Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Semin Thorac Cardiovasc Surg ; 35(2): 251-258, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-34995752

RESUMEN

Hypothermic circulatory arrest is a protective technique used when complete cessation of circulation is required during cardiac surgery. Prior efforts to decrease neurologic injury with the NMDA receptor antagonist MK801 were limited by unacceptable side effects. We hypothesized that ketamine would provide neuroprotection without dose-limiting side effects. Canines were peripherally cannulated for cardiopulmonary bypass, cooled to 18°C, and underwent 90 minutes of circulatory arrest. Ketamine-treated canines (n = 5; total dose 2.85 mg/kg) were compared to untreated controls (n = 10). A validated neurobehavioral deficit score was obtained at 24, 48, and 72 hours (0 = no deficits/normal exam; higher score represents increasing deficits). Biomarkers of neuronal injury in the cerebrospinal fluid were examined at baseline and at 8, 24, 48, and 72 hours. Brain histopathologic injury was scored at 72 hours (higher score indicates more necrosis and apoptosis). Ketamine-treated canines had significantly improved, lower neurobehavioral deficit scores compared to controls (overall P = 0.003; 24 hours: median 72 vs 112, P = 0.030; 48 hours: 47 vs 90, P = 0.021; 72 hours: 30 vs 89, P = 0.069). Although the histopathologic injury scores of ketamine-treated canines (median 12) were lower than controls (16), there was no statistical difference (P = 0.10). Levels of phosphorylated neurofilament-H and neuron specific enolase, markers of neuronal injury, were significantly lower in ketamine-treated animals (P = 0.010 and = 0.039, respectively). Ketamine significantly reduced neurologic deficits and biomarkers of injury in canines after hypothermic circulatory arrest. Ketamine represents a safe and approved medication that may be useful as a pharmacologic neuroprotectant during cardiac surgery with circulatory arrest.


Asunto(s)
Hipotermia Inducida , Ketamina , Animales , Perros , Ketamina/toxicidad , Hipotermia Inducida/efectos adversos , Hipotermia Inducida/métodos , Resultado del Tratamiento , Puente Cardiopulmonar/efectos adversos , Biomarcadores , Paro Cardíaco Inducido/efectos adversos , Encéfalo
2.
Cancer Lett ; 517: 35-45, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34098063

RESUMEN

Tumor-associated microglia/macrophages (TAMs) are the main innate immune effector cells in malignant gliomas and have both pro- and anti-tumor functions. The plasticity of TAMs is partially dictated by oncogenic mutations in tumor cells. Heterozygous IDH1 mutation is a cancer driver gene prevalent in grade II/III gliomas, and IDH1 mutant gliomas have relatively favorable clinical outcomes. It is largely unknown how IDH mutation alters TAM phenotypes to influence glioma growth. Here we established clinically relevant isogenic glioma models carrying monoallelic IDH1 R132H mutation (IDH1R132H/WT) and found that IDH1R132H/WT significantly downregulated immune response-related pathways in glioma cells, indicating an immunomodulation role of mutant IDH1. Co-culturing IDH1R132H/WT glioma cells with human macrophages promoted anti-tumor phenotypes of macrophages and increased macrophage migration and phagocytic capacity. In orthotopic xenografts, IDH1R132H/WT decreased tumor growth and prolonged animal survival, accompanied by increased TAM recruitment and upregulated phagocytosis markers, suggesting the induction of anti-tumor TAM functions. Using human cytokine arrays that query 36 proteins, we identified significant downregulation of ICAM-1/CD54 in IDH1R132H/WT gliomas, which was further confirmed by ELISA and immunoblotting analyses. ICAM1 gain-of-function studies revealed that ICAM1 downregulation in IDH1R132H/WT cells played a mechanistic role to mediate the immunomodulation function of IDH1R132H/WT. ICAM-1 silencing in IDH1 wild-type glioma cells decreased tumor growth and increased the anti-tumor function of TAMs. Together, our studies support a new TAM-mediated phagocytic function within IDH1 mutant gliomas, and improved understanding of this process may uncover novel approaches to targeting IDH1 wild type gliomas.


Asunto(s)
Regulación hacia Abajo/genética , Glioma/genética , Molécula 1 de Adhesión Intercelular/genética , Isocitrato Deshidrogenasa/genética , Macrófagos/metabolismo , Microglía/metabolismo , Mutación/genética , Animales , Línea Celular , Línea Celular Tumoral , Femenino , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos Mononucleares , Ratones , Ratones SCID , Células THP-1
3.
J Surg Res ; 264: 260-273, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33839341

RESUMEN

BACKGROUND: Hypothermic circulatory arrest (HCA) is a technique used for complex repair of the aorta, but it can be associated with neurologic morbidity. To better understand the molecular changes that underlie ischemic brain injury, we assessed gene expression and cytokine/chemokine polypeptide concentration in brain tissue and cerebrospinal fluid (CSF) of canines that underwent two hours of HCA. MATERIALS AND METHODS: Adult male canines were cannulated peripherally for cardiopulmonary bypass, cooled to 18°C, and arrested for two hours. Animals were euthanized two, eight, or 24 hours post-HCA (n = 8 per group), and their brains were compared to brains from eight normal canines, using gene expression microarray analysis, cytokine assay, and histopathology. RESULTS: Two to eight hours after HCA, pro-inflammatory cytokine mRNAs increased markedly, and gene expression was enriched within signaling pathways related to neuroinflammation or ischemic injury. Concentrations of pro-inflammatory cytokine polypeptides IL-6, IL-8, IL-1ß, and CCL2 were very low in normal canine brain, whereas anti-inflammatory IL-10 and TGF-ß1 were expressed at moderate levels. Pro-inflammatory cytokine concentrations rose robustly in cerebral tissue and CSF after HCA. IL-6 and IL-8 peaked at eight hours and declined at 24 hours, while IL-1ß and CCL2 remained elevated. Concentrations of anti-inflammatory IL-10 and TGF-ß1 were maintained after HCA, with a significant increase in TGF-ß1 at 24 hours. CONCLUSIONS: These cytokines represent potential diagnostic markers for ischemic neurologic injury that could be used to assess neurologic injury in patients undergoing HCA. The cellular mechanisms underlying this pro-inflammatory, ischemic-induced injury represent potential targets for neuroprotection in the future.


Asunto(s)
Isquemia Encefálica/inmunología , Paro Circulatorio Inducido por Hipotermia Profunda/efectos adversos , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Animales , Biomarcadores/líquido cefalorraquídeo , Biomarcadores/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Encéfalo/patología , Isquemia Encefálica/líquido cefalorraquídeo , Isquemia Encefálica/diagnóstico , Isquemia Encefálica/patología , Citocinas/líquido cefalorraquídeo , Modelos Animales de Enfermedad , Perros , Perfilación de la Expresión Génica , Humanos , Mediadores de Inflamación/líquido cefalorraquídeo , Masculino
4.
J Surg Res ; 260: 177-189, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33348169

RESUMEN

BACKGROUND: Hypothermic circulatory arrest (HCA) is associated with neurologic morbidity, in part mediated by activation of the N-methyl-D-aspartate glutamate receptor causing excitotoxicity and neuronal apoptosis. Using a canine model, we hypothesized that the N-methyl-D-aspartate receptor antagonist MK801 would provide neuroprotection and that MK801 conjugation to dendrimer nanoparticles would improve efficacy. MATERIALS AND METHODS: Male hound dogs were placed on cardiopulmonary bypass, cooled to 18°C, and underwent 90 min of HCA. Dendrimer conjugates (d-MK801) were prepared by covalently linking dendrimer surface OH groups to MK801. Six experimental groups received either saline (control), medium- (0.15 mg/kg) or high-dose (1.56 mg/kg) MK801, or low- (0.05 mg/kg), medium-, or high-dose d-MK801. At 24, 48, and 72 h after HCA, animals were scored by a standardized neurobehavioral paradigm (higher scores indicate increasing deficits). Cerebrospinal fluid was obtained at baseline, eight, 24, 48, and 72 h after HCA. At 72 h, brains were examined for histopathologic injury in a blinded manner (higher scores indicate more injury). RESULTS: Neurobehavioral deficit scores were reduced by low-dose d-MK801 on postoperative day two (P < 0.05) and by medium-dose d-MK801 on postoperative day 3 (P = 0.05) compared with saline controls, but free drug had no effect. In contrast, high-dose free MK801 significantly improved histopathology scores compared with saline (P < 0.05) and altered biomarkers of injury in cerebrospinal fluid, with a significant reduction in phosphorylated neurofilament-H for high-dose MK801 versus saline (P < 0.05). CONCLUSIONS: Treatment with MK-801 demonstrated significant improvement in neurobehavioral and histopathology scores after HCA, although not consistently across doses and conjugates.


Asunto(s)
Paro Circulatorio Inducido por Hipotermia Profunda/efectos adversos , Maleato de Dizocilpina/farmacología , Fármacos Neuroprotectores/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Animales , Encéfalo/patología , Cognición , Perros , Masculino
5.
Cancer Res ; 79(10): 2697-2708, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30898840

RESUMEN

Glioblastomas (GBM) are highly infiltrated by myeloid-derived innate immune cells that contribute to the immunosuppressive nature of the brain tumor microenvironment (TME). CD47 has been shown to mediate immune evasion, as the CD47-SIRPα axis prevents phagocytosis of tumor cells by macrophages and other myeloid cells. In this study, we established CD47 homozygous deletion (CD47-/-) in human and mouse GBM cells and investigated the impact of eliminating the "don't eat me" signal on tumor growth and tumor-TME interactions. CD47 knockout (KO) did not significantly alter tumor cell proliferation in vitro but significantly increased phagocytosis of tumor cells by macrophages in cocultures. Compared with CD47 wild-type xenografts, orthotopic xenografts derived from CD47-/- tumor cells grew significantly slower with enhanced tumor cell phagocytosis and increased recruitment of M2-like tumor-associated microglia/macrophages (TAM). CD47 KO increased tumor-associated extracellular matrix protein tenascin C (TNC) in xenografts, which was further examined in vitro. CD47 loss of function upregulated TNC expression in tumor cells via a Notch pathway-mediated mechanism. Depletion of TNC in tumor cells enhanced the growth of CD47-/- xenografts in vivo and decreased the number of TAM. TNC knockdown also inhibited phagocytosis of CD47-/- tumor cells in cocultures. Furthermore, TNC stimulated release of proinflammatory factors including TNFα via a Toll-like receptor 4 and STAT3-dependent mechanism in human macrophage cells. These results reveal a vital role for TNC in immunomodulation in brain tumor biology and demonstrate the prominence of the TME extracellular matrix in affecting the antitumor function of brain innate immune cells. SIGNIFICANCE: These findings link TNC to CD47-driven phagocytosis and demonstrate that TNC affects the antitumor function of brain TAM, facilitating the development of novel innate immune system-based therapies for brain tumors.


Asunto(s)
Neoplasias Encefálicas/inmunología , Antígeno CD47/inmunología , Glioblastoma/inmunología , Mutación con Pérdida de Función , Fagocitosis , Tenascina/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Antígeno CD47/genética , Línea Celular Tumoral , Glioblastoma/metabolismo , Glioblastoma/patología , Xenoinjertos , Humanos , Inmunidad Innata , Ratones , Ratones Noqueados
6.
Front Neurol ; 9: 304, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29867720

RESUMEN

Perinatal hypoxic-ischemic encephalopathy (HIE) can lead to neurodevelopmental disorders, including cerebral palsy. Standard care for neonatal HIE includes therapeutic hypothermia, which provides partial neuroprotection; magnetic resonance imaging (MRI) is often used to assess injury and predict outcome after HIE. Immature rodent models of HIE are used to evaluate mechanisms of injury and to examine the efficacy and mechanisms of neuroprotective interventions such as hypothermia. In this study, we first confirmed that, in the CD1 mouse model of perinatal HIE used for our research, MRI obtained 3 h after hypoxic ischemia (HI) could reliably assess initial brain injury and predict histopathological outcome. Mice were subjected to HI (unilateral carotid ligation followed by exposure to hypoxia) on postnatal day 7 and were imaged with T2-weighted MRI and diffusion-weighted MRI (DWI), 3 h after HI. Clearly defined regions of increased signal were comparable in T2 MRI and DWI, and we found a strong correlation between T2 MRI injury scores 3 h after HI and histopathological brain injury 7 days after HI, validating this method for evaluating initial injury in this model of HIE. The more efficient, higher resolution T2 MRI was used to score initial brain injury in subsequent studies. In mice treated with hypothermia, we found a significant reduction in T2 MRI injury scores 3 h after HI, compared to normothermic littermates. Early hypothermic neuroprotection was maintained 7 days after HI, in both T2 MRI injury scores and histopathology. In the normothermic group, T2 MRI injury scores 3 h after HI were comparable to those obtained 7 days after HI. However, in the hypothermic group, brain injury was significantly less 7 days after HI than at 3 h. Thus, early neuroprotective effects of hypothermia were enhanced by 7 days, which may reflect the additional 3 h of hypothermia after imaging or effects on later mechanisms of injury, such as delayed cell death and inflammation. Our results demonstrate that hypothermia has early neuroprotective effects in this model. These findings suggest that hypothermia has an impact on early mechanisms of excitotoxic injury and support initiation of hypothermic intervention as soon as possible after diagnosis of HIE.

7.
Nanomedicine ; 13(7): 2359-2369, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28669854

RESUMEN

Perinatal hypoxic-ischemic encephalopathy (HIE) can result in neurodevelopmental disability, including cerebral palsy. The only treatment, hypothermia, provides incomplete neuroprotection. Hydroxyl polyamidoamine (PAMAM) dendrimers are being explored for targeted delivery of therapy for HIE. Understanding the biodistribution of dendrimer-conjugated drugs into microglia, neurons and astrocytes after brain injury is essential for optimizing drug delivery. We conjugated N-acetyl-L-cysteine to Cy5-labeled PAMAM dendrimer (Cy5-D-NAC) and used a mouse model of perinatal HIE to study effects of timing of administration, hypothermia, brain injury, and microglial activation on uptake. Dendrimer conjugation delivered therapy most effectively to activated microglia but also targeted some astrocytes and injured neurons. Cy5-D-NAC uptake was correlated with brain injury in all cell types and with activated morphology in microglia. Uptake was not inhibited by hypothermia, except in CD68+ microglia. Thus, dendrimer-conjugated drug delivery can target microglia, astrocytes and neurons and can be used in combination with hypothermia for treatment of HIE.


Asunto(s)
Acetilcisteína/administración & dosificación , Antioxidantes/administración & dosificación , Dendrímeros/química , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Acetilcisteína/farmacocinética , Acetilcisteína/uso terapéutico , Animales , Animales Recién Nacidos , Antioxidantes/farmacocinética , Antioxidantes/uso terapéutico , Parálisis Cerebral/tratamiento farmacológico , Parálisis Cerebral/patología , Modelos Animales de Enfermedad , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipotermia Inducida , Hipoxia-Isquemia Encefálica/patología , Hipoxia-Isquemia Encefálica/terapia , Ratones , Microglía/efectos de los fármacos , Microglía/patología , Distribución Tisular
8.
J Control Release ; 249: 173-182, 2017 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-28137632

RESUMEN

Hypothermic circulatory arrest (HCA) provides neuroprotection during cardiac surgery but entails an ischemic period that can lead to excitotoxicity, neuroinflammation, and subsequent neurologic injury. Hydroxyl polyamidoamine (PAMAM) dendrimers target activated microglia and damaged neurons in the injured brain, and deliver therapeutics in small and large animal models. We investigated the effect of dendrimer size on brain uptake and explored the pharmacokinetics in a clinically-relevant canine model of HCA-induced brain injury. Generation 6 (G6, ~6.7nm) dendrimers showed extended blood circulation times and increased accumulation in the injured brain compared to generation 4 dendrimers (G4, ~4.3nm), which were undetectable in the brain by 48h after final administration. High levels of G6 dendrimers were found in cerebrospinal fluid (CSF) of injured animals with a CSF/serum ratio of ~20% at peak, a ratio higher than that of many neurologic pharmacotherapies already in clinical use. Brain penetration (measured by drug CSF/serum level) of G6 dendrimers correlated with the severity of neuroinflammation observed. G6 dendrimers also showed decreased renal clearance rate, slightly increased liver and spleen uptake compared to G4 dendrimers. These results, in a large animal model, may offer insights into the potential clinical translation of dendrimers.


Asunto(s)
Encéfalo/metabolismo , Dendrímeros/química , Dendrímeros/farmacocinética , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Sistemas de Liberación de Medicamentos , Animales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Dendrímeros/administración & dosificación , Modelos Animales de Enfermedad , Perros , Portadores de Fármacos/administración & dosificación , Riñón/metabolismo , Hígado/metabolismo , Masculino
9.
Stem Cells Dev ; 25(13): 975-85, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27149035

RESUMEN

Glial-Restricted Precursors (GRPs) are tripotential progenitors that have been shown to exhibit beneficial effects in several preclinical models of neurological disorders, including neonatal brain injury. The mechanisms of action of these cells, however, require further study, as do clinically relevant questions such as timing and route of cell administration. Here, we explored the effects of GRPs on neonatal hypoxia-ischemia during acute and subacute stages, using an in vitro transwell co-culture system with organotypic brain slices exposed to oxygen-glucose deprivation (OGD). OGD-exposed slices that were then co-cultured with GRPs without direct cell contact had decreased tissue injury and cortical cell death, as evaluated by lactate dehydrogenase (LDH) release and propidium iodide (PI) staining. This effect was more pronounced when cells were added during the subacute phase of the injury. Furthermore, GRPs reduced the amount of glutamate in the slice supernatant and changed the proliferation pattern of endogenous progenitor cells in brain slices. In summary, we show that GRPs exert a neuroprotective effect on neonatal hypoxia-ischemia without the need for direct cell-cell contact, thus confirming the rising view that beneficial actions of stem cells are more likely attributable to trophic or immunomodulatory support rather than to long-term integration.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/patología , Neuroglía/citología , Fármacos Neuroprotectores/metabolismo , Células Madre/citología , Animales , Animales Recién Nacidos , Antígenos/metabolismo , Isquemia Encefálica/complicaciones , Bromodesoxiuridina/metabolismo , Muerte Celular , Hipoxia de la Célula , Proliferación Celular , Técnicas de Cocultivo , Proteínas de Dominio Doblecortina , Femenino , Glucosa/deficiencia , Glutamatos/metabolismo , Hipocampo/patología , L-Lactato Deshidrogenasa , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Neuroglía/metabolismo , Neuropéptidos/metabolismo , Técnicas de Cultivo de Órganos , Oxígeno , Embarazo , Proteoglicanos/metabolismo
10.
Glia ; 63(3): 452-65, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25377280

RESUMEN

OBJECTIVE: Neonatal white matter injury (NWMI) is the leading cause of cerebral palsy and other neurocognitive deficits in prematurely-born children, and no restorative therapies exist. Our objective was to determine the fate and effect of glial restricted precursor cell (GRP) transplantation in an ischemic mouse model of NWMI. METHODS: Neonatal CD-1 mice underwent unilateral carotid artery ligation on postnatal-Day 5 (P5). At P22, intracallosal injections of either enhanced green fluorescent protein (eGFP) + GRPs or saline were performed in control and ligated mice. Neurobehavioral and postmortem studies were performed at 4 and 8 weeks post-transplantation. RESULTS: GRP survival was comparable at 1 month but significantly lower at 2 months post-transplantation in NWMI mice compared with unligated controls. Surviving cells showed better migration capability in controls; however, the differentiation capacity of transplanted cells was similar in control and NWMI. Saline-treated NWMI mice showed significantly altered response in startle amplitude and prepulse inhibition (PPI) paradigms compared with unligated controls, while these behavioral tests were completely normal in GRP-transplanted animals. Similarly, there was significant increase in hemispheric myelin basic protein density, along with significant decrease in pathologic axonal staining in cell-treated NWMI mice compared with saline-treated NWMI animals. INTERPRETATION: The reduced long-term survival and migration of transplanted GRPs in an ischemia-induced NWMI model suggests that neonatal ischemia leads to long-lasting detrimental effects on oligodendroglia even months after the initial insult. Despite limited GRP-survival, behavioral, and neuropathological outcomes were improved after GRP-transplantation. Our results suggest that exogenous GRPs improve myelination through trophic effects in addition to differentiation into mature oligodendrocytes.


Asunto(s)
Isquemia Encefálica/fisiopatología , Supervivencia Celular/fisiología , Neuroglía/trasplante , Trasplante de Células Madre , Células Madre/fisiología , Sustancia Blanca/lesiones , Animales , Animales Recién Nacidos , Axones/patología , Axones/fisiología , Encéfalo/patología , Encéfalo/fisiopatología , Isquemia Encefálica/patología , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Básica de Mielina/metabolismo , Neuroglía/fisiología , Médula Espinal/fisiología , Médula Espinal/trasplante , Trasplante de Células Madre/métodos , Resultado del Tratamiento , Sustancia Blanca/patología , Sustancia Blanca/fisiopatología
11.
J Neuropathol Exp Neurol ; 73(12): 1134-43, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25383634

RESUMEN

Neuropathology and neurologic impairment were characterized in a clinically relevant canine model of hypothermic (18°C) circulatory arrest (HCA) and cardiopulmonary bypass (CPB). Adult dogs underwent 2 hours of HCA (n = 39), 1 hour of HCA (n = 20), or standard CPB (n = 22) and survived 2, 8, 24, or 72 hours. Neurologic impairment and neuropathology were much more severe after 2-hour HCA than after 1-hour HCA or CPB; histopathology and neurologic deficit scores were significantly correlated. Apoptosis developed as early as 2 hours after injury and was most severe in the granule cells of the hippocampal dentate gyrus. Necrosis evolved more slowly and was most severe in amygdala and pyramidal neurons in the cornu ammonis hippocampus. Neuronal injury was minimal up to 24 hours after 1-hour HCA, but 1 dog that survived to 72 hours showed substantial necrosis in the hippocampus, suggesting that, with longer survival time, the injury was worse. Although neuronal injury was minimal after CPB, we observed rare apoptotic and necrotic neurons in hippocampi and caudate nuclei. These results have important implications for CPB in humans and may help explain the subtle cognitive changes experienced by patients after CPB.


Asunto(s)
Lesiones Encefálicas/diagnóstico , Lesiones Encefálicas/etiología , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Modelos Animales de Enfermedad , Animales , Perros , Masculino
12.
Dev Neurosci ; 35(2-3): 182-96, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23445614

RESUMEN

Microglial activation in crossing white matter tracts is a hallmark of noncystic periventricular leukomalacia (PVL), the leading pathology underlying cerebral palsy in prematurely born infants. Recent studies indicate that neuroinflammation within an early time window can produce long-lasting defects in oligodendroglial maturation, myelination deficit, as well as disruption of transcription factors important in oligodendroglial maturation. We recently reported an ischemic mouse model of PVL, induced by unilateral neonatal carotid artery ligation, leading to selective long-lasting bilateral myelination deficits, ipsilateral thinning of the corpus callosum, ventriculomegaly, as well as evidence of axonopathy. Here, we report that permanent unilateral carotid ligation on postnatal day 5 in CD-1 mice induces an inflammatory response, as defined by microglial activation and recruitment, as well as significant changes in cytokine expression (increased IL-1ß, IL-6, TGF-ß1, and TNF-α) following ischemia. Transient reduction in counts of oligodendrocyte progenitor cells (OPCs) at 24 and 48 h after ischemia, a shift in OPC cell size and morphology towards the more immature form, as well as likely migration of OPCs were found. These OPC changes were topographically associated with areas showing microglial activation, and OPC counts negatively correlated with increased microglial staining. The presented data show a striking neuroinflammatory response in an ischemia-induced model of PVL, associated with oligodendroglial injury. Future studies modulating the neuroinflammatory response in this model may contribute to a better understanding of the interaction between microglia and OPCs in PVL and open opportunities for future therapies.


Asunto(s)
Encéfalo/patología , Inflamación/patología , Leucomalacia Periventricular/patología , Oligodendroglía/patología , Células Madre/patología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Hipoxia-Isquemia Encefálica/patología , Inmunohistoquímica , Inflamación/complicaciones , Ratones , Microglía/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
World J Clin Pediatr ; 2(3): 16-25, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-25254170

RESUMEN

The Food and Drug Administration (FDA) has approved two mechanism-based treatments for tuberous sclerosis complex (TSC)-everolimus and vigabatrin. However, these treatments have not been systematically studied in individuals with TSC and severe autism. The aim of this review is to identify the clinical features of severe autism in TSC, applicable preclinical models, and potential barriers that may warrant strategic planning in the design phase of clinical trial development. A comprehensive search strategy was formed and searched across PubMed, Embase and SCOPUS from their inception to 2/21/12, 3/16/12, and 3/12/12 respectively. After the final search date, relevant, updated articles were selected from PubMed abstracts generated electronically and emailed daily from PubMed. The references of selected articles were searched, and relevant articles were selected. A search of clinicaltrials.gov was completed using the search term "TSC" and "tuberous sclerosis complex". Autism has been reported in as many as 60% of individuals with TSC; however, review of the literature revealed few data to support clear classification of the severity of autism in TSC. Variability was identified in the diagnostic approach, assessment of cognition, and functional outcome among the reviewed studies and case reports. Objective outcome measures were not used in many early studies; however, diffusion tensor imaging of white matter, neurophysiologic variability in infantile spasms, and cortical tuber subcategories were examined in recent studies and may be useful for objective classification of TSC in future studies. Mechanism-based treatments for TSC are currently available. However, this literature review revealed two potential barriers to successful design and implementation of clinical trials in individuals with severe autism-an unclear definition of the population and lack of validated outcome measures. Recent studies of objective outcome measures in TSC and further study of applicable preclinical models present an opportunity to overcome these barriers.

14.
J Thorac Cardiovasc Surg ; 142(4): 902-910.e1, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21924148

RESUMEN

OBJECTIVES: Ubiquitin carboxyl-terminal esterase-L1 (UCHL1) is a protein highly selectively expressed in neurons and has been linked to neurodegenerative disease in humans. We hypothesize that UCHL1 would be an effective serum biomarker for brain injury as tested in canine models of hypothermic circulatory arrest (HCA) and cardiopulmonary bypass (CPB). METHODS: Dogs were exposed to CPB (n = 14), 1 hour of HCA (1h-HCA; n = 11), or 2 hours of HCA (2h-HCA; n = 20). Cerebrospinal fluid and serum were collected at baseline, 8 hours, and 24 hours after treatment. UCHL1 levels were measured using a sandwich enzyme-linked immunosorbent assay. Neurologic function and histopathology were scored at 24 hours, and UCHL1 immunoreactivity was examined at 8 hours. RESULTS: Baseline UCHL1 protein levels in cerebrospinal fluid and serum were similar for all groups. In serum, UCHL1 levels were elevated at 8 hours after treatment for 2h-HCA subjects compared with baseline values (P < .01) and also compared with CPB dogs at 8 hours (P < .01). A serum UCHL1 level above 3.9 ng/(mg total protein) at 8 hours had the best discriminatory power for predicting functional disability. In cerebrospinal fluid, UCHL1 was elevated in all groups at 8 hours after treatment compared with baseline (P < .01). However, UCHL1 levels in cerebrospinal fluid remained elevated at 24 hours only in 2h-HCA subjects (P < .01). Functional and histopathologic scores were closely correlated (Pearson coefficient, 0.66; P < .01) and were significantly worse in 2h-HCA animals. CONCLUSIONS: This is the first report associating elevated serum UCHL1 with brain injury. The novel neuronal biomarker UCHL1 is increased in serum 8 hours after severe neurologic insult in 2h-HCA animals compared with CPB animals. These results support the potential for use in cardiac surgery patients and form the basis for clinical correlation in humans.


Asunto(s)
Lesiones Encefálicas/enzimología , Puente Cardiopulmonar/efectos adversos , Paro Circulatorio Inducido por Hipotermia Profunda/efectos adversos , Neuronas/enzimología , Ubiquitina Tiolesterasa/sangre , Animales , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Lesiones Encefálicas/etiología , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Modelos Animales de Enfermedad , Perros , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Inmunohistoquímica , Masculino , Examen Neurológico , Neuronas/patología , Factores de Tiempo , Ubiquitina Tiolesterasa/líquido cefalorraquídeo , Regulación hacia Arriba
15.
J Cereb Blood Flow Metab ; 31(10): 2009-18, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21540870

RESUMEN

Periventricular leukomalacia, PVL, is the leading cause of cerebral palsy in prematurely born infants, and therefore more effective interventions are required. The objective of this study was to develop an ischemic injury model of PVL in mice and to determine the feasibility of in vivo magnetization transfer (MT) magnetic resonance imaging (MRI) as a potential monitoring tool for the evaluation of disease severity and experimental therapeutics. Neonatal CD-1 mice underwent unilateral carotid artery ligation on postnatal day 5 (P5); at P60, in vivo T2-weighted (T2w) and MT-MRI were performed and correlated with postmortem histopathology. In vivo T2w MRI showed thinning of the right corpus callosum, but no significant changes in hippocampal and hemispheric volumes. Magnetization transfer MRI revealed significant white matter abnormalities in the bilateral corpus callosum and internal capsule. These quantitative MT-MRI changes correlated highly with postmortem findings of reduced myelin basic protein in bilateral white matter tracts. Ventriculomegaly and persistent astrogliosis were observed on the ligated side, along with evidence of axonopathy and fewer oligodendrocytes in the corpus callosum. We present an ischemia-induced mouse model of PVL, which has pathologic abnormalities resembling autopsy reports in infants with PVL. We further validate in vivo MRI techniques as quantitative monitoring tools that highly correlate with postmortem histopathology.


Asunto(s)
Cuerpo Calloso/diagnóstico por imagen , Cuerpo Calloso/fisiopatología , Modelos Animales de Enfermedad , Leucomalacia Periventricular/diagnóstico por imagen , Leucomalacia Periventricular/fisiopatología , Imagen por Resonancia Magnética/métodos , Animales , Isquemia Encefálica/diagnóstico por imagen , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Parálisis Cerebral/diagnóstico por imagen , Parálisis Cerebral/metabolismo , Parálisis Cerebral/patología , Parálisis Cerebral/fisiopatología , Cuerpo Calloso/metabolismo , Humanos , Recién Nacido , Recien Nacido Prematuro , Leucomalacia Periventricular/metabolismo , Leucomalacia Periventricular/patología , Ratones , Proteína Básica de Mielina/metabolismo , Radiografía , Factores de Tiempo
16.
Lancet Neurol ; 10(4): 372-82, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21435600

RESUMEN

Knowledge of the nature, prognosis, and ways to treat brain lesions in neonatal infants has increased remarkably. Neonatal hypoxic-ischaemic encephalopathy (HIE) in term infants, mirrors a progressive cascade of excito-oxidative events that unfold in the brain after an asphyxial insult. In the laboratory, this cascade can be blocked to protect brain tissue through the process of neuroprotection. However, proof of a clinical effect was lacking until the publication of three positive randomised controlled trials of moderate hypothermia for term infants with HIE. These results have greatly improved treatment prospects for babies with asphyxia and altered understanding of the theory of neuroprotection. The studies show that moderate hypothermia within 6 h of asphyxia improves survival without cerebral palsy or other disability by about 40% and reduces death or neurological disability by nearly 30%. The search is on to discover adjuvant treatments that can further enhance the effects of hypothermia.


Asunto(s)
Hipoxia-Isquemia Encefálica/terapia , Ensayos Clínicos como Asunto , Humanos , Hipotermia Inducida , Hipoxia-Isquemia Encefálica/etiología , Recién Nacido , Pronóstico , Resultado del Tratamiento
17.
ASN Neuro ; 3(2)2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-21382016

RESUMEN

Neuronal death pathways following hypoxia-ischaemia are sexually dimorphic, but the underlying mechanisms are unclear. We examined cell death mechanisms during OGD (oxygen-glucose deprivation) followed by Reox (reoxygenation) in segregated male (XY) and female (XX) mouse primary CGNs (cerebellar granule neurons) that are WT (wild-type) or Parp-1 [poly(ADP-ribose) polymerase 1] KO (knockout). Exposure of CGNs to OGD (1.5 h)/Reox (7 h) caused cell death in XY and XX neurons, but cell death during Reox was greater in XX neurons. ATP levels were significantly lower after OGD/Reox in WT-XX neurons than in XY neurons; this difference was eliminated in Parp-1 KO-XX neurons. AIF (apoptosis-inducing factor) was released from mitochondria and translocated to the nucleus by 1 h exclusively in WT-XY neurons. In contrast, there was a release of Cyt C (cytochrome C) from mitochondria in WT-XX and Parp-1 KO neurons of both sexes; delayed activation of caspase 3 was observed in the same three groups. Thus deletion of Parp-1 shunted cell death towards caspase 3-dependent apoptosis. Delayed activation of caspase 8 was also observed in all groups after OGD/Reox, but was much greater in XX neurons, and caspase 8 translocated to the nucleus in XX neurons only. Caspase 8 activation may contribute to increased XX neuronal death during Reox, via caspase 3 activation. Thus, OGD/Reox induces death of XY neurons via a PARP-1-AIF-dependent mechanism, but blockade of PARP-1-AIF pathway shifts neuronal death towards a caspase-dependent mechanism. In XX neurons, OGD/Reox caused prolonged depletion of ATP and delayed activation of caspase 8 and caspase 3, culminating in greater cell death during Reox.


Asunto(s)
Muerte Celular/fisiología , Glucosa/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Neuronas/fisiología , Oxígeno/metabolismo , Caracteres Sexuales , Transducción de Señal/fisiología , Adenosina Trifosfato/metabolismo , Animales , Factor Inductor de la Apoptosis/metabolismo , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Cerebelo/citología , Femenino , Masculino , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Neuronas/citología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo
18.
Ann Thorac Surg ; 89(6): 1965-71, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20494057

RESUMEN

BACKGROUND: Little is known about the molecular mechanisms of neurologic complications after hypothermic circulatory arrest (HCA) with cardiopulmonary bypass (CPB). Canine genome sequencing allows profiling of genomic changes after HCA and CPB alone. We hypothesize that gene regulation will increase with increased severity of injury. METHODS: Dogs underwent 2-hour HCA at 18 degrees C (n = 10), 1-hour HCA (n = 8), or 2-hour CPB at 32 degrees C alone (n = 8). In each group, half were sacrificed at 8 hours and half at 24 hours after treatment. After neurologic scoring, brains were harvested for genomic analysis. Hippocampal RNA isolates were analyzed using canine oligonucleotide expression arrays containing 42,028 probes. RESULTS: Consistent with prior work, dogs that underwent 2-hour HCA experienced severe neurologic injury. One hour of HCA caused intermediate clinical damage. Cardiopulmonary bypass alone yielded normal clinical scores. Cardiopulmonary bypass, 1-hour HCA, and 2-hour HCA groups historically demonstrated increasing degrees of histopathologic damage (previously published). Exploratory analysis revealed differences in significantly regulated genes (false discovery rate < 10%, absolute fold change > or = 1.2), with increases in differential gene expression with injury severity. At 8 hours and 24 hours after insult, 2-hour HCA dogs had 502 and 1,057 genes regulated, respectively; 1-hour HCA dogs had 179 and 56 genes regulated; and CPB alone dogs had 5 and 0 genes regulated. CONCLUSIONS: Our genomic profile of canine brains after HCA and CPB revealed 1-hour and 2-hour HCA induced markedly increased gene regulation, in contrast to the minimal effect of CPB alone. This adds to the body of neurologic literature supporting the safety of CPB alone and the minimal effect of CPB on a normal brain, while illuminating genomic results of both.


Asunto(s)
Encefalopatías/etiología , Encefalopatías/genética , Puente Cardiopulmonar/efectos adversos , Paro Circulatorio Inducido por Hipotermia Profunda/efectos adversos , Perfilación de la Expresión Génica , Animales , Perros , Puntaje de Gravedad del Traumatismo , Masculino
19.
Clin Perinatol ; 36(4): 835-58, vii, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19944838

RESUMEN

Hypoxia-ischemia in the perinatal period is an important cause of cerebral palsy and associated disabilities in children. There has been significant research progress in hypoxic-ischemic encephalopathy over the last 2 decades, and many new molecular mechanisms have been identified. Despite all these advances, therapeutic interventions are still limited. In this article the authors discuss several molecular pathways involved in hypoxia-ischemia, and potential therapeutic targets.


Asunto(s)
Parálisis Cerebral/etiología , Hipoxia-Isquemia Encefálica , Animales , Antioxidantes/uso terapéutico , Apoptosis , Parálisis Cerebral/prevención & control , Diagnóstico por Imagen , Niños con Discapacidad , Humanos , Hipotermia Inducida/métodos , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/terapia , Recién Nacido , Estrés Oxidativo/fisiología , Pronóstico
20.
Ann Thorac Surg ; 88(2): 543-50, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19632410

RESUMEN

BACKGROUND: The development of specific biomarkers to aid in the diagnosis and prognosis of neuronal injury is of paramount importance in cardiac surgery. Alpha II-spectrin is a structural protein abundant in neurons of the central nervous system and cleaved into signature fragments by proteases involved in necrotic and apoptotic cell death. We measured cerebrospinal fluid alpha II-spectrin breakdown products (alphaII-SBDPs) in a canine model of hypothermic circulatory arrest (HCA) and cardiopulmonary bypass. METHODS: Canine subjects were exposed to either 1 hour of HCA (n = 8; mean lowest tympanic temperature 18.0 +/- 1.2 degrees C) or standard cardiopulmonary bypass (n = 7). Cerebrospinal fluid samples were collected before treatment and 8 and 24 hours after treatment. Using polyacrylamide gel electrophoresis and immunoblotting, SBDPs were isolated and compared between groups using computer-assisted densitometric scanning. Necrotic versus apoptotic cell death was indexed by measuring calpain and caspase-3 cleaved alphaII-SBDPs (SBDP 145+150 and SBDP 120, respectively). RESULTS: Animals undergoing HCA demonstrated mild patterns of histologic cellular injury and clinically detectable neurologic dysfunction. Calpain-produced alphaII-SBDPs (150 kDa+145 kDa bands-necrosis) 8 hours after HCA were significantly increased (p = 0.02) as compared with levels before HCA, and remained elevated at 24 hours after HCA. In contrast, caspase-3 alphaII-SBDP (120 kDa band-apoptosis) was not significantly increased. Animals receiving cardiopulmonary bypass did not demonstrate clinical or histologic evidence of injury, with no increases in necrotic or apoptotic cellular markers. CONCLUSIONS: We report the use of alphaII-SBDPs as markers of neurologic injury after cardiac surgery. Our analysis demonstrates that calpain- and caspase-produced alphaII-SBDPs may be an important and novel marker of neurologic injury after HCA.


Asunto(s)
Biomarcadores/líquido cefalorraquídeo , Lesiones Encefálicas/líquido cefalorraquídeo , Paro Cardíaco Inducido/efectos adversos , Espectrina/líquido cefalorraquídeo , Animales , Apoptosis/fisiología , Ganglios Basales/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Calpaína/metabolismo , Caspasas/metabolismo , Cerebelo/patología , Giro Dentado/patología , Perros , Electroforesis en Gel de Poliacrilamida , Hipotermia Inducida , Immunoblotting , Masculino , Modelos Animales , Necrosis/líquido cefalorraquídeo , Lóbulo Parietal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA