Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neuroinflammation ; 20(1): 124, 2023 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-37226206

RESUMEN

BACKGROUND: Antenatal infection/inflammation is associated with disturbances in neuronal connectivity, impaired cortical growth and poor neurodevelopmental outcomes. The pathophysiological substrate that underpins these changes is poorly understood. We tested the hypothesis that progressive inflammation in late gestation fetal sheep would alter cortical neuronal microstructure and neural function assessed using electroencephalogram band power analysis. METHODS: Fetal sheep (0.85 of gestation) were surgically instrumented for continuous electroencephalogram (EEG) recording and randomly assigned to repeated saline (control; n = 9) or LPS (0 h = 300 ng, 24 h = 600 ng, 48 h = 1200 ng; n = 8) infusions to induce inflammation. Sheep were euthanised 4 days after the first LPS infusion for assessment of inflammatory gene expression, histopathology and neuronal dendritic morphology in the somatosensory cortex. RESULTS: LPS infusions increased delta power between 8 and 50 h, with reduced beta power from 18 to 96 h (P < 0.05 vs. control). Basal dendritic length, numbers of dendritic terminals, dendritic arborisation and numbers of dendritic spines were reduced in LPS-exposed fetuses (P < 0.05 vs. control) within the somatosensory cortex. Numbers of microglia and interleukin (IL)-1ß immunoreactivity were increased in LPS-exposed fetuses compared with controls (P < 0.05). There were no differences in total numbers of cortical NeuN + neurons or cortical area between the groups. CONCLUSIONS: Exposure to antenatal infection/inflammation was associated with impaired dendritic arborisation, spine number and loss of high-frequency EEG activity, despite normal numbers of neurons, that may contribute to disturbed cortical development and connectivity.


Asunto(s)
Corteza Cerebral , Electroencefalografía , Inflamación , Animales , Femenino , Embarazo , Feto , Inflamación/inducido químicamente , Lipopolisacáridos/toxicidad , Microglía , Ovinos , Dendritas , Corteza Cerebral/crecimiento & desarrollo
2.
Int J Mol Sci ; 24(4)2023 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-36835117

RESUMEN

Therapeutic hypothermia significantly improves outcomes after neonatal hypoxic-ischemic (HI) encephalopathy but is only partially protective. There is evidence that cortical inhibitory interneuron circuits are particularly vulnerable to HI and that loss of interneurons may be an important contributor to long-term neurological dysfunction in these infants. In the present study, we examined the hypothesis that the duration of hypothermia has differential effects on interneuron survival after HI. Near-term fetal sheep received sham ischemia or cerebral ischemia for 30 min, followed by cerebral hypothermia from 3 h after ischemia end and continued up to 48 h, 72 h, or 120 h recovery. Sheep were euthanized after 7 days for histology. Hypothermia up to 48 h recovery resulted in moderate neuroprotection of glutamate decarboxylase (GAD)+ and parvalbumin+ interneurons but did not improve survival of calbindin+ cells. Hypothermia up to 72 h recovery was associated with significantly increased survival of all three interneuron phenotypes compared with sham controls. By contrast, while hypothermia up to 120 h recovery did not further improve (or impair) GAD+ or parvalbumin+ neuronal survival compared with hypothermia up to 72 h, it was associated with decreased survival of calbindin+ interneurons. Finally, protection of parvalbumin+ and GAD+ interneurons, but not calbindin+ interneurons, with hypothermia was associated with improved recovery of electroencephalographic (EEG) power and frequency by day 7 after HI. The present study demonstrates differential effects of increasing the duration of hypothermia on interneuron survival after HI in near-term fetal sheep. These findings may contribute to the apparent preclinical and clinical lack of benefit of very prolonged hypothermia.


Asunto(s)
Infarto Cerebral , Hipotermia Inducida , Hipoxia-Isquemia Encefálica , Animales , Infarto Cerebral/patología , Infarto Cerebral/terapia , Hipotermia Inducida/métodos , Hipoxia-Isquemia Encefálica/patología , Interneuronas/patología , Isquemia/patología , Isquemia/terapia , Parvalbúminas , Ovinos
3.
Int J Mol Sci ; 24(23)2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38069249

RESUMEN

Seizures are common in preterm newborns and are associated with poor neurodevelopmental outcomes. Current anticonvulsants have poor efficacy, and many have been associated with upregulation of apoptosis in the developing brain. Apigenin, a natural bioactive flavonoid, is a potent inhibitor of hyaluronidase and reduces seizures in adult animal models. However, its impact on perinatal seizures is unclear. In the present study, we examined the effect of apigenin and S3, a synthetic, selective hyaluronidase inhibitor, on seizures after cerebral ischemia in preterm fetal sheep at 0.7 gestation (98-99 days, term ~147 days). Fetuses received sham ischemia (n = 9) or ischemia induced by bilateral carotid occlusion for 25 min. Immediately after ischemia, fetuses received either a continuous infusion of vehicle (0.036% dimethyl sulfoxide, n = 8) or apigenin (50 µM, n = 6). In a pilot study, we also tested infusion of S3 (2 µM, n = 3). Fetuses were monitored continuously for 72 h after ischemia. Infusion of apigenin or S3 were both associated with reduced numbers of animals with seizures, total seizure time, and mean seizure burden. S3 was also associated with a reduction in the total number of seizures over the 72 h recovery period. In animals that developed seizures, apigenin was associated with earlier cessation of seizures. However, apigenin or S3 treatment did not alter recovery of electroencephalographic power or spectral edge frequency. These data support that targeting brain hyaluronidase activity with apigenin or S3 may be an effective strategy to reduce perinatal seizures following ischemia. Further studies are required to determine their effects on neurohistological outcomes.


Asunto(s)
Apigenina , Hipoxia-Isquemia Encefálica , Embarazo , Femenino , Ovinos , Animales , Apigenina/farmacología , Apigenina/uso terapéutico , Hialuronoglucosaminidasa , Proyectos Piloto , Convulsiones/tratamiento farmacológico , Feto/patología , Isquemia , Electroencefalografía , Hipoxia-Isquemia Encefálica/patología
4.
J Neuroinflammation ; 18(1): 189, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34465372

RESUMEN

BACKGROUND: Increased systemic and tissue levels of interleukin (IL)-1ß are associated with greater risk of impaired neurodevelopment after birth. In this study, we tested the hypothesis that systemic IL-1 receptor antagonist (Ra) administration would attenuate brain inflammation and injury in near-term fetal sheep exposed to lipopolysaccharide (LPS). METHODS: Chronically instrumented near-term fetal sheep at 0.85 of gestation were randomly assigned to saline infusion (control, n = 9), repeated LPS infusions (0 h = 300 ng, 24 h = 600 ng, 48 h = 1200 ng, n = 8) or repeated LPS plus IL-1Ra infusions (13 mg/kg infused over 4 h) started 1 h after each LPS infusion (n = 9). Sheep were euthanized 4 days after starting infusions for histology. RESULTS: LPS infusions increased circulating cytokines and were associated with electroencephalogram (EEG) suppression with transiently reduced mean arterial blood pressure, and increased carotid artery perfusion and fetal heart rate (P < 0.05 vs. control for all). In the periventricular and intragyral white matter, LPS-exposure increased IL-1ß immunoreactivity, numbers of caspase 3+ cells and microglia, reduced astrocyte and olig-2+ oligodendrocyte survival but did not change numbers of mature CC1+ oligodendrocytes, myelin expression or numbers of neurons in the cortex and subcortical regions. IL-1Ra infusions reduced circulating cytokines and improved recovery of EEG activity and carotid artery perfusion. Histologically, IL-1Ra reduced microgliosis, IL-1ß expression and caspase-3+ cells, and improved olig-2+ oligodendrocyte survival. CONCLUSION: IL-1Ra improved EEG activity and markedly attenuated systemic inflammation, microgliosis and oligodendrocyte loss following LPS exposure in near-term fetal sheep. Further studies examining the long-term effects on brain maturation are now needed.


Asunto(s)
Encéfalo/efectos de los fármacos , Encefalitis/tratamiento farmacológico , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Lipopolisacáridos/farmacología , Oligodendroglía/efectos de los fármacos , Sustancia Blanca/efectos de los fármacos , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encefalitis/metabolismo , Encefalitis/patología , Femenino , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Oligodendroglía/metabolismo , Oligodendroglía/patología , Embarazo , Ovinos , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
5.
Brain Behav Immun ; 94: 338-356, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33307171

RESUMEN

Severe postnatal systemic infection is highly associated with persistent disturbances in brain development and neurobehavioral outcomes in survivors of preterm birth. However, the contribution of less severe but prolonged postnatal infection and inflammation to such disturbances is unclear. Further, the ability of modern imaging techniques to detect the underlying changes in cellular microstructure of the brain in these infants remains to be validated. We used high-field ex-vivo MRI, neurohistopathology, and behavioral tests in newborn rats to demonstrate that prolonged postnatal systemic inflammation causes subtle, persisting disturbances in brain development, with neurodevelopmental delays and mild motor impairments. Diffusion-tensor MRI and neurite orientation dispersion and density imaging (NODDI) revealed delayed maturation of neocortical and subcortical white matter microstructure. Analysis of pyramidal neurons showed that the cortical deficits involved impaired dendritic arborization and spine formation. Analysis of oligodendrocytes showed that the white matter deficits involved impaired oligodendrocyte maturation and axonal myelination. These findings indicate that prolonged postnatal inflammation, without severe infection, may critically contribute to the diffuse spectrum of brain pathology and subtle long-term disability in preterm infants, with a cellular mechanism involving oligodendrocyte and neuronal dysmaturation. NODDI may be useful for clinical detection of these microstructural deficits.


Asunto(s)
Neocórtex , Nacimiento Prematuro , Sustancia Blanca , Animales , Encéfalo/diagnóstico por imagen , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro , Inflamación , Neocórtex/diagnóstico por imagen , Embarazo , Ratas , Sustancia Blanca/diagnóstico por imagen
6.
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
7.
Int J Mol Sci ; 22(8)2021 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-33924540

RESUMEN

Despite the prevalence of preterm brain injury, there are no established neuroprotective strategies to prevent or alleviate mild-to-moderate inflammation-related brain injury. Perinatal infection and inflammation have been shown to trigger acute neuroinflammation, including proinflammatory cytokine release and gliosis, which are associated with acute and chronic disturbances in brain cell survival and maturation. These findings suggest the hypothesis that the inhibition of peripheral immune responses following infection or nonspecific inflammation may be a therapeutic strategy to reduce the associated brain injury and neurobehavioral deficits. This review provides an overview of the neonatal immunity, neuroinflammation, and mechanisms of inflammation-related brain injury in preterm infants and explores the safety and efficacy of anti-inflammatory agents as potentially neurotherapeutics.


Asunto(s)
Antiinflamatorios/uso terapéutico , Lesiones Encefálicas/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/inmunología , Citocinas/metabolismo , Humanos , Recién Nacido , Recien Nacido Prematuro , Inflamación/complicaciones , Modelos Biológicos
8.
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
9.
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
10.
Pediatr Res ; 88(1): 48-56, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31234193

RESUMEN

BACKGROUND: Therapeutic hypothermia is partially protective for neonatal hypoxic-ischemic encephalopathy (HIE). Damage to the white matter tracts is highly associated with adverse outcomes after HIE, but the effectiveness and optimal duration of hypothermia to attenuate axonal injury are unclear. METHODS: Near-term fetal sheep were randomized to sham control or cerebral ischemia for 30 min with normothermia or cerebral hypothermia from 3 to either 48 or 72 h. Sheep were killed after 7 days. SMI-312-labeled axons and myelin basic protein were quantified in the intragyral white matter of the first and second parasagittal gyri. RESULTS: Ischemia was associated with reduced axonal and myelin area fraction (p < 0.05); loss of axonal and myelin linearity (p < 0.05); and thin, sparse axons, with spheroids, compared to dense, linear morphology in sham controls and associated with induction of microglia in an amoeboid morphology. Both ischemia-48 h hypothermia and ischemia-72 h hypothermia improved axonal area fraction and linearity (p < 0.05), although abnormal morphological features were seen in a subset. Microglial induction was partially suppressed by ischemia-48 h hypothermia, with a ramified morphology. CONCLUSIONS: These data suggest that therapeutic hypothermia can alleviate post-ischemic axonopathy, in part by suppressing secondary inflammation.


Asunto(s)
Axones/fisiología , Encéfalo/embriología , Encéfalo/fisiopatología , Hipotermia Inducida/métodos , Animales , Axones/patología , Análisis de los Gases de la Sangre , Isquemia Encefálica/fisiopatología , Electroencefalografía , Feto/metabolismo , Hipotermia/fisiopatología , Hipoxia-Isquemia Encefálica/terapia , Inflamación , Microglía/metabolismo , Vaina de Mielina/metabolismo , Oligodendroglía/patología , Ovinos , Factores de Tiempo , Sustancia Blanca/patología
11.
Pediatr Int ; 62(7): 770-778, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32119180

RESUMEN

Therapeutic hypothermia is now well established to partially reduce disability in term and near-term infants with moderate-severe hypoxic-ischemic encephalopathy. Preclinical and clinical studies have confirmed that current protocols for therapeutic hypothermia are near optimal. The challenge is now to identify complementary therapies that can further improve outcomes, in combination with therapeutic hypothermia. Overall, anti-excitatory and anti-apoptotic agents have shown variable or even no benefit in combination with hypothermia, suggesting overlapping mechanisms of neuroprotection. Inflammation appears to play a critical role in the pathogenesis of injury in the neonatal brain, and thus, there is potential for drugs with immunomodulatory properties that target inflammation to be used as a therapy in neonates. In this review, we examine the evidence for neuroprotection with immunomodulation after hypoxia-ischemia. For example, stem cell therapy can reduce inflammation, increase cell survival, and promote cell maturation and repair. There are also encouraging preclinical data from small animals suggesting that stem cell therapy can augment hypothermic neuroprotection. However, there is conflicting evidence, and rigorous testing in translational animal models is now needed.


Asunto(s)
Hipotermia Inducida/métodos , Hipoxia-Isquemia Encefálica/terapia , Inmunomodulación , Animales , Antiinflamatorios/uso terapéutico , Lesiones Encefálicas/terapia , Frío , Terapia Combinada , Humanos , Hipoxia-Isquemia Encefálica/complicaciones , Factores Inmunológicos/uso terapéutico , Recién Nacido , Inflamación/complicaciones , Inflamación/terapia , Neuroprotección , Células Madre
12.
Int J Mol Sci ; 21(18)2020 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-32899855

RESUMEN

Perinatal hypoxia-ischemia is associated with disruption of cortical gamma-aminobutyric acid (GABA)ergic interneurons and their surrounding perineuronal nets, which may contribute to persisting neurological deficits. Blockade of connexin43 hemichannels using a mimetic peptide can alleviate seizures and injury after hypoxia-ischemia. In this study, we tested the hypothesis that connexin43 hemichannel blockade improves the integrity of cortical interneurons and perineuronal nets. Term-equivalent fetal sheep received 30 min of bilateral carotid artery occlusion, recovery for 90 min, followed by a 25-h intracerebroventricular infusion of vehicle or a mimetic peptide that blocks connexin hemichannels or by a sham ischemia + vehicle infusion. Brain tissues were stained for interneuronal markers or perineuronal nets. Cerebral ischemia was associated with loss of cortical interneurons and perineuronal nets. The mimetic peptide infusion reduced loss of glutamic acid decarboxylase-, calretinin-, and parvalbumin-expressing interneurons and perineuronal nets. The interneuron and perineuronal net densities were negatively correlated with total seizure burden after ischemia. These data suggest that the opening of connexin43 hemichannels after perinatal hypoxia-ischemia causes loss of cortical interneurons and perineuronal nets and that this exacerbates seizures. Connexin43 hemichannel blockade may be an effective strategy to attenuate seizures and may improve long-term neurological outcomes after perinatal hypoxia-ischemia.


Asunto(s)
Conexina 43/efectos de los fármacos , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Péptidos/farmacología , Animales , Biomimética/métodos , Isquemia Encefálica/tratamiento farmacológico , Infarto Cerebral/tratamiento farmacológico , Conexina 43/antagonistas & inhibidores , Conexina 43/metabolismo , Conexinas/antagonistas & inhibidores , Conexinas/metabolismo , Matriz Extracelular/metabolismo , Femenino , Feto/metabolismo , Hipoxia/fisiopatología , Infusiones Intraventriculares , Interneuronas/metabolismo , Masculino , Parvalbúminas/metabolismo , Péptidos/administración & dosificación , Embarazo , Convulsiones/tratamiento farmacológico , Convulsiones/fisiopatología , Convulsiones/prevención & control , Ovinos
13.
Int J Mol Sci ; 21(23)2020 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-33255257

RESUMEN

Progressive fetal infection/inflammation is strongly associated with neural injury after preterm birth. We aimed to test the hypotheses that progressively developing fetal inflammation leads to neuroinflammation and impaired white matter development and that the histopathological changes can be detected using high-field diffusion tensor magnetic resonance imaging (MRI). Chronically instrumented preterm fetal sheep at 0.7 of gestation were randomly assigned to receive intravenous saline (control; n = 6) or a progressive infusion of lipopolysaccharide (LPS, 200 ng intravenous over 24 h then doubled every 24 h for 5 days to induce fetal inflammation, n = 7). Sheep were killed 10 days after starting the infusions, for histology and high-field diffusion tensor MRI. Progressive LPS infusion was associated with increased circulating interleukin (IL)-6 concentrations and moderate increases in carotid artery perfusion and the frequency of electroencephalogram (EEG) activity (p < 0.05 vs. control). In the periventricular white matter, fractional anisotropy (FA) was increased, and orientation dispersion index (ODI) was reduced (p < 0.05 vs. control for both). Histologically, in the same brain region, LPS infusion increased microglial activation and astrocyte numbers and reduced the total number of oligodendrocytes with no change in myelination or numbers of immature/mature oligodendrocytes. Numbers of astrocytes in the periventricular white matter were correlated with increased FA and reduced ODI signal intensities. Astrocyte coherence was associated with increased FA. Moderate astrogliosis, but not loss of total oligodendrocytes, after progressive fetal inflammation can be detected with high-field diffusion tensor MRI.


Asunto(s)
Gliosis/diagnóstico por imagen , Inflamación/diagnóstico por imagen , Leucoencefalopatías/diagnóstico por imagen , Imagen por Resonancia Magnética , Animales , Gliosis/fisiopatología , Gliosis/veterinaria , Inflamación/fisiopatología , Inflamación/veterinaria , Leucoencefalopatías/fisiopatología , Leucoencefalopatías/veterinaria , Ovinos , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/fisiopatología
14.
Dev Neurosci ; 40(2): 145-161, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29680832

RESUMEN

Fatty acid-binding proteins (FABPs) are a family of transport proteins that facilitate intracellular transport of fatty acids. Despite abundant expression in the brain, the role that FABPs play in the process of cell proliferation and migration in the subventricular zone (SVZ) remains unclear. Our results provide a detailed characterisation of FABP3, 5, and 7 expression in adult and fetal human and sheep SVZ. High FABP5 expression was specifically observed in the adult human SVZ and co-labelled with polysialylated neural cell adhesion molecule (PSA-NCAM), glial fibrillary acidic protein (GFAP), GFAPδ, and proliferating cell nuclear antigen (PCNA), indicating a role for FABP5 throughout the full maturation process of astrocytes and neuroblasts. Some FABP5+ cells had a radial glial-like appearance and co-labelled with the radial glia markers vimentin (40E-C) and GFAP. In the fetal human brain, FABP5 was expressed by radial glia cells throughout the ventricular zone. In contrast, radial glia-like cells in sheep highly expressed FABP3. Taken together, these differences highlight the species-specific expression profile of FABPs in the SVZ. In this study, we demonstrate the distribution of FABP in the adult human SVZ and fetal ventricular zone and reveal its expression on persistent radial glia that may be involved in adult neurogenesis.


Asunto(s)
Células Ependimogliales/metabolismo , Proteínas de Unión a Ácidos Grasos/biosíntesis , Ventrículos Laterales/metabolismo , Neurogénesis/fisiología , Adulto , Anciano , Animales , Femenino , Feto , Humanos , Masculino , Persona de Mediana Edad , Ovinos , Especificidad de la Especie
15.
Dev Neurosci ; 40(3): 258-270, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30179864

RESUMEN

BACKGROUND: Infants born preterm following exposure to in utero inflammation/chorioamnionitis are at high risk of brain injury and life-long neurological deficits. In this study, we assessed the efficacy of early intervention umbilical cord blood (UCB) cell therapy in a large animal model of preterm brain inflammation and injury. We hypothesised that UCB treatment would be neuroprotective for the preterm brain following subclinical fetal inflammation. METHODS: Chronically instrumented fetal sheep at 0.65 gestation were administered lipopolysaccharide (LPS, 150 ng, 055:B5) intravenously over 3 consecutive days, followed by 100 million human UCB mononuclear cells 6 h after the final LPS dose. Controls were administered saline instead of LPS and cells. Ten days after the first LPS dose, the fetal brain and cerebrospinal fluid were collected for analysis of subcortical and periventricular white matter injury and inflammation. RESULTS: LPS administration increased microglial aggregate size, neutrophil recruitment, astrogliosis and cell death compared with controls. LPS also reduced total oligodendrocyte count and decreased mature myelinating oligodendrocytes. UCB cell therapy attenuated cell death and inflammation, and recovered total and mature oligodendrocytes, compared with LPS. CONCLUSIONS: UCB cell treatment following inflammation reduces preterm white matter brain injury, likely mediated via anti-inflammatory actions.


Asunto(s)
Lesiones Encefálicas/terapia , Encefalitis/terapia , Sangre Fetal/citología , Lipopolisacáridos/farmacología , Animales , Corioamnionitis/terapia , Modelos Animales de Enfermedad , Femenino , Feto/citología , Humanos , Microglía/citología , Embarazo , Ovinos , Sustancia Blanca/efectos de los fármacos
16.
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
18.
Dev Neurosci ; 39(1-4): 7-22, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27988510

RESUMEN

In the era of therapeutic hypothermia, reliable preclinical studies are integral to successfully identify neuroprotective strategies to further improve outcomes of encephalopathy at term. We reviewed preclinical neuroprotection studies reported between January 2014 and June 2016 to assess the use of effective temperature monitoring and control. As a secondary measure, we examined whether studies addressed other methodological issues such as stage of brain development, sex differences, the timing of the treatment relative to the insult, and the histological and functional endpoints used after hypoxia-ischemia. The extent and duration of temperature monitoring was highly inconsistent. Only a minority of papers monitored core (19/61; 31%) or brain temperature (3/61; 5%). Most (40/45) of the neuroprotectants either were likely to affect thermoregulation or their impact is unknown. In 85% of papers neonatal rodents were used (67% at P7); 51% of papers did not report the sex of the animals or tested the effect of potential neuroprotectants on just one sex. In 76% of studies, treatment was before or immediately after the insult (within the first 2 h), and few studies assessed long-term histological and behavioral outcomes. In conclusion, many recent preclinical neonatal studies cannot exclude the possibility that apparent neuroprotection might be related to drug-induced hypothermia or to other methodological choices. Close monitoring and control of brain temperature during, as well as for many days after, experimental hypoxia-ischemia are now critical to reliably develop new ways to improve neurodevelopmental outcomes after perinatal hypoxic-ischemic encephalopathy.


Asunto(s)
Hipotermia Inducida/métodos , Hipoxia-Isquemia Encefálica/terapia , Fármacos Neuroprotectores/uso terapéutico , Animales , Asfixia Neonatal/complicaciones , Asfixia Neonatal/terapia , Humanos , Recién Nacido , Proyectos de Investigación , Temperatura
19.
Dev Neurosci ; 37(4-5): 289-304, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25720344

RESUMEN

One of the central, unanswered questions in perinatology is why preterm infants continue to have such poor long-term neurodevelopmental, cognitive and learning outcomes, even though severe brain injury is now rare. There is now strong clinical evidence that one factor underlying disability may be infection, as well as nonspecific inflammation, during fetal and early postnatal life. In this review, we examine the experimental evidence linking both acute and chronic infection/inflammation with perinatal brain injury and consider key experimental determinants, including the microglia response, relative brain and immune maturity and the pattern of exposure to infection. We highlight the importance of the origin and derivation of the bacterial cell wall component lipopolysaccharide. Such experimental paradigms are essential to determine the precise time course of the inflammatory reaction and to design targeted neuroprotective strategies to protect the perinatal brain from infection and inflammation.


Asunto(s)
Encefalopatías , Modelos Animales de Enfermedad , Enfermedades Fetales , Enfermedades del Prematuro , Infecciones , Inflamación , Animales , Humanos
20.
Ann Neurol ; 75(4): 508-24, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24395459

RESUMEN

OBJECTIVE: Recently, we reported that the neocortex displays impaired growth after transient cerebral hypoxia-ischemia (HI) at preterm gestation that is unrelated to neuronal death but is associated with decreased dendritic arbor complexity of cortical projection neurons. We hypothesized that these morphological changes constituted part of a more widespread neuronal dysmaturation response to HI in the caudate nucleus (CN), which contributes to motor and cognitive disability in preterm survivors. METHODS: Ex vivo magnetic resonance imaging (MRI), immunohistochemistry, and Golgi staining defined CN growth, cell death, proliferation, and dendritic maturation in preterm fetal sheep 4 weeks after HI. Patch-clamp recording was used to analyze glutamatergic synaptic currents in CN neurons. RESULTS: MRI-defined growth of the CN was reduced after ischemia compared to controls. However, no significant acute or delayed neuronal death was seen in the CN or white matter. Nor was there significant loss of calbindin-positive medium spiny projection neurons (MSNs) or CN interneurons expressing somatostatin, calretinin, parvalbumin, or tyrosine hydroxylase. Morphologically, ischemic MSNs showed a markedly immature dendritic arbor, with fewer dendritic branches, nodes, endings, and spines. The magnitude and kinetics of synaptic currents, and the relative contribution of glutamate receptor subtypes in the CN were significantly altered. INTERPRETATION: The marked MSN dendritic and functional abnormalities after preterm cerebral HI, despite the marked resistance of immature CN neurons to cell death, are consistent with widespread susceptibility of projection neurons to HI-induced dysmaturation. These global disturbances in dendritic maturation and glutamatergic synaptic transmission suggest a new mechanism for long-term motor and behavioral disabilities in preterm survivors via widespread disruption of neuronal connectivity.


Asunto(s)
Isquemia Encefálica/patología , Núcleo Caudado/patología , Hipoxia Fetal/patología , Regulación del Desarrollo de la Expresión Génica/fisiología , Neuronas/patología , Nacimiento Prematuro/fisiopatología , Potenciales de Acción/efectos de los fármacos , Animales , Isquemia Encefálica/sangre , Caspasa 3/metabolismo , Dendritas/patología , Dendritas/ultraestructura , Modelos Animales de Enfermedad , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , GABAérgicos/farmacología , Cabras , Antígeno Ki-67/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/ultraestructura , Embarazo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA