed.


Asunto(s)
Gliosis/tratamiento farmacológico , Mediadores de Inflamación/antagonistas & inhibidores , Nacimiento Prematuro/tratamiento farmacológico , Inhibidores del Factor de Necrosis Tumoral/administración & dosificación , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Sustancia Blanca/efectos de los fármacos , Animales , Etanercept/administración & dosificación , Femenino , Feto , Gliosis/metabolismo , Mediadores de Inflamación/metabolismo , Infusiones Intravenosas , Embarazo , Nacimiento Prematuro/metabolismo , Ovinos , Factor de Necrosis Tumoral alfa/metabolismo , Sustancia Blanca/metabolismo
4.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R369-R378, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31913689

RESUMEN

Activation of Toll-like receptors (TLRs) after hypoxic-ischemic brain injury can exacerbate injury but also alleviate cell loss, as recently demonstrated with the TLR7 agonist Gardiquimod (GDQ). However, TLR agonists also modulate vascular function and neuronal excitability. Thus, we examined the effects of TLR7 activation with GDQ on cardiovascular function and seizures after asphyxia in preterm fetal sheep at 0.7 gestation (104 days, term ∼147 days). Fetuses received sham asphyxia or asphyxia induced by umbilical cord occlusion for 25 min or asphyxia followed by a continuous intracerebroventricular infusion of 3.34 mg of GDQ from 1 to 4 h after asphyxia. Fetuses were monitored continuously for 72 h postasphyxia. GDQ treatment was associated with sustained, moderate hypertension for 72 h (P < 0.05), with a transient increase in heart rate. Electroencephalographic (EEG) power was suppressed for the entire postasphyxial period in both groups, whereas EEG spectral edge transiently increased during the GDQ infusion compared with asphyxia alone (P < 0.05), with higher ß- and lower δ-EEG frequencies (P < 0.05). This increase in EEG frequency was not related to epileptiform activity. After the GDQ infusion, there was earlier onset of high-amplitude stereotypic evolving seizures, with increased numbers of seizures and seizure burden (P < 0.05). Hemodynamic function and seizure activity are important indices of preterm wellbeing. These data highlight the importance of physiological monitoring during preclinical testing of potential neuroprotective strategies.


Asunto(s)
Aminoquinolinas/toxicidad , Asfixia Neonatal/tratamiento farmacológico , Hipertensión/inducido químicamente , Imidazoles/toxicidad , Fármacos Neuroprotectores/toxicidad , Nacimiento Prematuro , Convulsiones/inducido químicamente , Taquicardia/inducido químicamente , Receptor Toll-Like 7/agonistas , Animales , Animales Recién Nacidos , Asfixia Neonatal/fisiopatología , Presión Sanguínea/efectos de los fármacos , Ondas Encefálicas/efectos de los fármacos , Modelos Animales de Enfermedad , Edad Gestacional , Frecuencia Cardíaca/efectos de los fármacos , Hipertensión/fisiopatología , Medición de Riesgo , Convulsiones/fisiopatología , Oveja Doméstica , Transducción de Señal , Taquicardia/fisiopatología , Factores de Tiempo
5.
Dev Med Child Neurol ; 60(2): 126-133, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29194585

RESUMEN

Children surviving preterm birth have a high risk of disability, particularly cognitive and learning problems. There is extensive clinical and experimental evidence that disability is now primarily related to dysmaturation of white and gray matter, defined by failure of oligodendrocyte maturation and neuronal dendritic arborization, rather than cell death alone. The etiology of this dysmaturation is multifactorial, with contributions from hypoxia-ischemia, infection/inflammation and barotrauma. Intriguingly, these factors can interact to both increase and decrease damage. In this review we summarize preclinical and clinical evidence that all of these factors trigger secondary or chronic inflammation and gliosis. Thus, we hypothesize that these shared pathological features play a key role in a final common pathway that leads to the impaired neural maturation and connectivity and cognitive/motor impairments that are commonly observed in infants born preterm. This raises the possibility that secondary or chronic inflammation may be a viable therapeutic target for delayed interventions to improve neurodevelopmental outcomes after preterm birth. WHAT THIS PAPER ADDS: Hypoxia-ischemia, infection/inflammation, and barotrauma/volutrauma all contribute to preterm brain injury. Multiple different triggers of preterm brain injury are associated with central nervous system dysmaturation. Secondary brain inflammation may be a viable target to improve neurodevelopment after preterm birth.


Asunto(s)
Hipoxia-Isquemia Encefálica/fisiopatología , Enfermedades del Prematuro/fisiopatología , Inflamación/etiología , Animales , Humanos
6.
Growth Factors ; 34(5-6): 203-209, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-28122472

RESUMEN

To investigate the relationship between maternal serum concentrations of placental growth hormone (GH-V), insulin-like growth factor (IGF)-1 and 2, IGF binding proteins (IGFBP)-1 and 3 and birth weight in appropriate-for-gestational-age (AGA), large-for-gestational-age (LGA) and small-for-gestational-age (SGA) cases in a nested case-control study. Maternal serum samples were selected from the Screening for Pregnancy Endpoints (SCOPE) biobank in Auckland, New Zealand. Serum hormone concentrations were determined by ELISA. We found that maternal serum GH-V concentrations at 20 weeks of gestation in LGA pregnancies were significantly higher than in AGA and SGA pregnancies. Maternal GH-V concentrations were positively correlated to birth weights and customized birth weight centiles, while IGFBP-1 concentrations were inversely related to birth weights and customized birth weight centiles. Our findings suggest that maternal serum GH-V and IGFBP-1 concentrations at 20 weeks' gestation are associated with fetal growth.


Asunto(s)
Peso al Nacer , Hormona del Crecimiento/sangre , Hormonas Placentarias/sangre , Adulto , Estudios de Casos y Controles , Femenino , Desarrollo Fetal , Humanos , Recién Nacido de Bajo Peso/sangre , Recién Nacido , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Factor I del Crecimiento Similar a la Insulina/metabolismo , Embarazo
7.
Dev Neurosci ; 37(6): 497-514, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26184807

RESUMEN

Acute exposure to subclinical infection modulates subsequent hypoxia-ischemia (HI) injury in a time-dependent manner, likely by cross-talk through Toll-like receptors (TLRs), but the specific pathways are unclear in the preterm-equivalent brain. In the present study, we tested the hypothesis that repeated low-dose exposure to lipopolysaccharide (LPS) before acute ischemia would be associated with induction of specific TLRs that are potentially neuroprotective. Fetal sheep at 0.65 gestation (term is ∼145 days) received intravenous boluses of low-dose LPS for 5 days (day 1, 50 ng/kg; days 2-5, 100 ng/kg) or the same volume of saline. Either 4 or 24 h after the last bolus of LPS, complete carotid occlusion was induced for 22 min. Five days after LPS, brains were collected. Pretreatment with LPS for 5 days decreased cellular apoptosis, microglial activation and reactive astrogliosis in response to HI injury induced 24 but not 4 h after the last dose of LPS. This was associated with upregulation of TLR4, TLR7 and IFN-ß mRNA, and increased fetal plasma IFN-ß concentrations. The association of reduced white matter apoptosis and astrogliosis after repeated low-dose LPS finishing 24 h but not 4 h before cerebral ischemia, with central and peripheral induction of IFN-ß, suggests the possibility that IFN-ß may be an important mediator of endogenous neuroprotection in the developing brain.


Asunto(s)
Apoptosis/efectos de los fármacos , Encéfalo/embriología , Hipoxia-Isquemia Encefálica/inmunología , Lipopolisacáridos/farmacología , Lesiones Prenatales/inmunología , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 7/inmunología , Animales , Encéfalo/inmunología , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/prevención & control , Modelos Animales de Enfermedad , Feto/efectos de los fármacos , Feto/inmunología , Interferón beta/sangre , Oveja Doméstica
8.
J Neuroinflammation ; 11: 89, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24886663

RESUMEN

BACKGROUND: Perinatal asphyxia and exposure to intrauterine infection are associated with impaired neurodevelopment in preterm infants. Acute exposure to non-injurious infection and/or inflammation can either protect or sensitize the brain to subsequent hypoxia-ischemia. However, the effects of subacute infection and/or inflammation are unclear. In this study we tested the hypothesis that acute-on-chronic exposure to lipopolysaccharide (LPS) would exacerbate white matter injury after subsequent asphyxia in preterm fetal sheep. METHODS: Fetal sheep at 0.7 gestational age received a continuous LPS infusion at 100 ng/kg for 24 hours, then 250 ng/kg/24 hours for 96 hours, plus 1 µg boluses of LPS at 48, 72, and 96 hours or the same volume of saline. Four hours after the last bolus, complete umbilical cord occlusion or sham occlusion was induced for 15 minutes. Sheep were sacrificed 10 days after the start of infusions. RESULTS: LPS exposure was associated with induction of microglia and astrocytes and loss of total and immature and mature oligodendrocytes (n = 9) compared to sham controls (n = 9). Umbilical cord occlusion with saline infusions was associated with induction of microglia, astrogliosis, and loss of immature and mature oligodendrocytes (n = 9). LPS exposure before asphyxia (n = 8) was associated with significantly reduced microglial activation and astrogliosis and improved numbers of immature and mature oligodendrocytes compared to either LPS exposure or asphyxia alone. CONCLUSIONS: Contrary to our initial hypothesis, the combination of acute-on-chronic LPS with subsequent asphyxia reduced neuroinflammation and white matter injury compared with either intervention alone.


Asunto(s)
Asfixia Neonatal/fisiopatología , Inflamación/prevención & control , Leucoencefalopatías/prevención & control , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Modelos Animales de Enfermedad , Embrión de Mamíferos , Endotoxinas/toxicidad , Femenino , Inflamación/etiología , Leucoencefalopatías/etiología , Leucoencefalopatías/patología , Lipopolisacáridos/toxicidad , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Ovinos , Factores de Tiempo
9.
Am J Physiol Regul Integr Comp Physiol ; 306(11): R787-95, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24647590

RESUMEN

Sympathetic nervous system (SNS)-mediated peripheral vasoconstriction plays a key role in initial maintenance of blood pressure during rapid-onset asphyxia in the mammalian fetus, but it is attenuated after the first few minutes. It is unclear whether the SNS response is sustained during the brief, but frequently repeated, episodes of asphyxia characteristic of labor. In the present study, 14 fetal sheep at 0.85 of gestation received either chemical sympathectomy with 6-hydroxydopamine (6-OHDA; n = 7) or sham injection (control; n = 7), followed 4-5 days later by repeated 2-min episodes of complete umbilical cord occlusion every 5 min for up to 4 h or until mean arterial blood pressure (MAP) fell to <20 mmHg for two successive occlusions. In controls, umbilical cord occlusions were associated with a rapid initial fall in fetal heart rate (FHR) and femoral blood flow (FBF), with initial hypertension, followed by progressive development of hypotension during ongoing occlusions. Sympathectomy was associated with attenuation of the initial rise in MAP during umbilical cord occlusion, and after the onset of hypotension, a markedly more rapid fall of MAP to the nadir, with a correspondingly slower fall in FBF (P < 0.05). In contrast, MAP and FHR between successive occlusions were higher after sympathectomy (P < 0.05). There was no significant difference in the number of occlusions before terminal hypotension (6-OHDA; 16.1 ± 2.2 vs. control; 18.7 ± 2.3). These data show that SNS activity provides ongoing support for fetal MAP during prolonged exposure to brief repeated asphyxia.


Asunto(s)
Presión Sanguínea/fisiología , Feto/fisiología , Sistema Nervioso Simpático/fisiología , Arterias Umbilicales/fisiopatología , Cordón Umbilical/irrigación sanguínea , Vasoconstricción/fisiología , Animales , Asfixia/fisiopatología , Femenino , Frecuencia Cardíaca/fisiología , Modelos Animales , Oxidopamina/farmacología , Embarazo , Flujo Sanguíneo Regional/fisiología , Ovinos , Simpatectomía Química/métodos , Sistema Nervioso Simpático/efectos de los fármacos , Simpaticolíticos/farmacología
10.
Am J Physiol Regul Integr Comp Physiol ; 307(4): R387-95, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24944248

RESUMEN

Perinatal exposure to infection is highly associated with adverse outcomes. Experimentally, acute, severe exposure to gram-negative bacterial lipopolysaccharide (LPS) is associated with increased fetal heart rate variability (FHRV). It is unknown whether FHRV is affected by subclinical infection with or without acute exacerbations. We therefore tested the hypothesis that FHRV would be associated with hypotension after acute on chronic exposure to LPS. Chronically instrumented fetal sheep at 0.7 gestation were exposed to a continuous low-dose LPS infusion (n = 12, 100 ng/kg over 24 h, followed by 250 ng·kg(-1)·24 h(-1) for a further 96 h) or the same volume of saline (n = 10). Boluses of either 1 µg LPS or saline were given at 48, 72, and 96 h. Low-dose infusion was not associated with hemodynamic or FHRV changes. The first LPS bolus was associated with tachycardia and suppression of nuchal electromyographic activity in all fetuses. Seven of twelve fetuses developed hypotension (a fall in mean arterial blood pressure ≥5 mmHg). FHRV was transiently increased only at the onset of hypotension, in association with increased cytokine induction and electroencephalogram suppression. FHRV then fell before the nadir of hypotension, with transient suppression of short-term FHRV. After the second LPS bolus, the hypotension group showed a biphasic pattern of a transient increase in FHRV followed by more prolonged suppression. These findings suggest that infection-related hypotension in the preterm fetus mediates the transient increase in FHRV and that repeated exposure to LPS leads to progressive loss of FHRV.


Asunto(s)
Bradicardia/fisiopatología , Corazón Fetal/fisiopatología , Frecuencia Cardíaca Fetal , Hipotensión/fisiopatología , Lipopolisacáridos , Sepsis/fisiopatología , Taquicardia/fisiopatología , Animales , Presión Arterial , Biomarcadores/sangre , Análisis de los Gases de la Sangre , Glucemia/metabolismo , Bradicardia/inducido químicamente , Bradicardia/diagnóstico , Ritmo Circadiano , Modelos Animales de Enfermedad , Electrocardiografía , Electroencefalografía , Electromiografía , Femenino , Sangre Fetal/metabolismo , Monitoreo Fetal/métodos , Edad Gestacional , Concentración de Iones de Hidrógeno , Hipotensión/inducido químicamente , Hipotensión/diagnóstico , Ácido Láctico/sangre , Embarazo , Sepsis/inducido químicamente , Sepsis/diagnóstico , Ovinos , Taquicardia/inducido químicamente , Taquicardia/diagnóstico , Factores de Tiempo
11.
Dev Neurosci ; 35(6): 461-73, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24193164

RESUMEN

To determine whether increased matrix metalloproteinase (MMP) proteolytic activity plays a pathological role in infection/inflammation-induced preterm brain injury, primary cultures of preterm (day 90 of gestation; term 145 days) fetal ovine mixed glia were exposed to 24-96 h of lipopolysaccharide (LPS, 1 µg/ml) or tumour necrosis factor-α (TNF-α, 100 ng/ml). MMP-2 mRNA levels were significantly increased after TNF-α (96 h) and LPS exposure (48 and 96 h), and MMP-9 mRNA levels were significantly increased at 48 and 96 h after TNF-α. On zymography, the active form of secreted MMP-2 was significantly increased 24 h after LPS, but not TNF-α. Both active and latent forms of MMP-9 gelatinolytic activity were significantly increased by TNF-α (96 h) and LPS (72 and 96 h). On reverse zymography, inhibitory activity of TIMP-1 but not TIMP-2 was significantly increased by TNF-α and LPS. SB-3CT-mediated MMP-2 and MMP-9 inhibition transiently reduced LPS-induced oligodendrocyte cell death but had no effect during TNF-α exposure. Collectively, these observations suggest a limited, transient effect of MMPs on immature white matter damage associated with infection but not TNF-α-mediated inflammation.


Asunto(s)
Lipopolisacáridos/farmacología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Neuroglía/metabolismo , Oligodendroglía/metabolismo , Animales , Lesiones Encefálicas/metabolismo , Muerte Celular/fisiología , Células Cultivadas , Humanos , Inflamación/metabolismo , Neuroglía/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Ovinos , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
12.
J Neuroinflammation ; 10: 153, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24344780

RESUMEN

BACKGROUND: White matter injury (WMI) is the major antecedent of cerebral palsy in premature infants, and is often associated with maternal infection and the fetal inflammatory response. The current study explores the therapeutic potential of glutamate receptor blockade or cyclooxygenase-2 (COX-2) inhibition for inflammatory WMI. METHODS: Using fetal ovine derived mixed glia cultures exposed to tumour necrosis factor-α (TNF-α) or lipopolysaccharide (LPS), the expression of alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionate (AMPA) and N-methyl D-aspartate (NMDA) glutamate receptors and their contribution to inflammation mediated pre-oligodendrocyte (OL) death was evaluated. The functional significance of TNF-α and COX-2 signalling in glutamate release in association with TNF-α and LPS exposure was also assessed. RESULTS: AMPA and NMDA receptors were expressed in primary mixed glial cultures on developing OLs, the main cell-type present in fetal white matter at a period of high risk for WMI. We show that glutamate receptor expression and configuration are regulated by TNF-α and LPS exposure, but AMPA and NMDA blockade, either alone or in combination, did not reduce pre-OL death. Furthermore, we demonstrate that glutamate and prostaglandin E2 (PGE2) release following TNF-α or LPS are mediated by a TNF-α-COX-2 dependent mechanism. CONCLUSIONS: Overall, these findings suggest that glial-localised glutamate receptors likely play a limited role in OL demise associated with chronic inflammation, but supports the COX-2 pathway as a potential therapeutic target for infection/inflammatory-mediated WMI.


Asunto(s)
Lesiones Encefálicas/metabolismo , Dinoprostona/metabolismo , Ácido Glutámico/metabolismo , Neuroglía/metabolismo , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Western Blotting , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Encefalitis/inducido químicamente , Encefalitis/metabolismo , Feto , Inmunohistoquímica , Lipopolisacáridos/toxicidad , Radioinmunoensayo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Glutamato/metabolismo , Ovinos , Factor de Necrosis Tumoral alfa/toxicidad
13.
Am J Physiol Regul Integr Comp Physiol ; 304(3): R189-97, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23235324

RESUMEN

Acute, high-dose exposure to endotoxin lipopolysaccharide (LPS) in preterm fetal sheep can trigger periventricular white matter lesions (PVL), in association with severe hypotension/hypoxemia and significant mortality. Intriguingly, however, chronic or repeated exposure to LPS can induce tachyphylaxis. We therefore tested the hypothesis that progressive, acute on chronic fetal infection would be associated with white matter injury with little fetal mortality. Chronically instrumented preterm (0.7 gestational age) fetal sheep were exposed to a continuous low-dose LPS infusion (100 ng over 24 h, followed by 250 ng/24 h for 96 h) or saline. Boluses of 1 µg LPS or saline were given at 48, 72, and 96 h; sheep were killed at day 10. Six of 11 fetal sheep exposed to saline infusion + LPS boluses died 4-7 h after the first bolus. In contrast, there was no fetal mortality after saline infusions alone (n = 9), low-dose LPS infusion + saline boluses (n = 5), or low-dose LPS + LPS boluses (n = 9). Low-dose LPS infusion + LPS boluses was associated with greater microglial induction than low-dose LPS + saline boluses but a similar area of periventricular white matter inflammation. One fetus developed severe focal white matter necrosis after LPS infusion + boluses. The acute cardiovascular compromise associated with high-dose, acute exposure to LPS is markedly attenuated by previous low-dose infusions, with limited apparent exacerbation of periventricular white matter injury compared with low-dose infusion alone.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Endotoxinas/toxicidad , Fibras Nerviosas Mielínicas/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Animales Recién Nacidos , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Embarazo , Nacimiento Prematuro , Ovinos , Tasa de Supervivencia , Pruebas de Toxicidad Aguda , Pruebas de Toxicidad Crónica
14.
Ann Neurol ; 71(1): 121-32, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22275258

RESUMEN

OBJECTIVE: Connexin hemichannels can open during ischemia, resulting in loss of membrane potential, calcium influx, and release of glutamate. In this study, we tested the hypothesis that opening of hemichannels after cerebral ischemia may contribute to delayed evolution of injury. METHODS: We infused a mimetic peptide that blocks connexin 43 hemichannels into the lateral ventricle of chronically instrumented fetal sheep in utero at 128 ± 1 days gestation (term is 147 days), starting 90 minutes after 30 minutes of severe ischemia induced by reversible bilateral carotid artery occlusion, for either 1 or 25 hours. Sheep were killed 7 days later. RESULTS: Peptide infusion was associated with a graded improvement in recovery of electroencephalographic power after 7 days recovery, from -13 ± 1.9 dB (n = 7) after ischemia-vehicle to -9 ± 1.6 dB (n = 7) after ischemia-short infusion and -5 ± 1.6 dB after ischemia-long infusion (n = 6, p < 0.05). Peptide infusion was associated with reduced seizure activity after ischemia, less frequent status epilepticus (p < 0.05), and earlier return of sleep state cycling (p < 0.05). Ischemia-long infusion (but not ischemia-short infusion) was associated with improved survival of oligodendrocytes in intragyral and periventricular white matter (p < 0.05) and increased brain weight (p < 0.05). Ischemia-long infusion was associated with an intermediate estimate of surviving neurons in the parasagittal cortex of 2.9 ± 0.8 × 10(6), in comparison to sham control (4.3 ± 0.9 × 10(6)) or ischemia-vehicle (1.5 ± 0.4 × 10(6); p < 0.05 vs sham control). INTERPRETATION: These data support the hypothesis that opening of connexin hemichannels is a significant mediator of postischemic white and gray matter dysfunction and injury.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevención & control , Conexina 43/antagonistas & inhibidores , Hipoxia Fetal/tratamiento farmacológico , Fragmentos de Péptidos/administración & dosificación , Animales , Isquemia Encefálica/fisiopatología , Supervivencia Celular/fisiología , Conexina 43/fisiología , Modelos Animales de Enfermedad , Electroencefalografía , Femenino , Hipoxia Fetal/metabolismo , Hipoxia Fetal/patología , Marcación de Gen , Masculino , Neuronas/patología , Neuronas/fisiología , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/uso terapéutico , Embarazo , Convulsiones/prevención & control , Ovinos , Fases del Sueño/fisiología , Resultado del Tratamiento
15.
Dev Neurosci ; 34(1): 30-42, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22627272

RESUMEN

Astrocytes, microglial cells and oligodendrocytes (OLs) have been employed separately in vitro to assess cellular pathways following a variety of stimuli. Mixed glial cell cultures, however, have not been utilized to the same extent, despite the observed discrepancy in outcomes resulting from cell-to-cell contact of different glia in culture. Our objective was to standardize and morphologically characterize a primary culture of preterm ovine glial cells in order to attain a relevant in vitro model to assess the intracellular effects of infection and inflammation. This would provide a high-throughput model necessary for in-depth studies on the various pathophysiological mechanisms of white matter injury (WMI), which may occur in the preterm infant as a consequence of maternal infection or the fetal inflammatory response. Glial cells from the forebrains of 0.65-gestation ovine fetuses (comparable to 24- to 26-week human fetal brain development) were mechanically and enzymatically isolated and plated at a final density of 250,000 cells per well. When reaching confluence at 5 days after plating, the cultures contained astrocytes, microglial cells, as well as progenitor, precursor and immature OLs. Glial cell morphology and phenotypic immunoreactivity were characteristic of and consistent with previous observations of separately cultured cell types. To determine the effects of infection or inflammation in our in vitro model, we then treated mixed glial cultures with tumour necrosis factor-α (TNF-α; 50 or 100 ng/ml) or lipopolysaccharide (LPS; 1 µg/ml) for a period of 48 h. Cytokine levels were measured by ELISA and cell numbers for specific glial cell types were determined along with OL proliferation and apoptosis by Ki67 and caspase-3 immunocytochemistry, respectively. Our results showed that exposure to TNF-α or LPS resulted in a characteristic inflammatory response entailed by up-regulation of pro-inflammatory cytokines, a lack of astrogliosis and a marked reduction in OLs attributable to increased apoptosis. In LPS-treated cultures, there was a marked increase in the pro-inflammatory cytokine TNF-α at both 24 and 48 h. In conclusion, this is the first report of the immunocytochemical description and characterization of fetal ovine-derived mixed glial cell primary cultures. This in vitro model provides a novel and efficient system to explore the mechanisms of infection/inflammation-mediated WMI at the cellular level and for screening candidate therapeutic strategies.


Asunto(s)
Modelos Animales de Enfermedad , Encefalitis/complicaciones , Leucoencefalopatías/etiología , Leucoencefalopatías/patología , Prosencéfalo/patología , Oveja Doméstica , Animales , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Astrocitos/metabolismo , Caspasa 3/análisis , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Citocinas/análisis , Citocinas/metabolismo , Encefalitis/patología , Femenino , Feto , Humanos , Infecciones/metabolismo , Antígeno Ki-67/análisis , Antígeno Ki-67/metabolismo , Leucoencefalopatías/metabolismo , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo , Modelos Biológicos , Neuroglía/efectos de los fármacos , Neuroglía/inmunología , Neuroglía/metabolismo , Oligodendroglía/efectos de los fármacos , Oligodendroglía/inmunología , Oligodendroglía/metabolismo , Embarazo , Factor de Necrosis Tumoral alfa/farmacología
16.
Dev Neurosci ; 34(5): 417-27, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23171520

RESUMEN

Perinatal hypoxic ischemic (HI) brain injury is a leading cause of long-term neurological handicap in newborn babies. Recently, excessive activity of matrix metalloproteinases (MMPs), and in particular MMP-9, has been implicated in the aetiology of HI injuries to the immature brain. Our previous study suggested that MMP-9 may be involved in the development of the delayed injury processes following HI injury to the developing brain. Given this, we therefore propose that MMP-9 may be a useful target for rescue therapies in the injured developing brain. To address this, we chose to use SB-3CT, a highly selective inhibitor that is known to target only MMP-2 and MMP-9, to attenuate the elevated MMP-9 activity seen following HI injury to the developing brain. Twenty-one-day-old postnatal Wistar rats were subjected to unilateral carotid artery occlusion followed by exposure to hypoxia (8% oxygen for 1 h). SB-3CT (50 mg/kg body weight in 25% dimethyl sulphoxide/75% polyethylene glycol) or an equal volume of vehicle or saline diluent was then administered intraperitoneally at 2, 5 and 14 h following the insult. Gelatin zymography revealed that pro-MMP-9 levels were significantly reduced at 6 h following hypoxic ischaemia (p ≤ 0.05). However, our results showed that despite significantly inhibiting brain pro-MMP-9 activity after hypoxic ischaemia, SB-3CT failed to confer significant neuroprotection in postnatal day 21 rats 3 days after an HI insult. Further investigations are warranted using a recently reported selective water-soluble version of SB-3CT or another MMP-9 selective inhibitor to resolve the role of MMP-9 in the aetiology of HI injury in the developing brain.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Inhibidores Enzimáticos/farmacología , Compuestos Heterocíclicos con 1 Anillo/farmacología , Hipoxia-Isquemia Encefálica/enzimología , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Sulfonas/farmacología , Animales , Conducta Animal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Infarto Cerebral/tratamiento farmacológico , Infarto Cerebral/enzimología , Cromatografía Líquida de Alta Presión , Inhibidores Enzimáticos/farmacocinética , Compuestos Heterocíclicos con 1 Anillo/farmacocinética , Hipoxia-Isquemia Encefálica/complicaciones , Metaloproteinasa 2 de la Matriz/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/prevención & control , Ratas , Ratas Wistar , Sulfonas/farmacocinética , Espectrometría de Masas en Tándem
17.
Am J Physiol Regul Integr Comp Physiol ; 303(3): R270-8, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22696578

RESUMEN

Exposure to chorioamnionitis is strongly associated with neurodevelopmental disability after premature birth; however, it remains unclear whether subclinical infection affects functional EEG maturation. Chronically instrumented 103-104-day-old (0.7 gestational age: term 147 days) fetal sheep in utero were randomized to receive either gram-negative LPS by continuous low-dose infusion (100 ng iv over 24 h, followed by 250 ng/24 h for 4 days; n = 6) or the same volume of normal saline (n = 9). Arterial plasma cortisol, ACTH, and IL-6 were measured. The delta (0-3.9 Hz), theta (4-7.9 Hz), alpha (8-12.9 Hz), and beta (13-22 Hz) components of the EEG were determined by power spectral analysis. Brains were taken after 10 days for histopathology. There were no changes in blood gases, cardiovascular variables, or EEG power during LPS infusion, but a transient rise in plasma cortisol and IL-6 (P < 0.05). LPS infusion was associated with loss of the maturational increase to higher frequency activity, with reduced alpha and beta power, and greater delta power than saline controls from 6 to 10 days (P < 0.05). Histologically, LPS was associated with increased numbers of microglia and TNF-α-positive cells in the periventricular white matter and frontoparietal cortex, increased caspase-3-positive cells in white matter, but no loss of CNPase-positive oligodendrocytes, Nurr-1 subplate cells, or gyral complexity. These data suggest that low-dose endotoxin exposure can impair EEG maturation in preterm fetal sheep in association with neural inflammation but without hemodynamic disturbances or cortical injury.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/embriología , Electroencefalografía/efectos de los fármacos , Endotoxinas/farmacología , Feto/efectos de los fármacos , Feto/embriología , Ritmo alfa/efectos de los fármacos , Ritmo alfa/fisiología , Animales , Ritmo beta/efectos de los fármacos , Ritmo beta/fisiología , Encéfalo/citología , Caspasa 3/metabolismo , Ritmo Delta/efectos de los fármacos , Ritmo Delta/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Feto/fisiología , Lipopolisacáridos/farmacología , Microglía/citología , Modelos Animales , Embarazo , Ovinos , Factor de Necrosis Tumoral alfa/metabolismo
18.
Dev Neurosci ; 33(3-4): 189-98, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21968048

RESUMEN

Sick preterm and term newborns are highly vulnerable to neural injury, and thus there has been a major search for new, safe and efficacious neuroprotective interventions in recent decades. Preclinical studies are essential to select candidate drugs for clinical trials in humans. This article focuses on 'negative' preclinical studies, i.e. studies where significant differences cannot be detected. Such findings are critical to inform both clinical and preclinical investigators, but historically they have been difficult to publish. A significant amount of time and resources is lost when negative results or nonpromising therapeutics are replicated in separate laboratories because these negative results were not shared with the research community in an open and accessible format. In this article, we discuss approaches to strengthen conclusions from negative preclinical studies and, conversely, to reduce false-negative preclinical evaluations of potential therapeutic compounds. Without being exhaustive, we address three major issues in conducting and interpreting preclinical experiments, including: (a) the choice of animal models, (b) the experimental design, and (c) issues concerning statistical analyses of the experiments. This general introduction is followed by synopses of negative data obtained from studies of three potential therapeutics for perinatal brain injury: (1) the somatostatin analog octreotide, (2) an AMPA/kainate receptor antagonist, topiramate, and (3) a pyruvate derivative, ethyl pyruvate.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/patología , Recién Nacido , Recien Nacido Prematuro , Fármacos Neuroprotectores/farmacología , Investigación , Animales , Ensayos Clínicos como Asunto , Humanos , Modelos Animales , Investigación/estadística & datos numéricos , Proyectos de Investigación , Ovinos
19.
Front Neurosci ; 15: 744840, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34630028

RESUMEN

This comprehensive review focuses on our current understanding of the proposed physiological and pathological functions of extracellular vesicles (EVs) in the developing brain. Furthermore, since EVs have attracted great interest as potential novel cell-free therapeutics, we discuss advances in the knowledge of stem cell- and astrocyte-derived EVs in relation to their potential for protection and repair following perinatal brain injury. This review identified 13 peer-reviewed studies evaluating the efficacy of EVs in animal models of perinatal brain injury; 12/13 utilized mesenchymal stem cell-derived EVs (MSC-EVs) and 1/13 utilized astrocyte-derived EVs. Animal model, method of EV isolation and size, route, timing, and dose administered varied between studies. Notwithstanding, EV treatment either improved and/or preserved perinatal brain structures both macroscopically and microscopically. Additionally, EV treatment modulated inflammatory responses and improved brain function. Collectively this suggests EVs can ameliorate, or repair damage associated with perinatal brain injury. These findings warrant further investigation to identify the optimal cell numbers, source, and dosage regimens of EVs, including long-term effects on functional outcomes.

20.
Stem Cells Transl Med ; 10(3): 427-440, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33103374

RESUMEN

There is increasing evidence that administration of many types of stem cells, including human amnion epithelial cells (hAECs), can reduce hypoxic-ischemic injury, including in the perinatal brain. However, the therapeutic window for single dose treatment is not known. We compared the effects of early and delayed intracerebroventricular administration of hAECs in fetal sheep at 0.7 gestation on brain injury induced by 25 minutes of complete umbilical cord occlusion (UCO) or sham occlusion. Fetuses received either 1 × 106 hAECs or vehicle alone, as an infusion over 1 hour, either 2 or 24 hours after UCO. Fetuses were killed for brain histology at 7 days post-UCO. hAEC infusion at both 2 and 24 hours had dramatic anti-inflammatory and anti-gliotic effects, including significantly attenuating the increase in microglia after UCO in the white and gray matter and the number of astrocytes in the white matter. Both protocols partially improved myelination, but had no effect on total or immature/mature numbers of oligodendrocytes. Neuronal survival in the hippocampus was increased by hAEC infusion at either 2 or 24 hours, whereas only hAECs at 24 hours were associated with improved neuronal survival in the striatum and thalamus. Neither protocol improved recovery of electroencephalographic (EEG) power. These data suggest that a single infusion of hAECs is anti-inflammatory, anti-gliotic, and neuroprotective in preterm fetal sheep when given up to 24 hours after hypoxia-ischemia, but was associated with limited white matter protection after 7 days recovery and no improvement in the recovery of EEG power.


Asunto(s)
Amnios , Gliosis , Hipoxia Encefálica/terapia , Trasplante de Células Madre , Amnios/citología , Animales , Células Epiteliales/trasplante , Femenino , Feto , Gliosis/terapia , Humanos , Embarazo , Ovinos , Células Madre , Cordón Umbilical
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda