Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
PLoS One ; 17(5): e0268282, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35544542

RESUMO

BACKGROUND: There is evidence from various models of hypoxic-ischemic injury (HII) that nitric oxide (NO) is protective. We hypothesized that either inhaled NO (iNO) or nitrite would alleviate brain injury in neonatal HII via modulation of mitochondrial function. METHODS: We tested the effects of iNO and nitrite on the Rice-Vannucci model of HII in 7-day-old rats. Brain mitochondria were isolated for flow cytometry, aconitase activity, electron paramagnetic resonance, and Seahorse assays. RESULTS: Pretreatment of pups with iNO decreased survival in the Rice-Vannucci model of HII, while iNO administered post-insult did not. MRI analysis demonstrated that pre-HII iNO at 40 ppm and post-HII iNO at 20 ppm decreased the brain lesion sizes from 6.3±1.3% to 1.0±0.4% and 1.8±0.8%, respectively. Intraperitoneal nitrite at 0.165 µg/g improved neurobehavioral performance but was harmful at higher doses and had no effect on brain infarct size. NO reacted with complex IV at the heme a3 site, decreased the oxidative stress of mitochondria challenged with anoxia and reoxygenation, and suppressed mitochondrial oxygen respiration. CONCLUSIONS: This study suggests that iNO administered following neonatal HII may be neuroprotective, possibly via its modulation of mitochondrial function.


Assuntos
Óxido Nítrico , Nitritos , Administração por Inalação , Animais , Animais Recém-Nascidos , Hipóxia , Ratos
2.
Cell Rep ; 31(6): 107622, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32402283

RESUMO

To date, no stem cell therapy has been directed to specific recipients-and, conversely, withheld from others-based on a clinical or molecular profile congruent with that cell's therapeutic mechanism-of-action (MOA) for that condition. We address this challenge preclinically with a prototypical scenario: human neural stem cells (hNSCs) against perinatal/neonatal cerebral hypoxic-ischemic injury (HII). We demonstrate that a clinically translatable magnetic resonance imaging (MRI) algorithm, hierarchical region splitting, provides a rigorous, expeditious, prospective, noninvasive "biomarker" for identifying subjects with lesions bearing a molecular profile indicative of responsiveness to hNSCs' neuroprotective MOA. Implanted hNSCs improve lesional, motor, and/or cognitive outcomes only when there is an MRI-measurable penumbra that can be forestalled from evolving into necrotic core; the core never improves. Unlike the core, a penumbra is characterized by a molecular profile associated with salvageability. Hence, only lesions characterized by penumbral > core volumes should be treated with cells, making such measurements arguably a regenerative medicine selection biomarker.


Assuntos
Biomarcadores/metabolismo , Lesões Encefálicas/terapia , Medicina Regenerativa/métodos , Transplante de Células-Tronco/métodos , Animais , Modelos Animais de Doenças , Ratos , Ratos Sprague-Dawley
3.
ASN Neuro ; 6(6)2014.
Artigo em Inglês | MEDLINE | ID: mdl-25424430

RESUMO

While hypothermia (HT) is the standard-of-care for neonates with hypoxic ischemic injury (HII), the mechanisms underlying its neuroprotective effect are poorly understood. We examined ischemic core/penumbra and cytokine/chemokine evolution in a 10-day-old rat pup model of HII. Pups were treated for 24 hr after HII with HT (32℃; n = 18) or normothermia (NT, 35℃; n = 15). Outcomes included magnetic resonance imaging (MRI), neurobehavioral testing, and brain cytokine/chemokine profiling (0, 24, 48, and 72 hr post-HII). Lesion volumes (24 hr) were reduced in HT pups (total 74%, p < .05; penumbra 68%, p < .05; core 85%, p = .19). Lesion volumes rebounded at 72 hr (48 hr post-HT) with no significant differences between NT and HT pups. HT reduced interleukin-1ß (IL-1ß) at all time points (p < .05); monocyte chemoattractant protein-1 (MCP-1) trended toward being decreased in HT pups (p = .09). The stem cell signaling molecule, stromal cell-derived factor-1 (SDF-1) was not altered by HT. Our data demonstrate that HT reduces total and penumbral lesion volumes (at 24 and 48 hr), potentially by decreasing IL-1ß without affecting SDF-1. Disassociation between the increasing trend in HII volumes from 48 to 72 hr post-HII when IL-1ß levels remained low suggests that after rewarming, mechanisms unrelated to IL-1ß expression are likely to contribute to this delayed increase in injury. Additional studies should be considered to determine what these mechanisms might be and also to explore whether extending the duration or degree of HT might ameliorate this delayed increase in injury.


Assuntos
Citocinas/metabolismo , Hipotermia Induzida/métodos , Hipóxia-Isquemia Encefálica/terapia , Análise de Variância , Animais , Animais Recém-Nascidos , Imagem de Difusão por Ressonância Magnética , Modelos Animais de Doenças , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/patologia , Imageamento por Ressonância Magnética , Transtornos dos Movimentos/etiologia , Transtornos dos Movimentos/prevenção & controle , Exame Neurológico , Ratos , Ratos Sprague-Dawley , Análise de Regressão
4.
Pediatr Res ; 75(5): 603-11, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24463490

RESUMO

BACKGROUND: Gender is increasingly recognized as an important influence on brain development, disease susceptibility, and response to pharmacologic/rehabilitative treatments. In regenerative medicine, it remains entirely unknown whether there is an interaction between transplanted stem cells and host gender that might bias efficacy and safety in some patients but not others. METHODS: We examined the role of recipient gender in a neonatal rat hypoxic-ischemic injury (HII) model, treated with female human neuronal stem cells (hNSCs), labeled with superparamagnetic iron oxide particles implanted into the contralateral cerebral ventricle. We monitored HII evolution (by magnetic resonance imaging, histopathology, behavioral testing) and hNSC fate (migration, replication, viability). RESULTS: Recipient gender after implantation did not influence the volume or location of ischemic injury (1, 30, or 90 d) or behavior (90 d). Superparamagnetic iron oxide labeling did not influence HII evolution. Implantation had its greatest benefit on mild/moderate injuries, which remained stable rather than increasing as in severe HII as is the natural history for such lesions. CONCLUSION: Our results suggest that hNSC treatment (including using hNSCs that are prelabeled with iron to allow tracking in real time by magnetic resonance imaging) would be equally safe and effective for male and female human newborns with mild-to-moderate HII.


Assuntos
Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/terapia , Células-Tronco Neurais/citologia , Fatores Sexuais , Transplante de Células-Tronco , Animais , Comportamento Animal , Feminino , Compostos Férricos/química , Imageamento por Ressonância Magnética , Masculino , Células-Tronco Neurais/transplante , Ratos
5.
J Cereb Blood Flow Metab ; 32(12): 2161-70, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22929436

RESUMO

Neonatal hypoxic-ischemic brain injury (HII) and arterial ischemic stroke (AIS) result in irreversibly injured (core) and salvageable (penumbral) tissue regions. Identification and reliable quantification of salvageable tissue is pivotal to any effective and safe intervention. Magnetic resonance imaging (MRI) is the current standard to distinguish core from penumbra using diffusion-perfusion mismatch (DPM). However, subtle MR signal variations between core-penumbral regions make their visual delineation difficult. We hypothesized that computational analysis of MRI data provides a more accurate assessment of core and penumbral tissue evolution in HII/AIS. We used two neonatal rat-pup models of HII/AIS (unilateral and global hypoxic-ischemia) and clinical data sets from neonates with AIS to test our noninvasive, automated computational approach, Hierarchical Region Splitting (HRS), to detect and quantify ischemic core-penumbra using only a single MRI modality (T2- or diffusion-weighted imaging, T2WI/DWI). We also validated our approach by comparing core-penumbral images (from HRS) to DPM with immunohistochemical validation of HII tissues. Our translational and clinical data results showed that HRS could accurately and reliably distinguish the ischemic core from penumbra and their spatiotemporal evolution, which may aid in the vetting and execution of effective therapeutic interventions as well as patient selection.


Assuntos
Lesões Encefálicas/patologia , Isquemia Encefálica/patologia , Encéfalo/patologia , Doenças do Recém-Nascido/patologia , Angiografia por Ressonância Magnética , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Lesões Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Recém-Nascido , Doenças do Recém-Nascido/metabolismo , Masculino , Ratos
6.
Ann Neurol ; 69(2): 282-91, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21387373

RESUMO

OBJECTIVE: Quantitative magnetic resonance imaging (MRI) can serially and noninvasively assess the degree of injury in rat pup models of hypoxic ischemic injury (HII). It can also noninvasively monitor stem cell migration following iron oxide prelabeling. Reports have shown that neural stem cells (NSCs) may help mediate neuroprotection or stimulate neuroreparative responses in adult and neonatal models of ischemic injury. We investigated the ability of high-field MRI to monitor and noninvasively quantify the migration, proliferation, and location of iron oxide-labeled NSCs over very long time periods (58 weeks) in real time while contemporaneously correlating this activity with the evolving severity and extent of neural damage. METHODS: Labeled clonal murine NSCs (mNSCs) were implanted 3 days after unilateral HII in 10-day-old rat pups into the contralateral striatum or ventricle. We developed methods for objectively quantifying key aspects of dynamic NSC behavior (eg, viability; extent, and speed of migration; degree of proliferation; extent of integration into host parenchyma). MRI images were validated with histological and immunohistochemical assessments. RESULTS: mNSCs rapidly migrated (100 µm/day) to the lesion site. Chains of migrating NSCs were observed in the corpus callosum. In pups subjected to HII, though not in intact control animals, we observed a 273% increase in the MR-derived volume of mNSCs 4 weeks after implantation (correlating with the known proliferative behavior of endogenous and exogenous NSCs) that slowly declined over the 58-week time course, with no adverse consequences. Large numbers of now quiescent mNSCs remained at the site of injury, many retaining their iron oxide label. INTERPRETATION: Our studies demonstrate that MRI can simultaneously monitor evolving neonatal cerebral injury as well as NSC migration and location. Most importantly, it can noninvasively monitor proliferation dynamically for prolonged time periods. To be able to pursue clinical trials in newborns using stem cell therapies it is axiomatic that safety be insured through the long-term real time monitoring of cell fate and activity, particularly with regard to observing unanticipated risks to the developing brain. This study supports the feasibility of reliably using MRI for this purpose.


Assuntos
Movimento Celular , Proliferação de Células , Hipóxia-Isquemia Encefálica/fisiopatologia , Células-Tronco Neurais/fisiologia , Animais , Diferenciação Celular , Hipóxia-Isquemia Encefálica/patologia , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Aprendizagem em Labirinto/fisiologia , Camundongos , Atividade Motora/fisiologia , Células-Tronco Neurais/patologia , Células-Tronco Neurais/transplante , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Teste de Desempenho do Rota-Rod
7.
J Cereb Blood Flow Metab ; 31(3): 819-31, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20877385

RESUMO

Neuroimaging with diffusion-weighted imaging is routinely used for clinical diagnosis/prognosis. Its quantitative parameter, the apparent diffusion coefficient (ADC), is thought to reflect water mobility in brain tissues. After injury, reduced ADC values are thought to be secondary to decreases in the extracellular space caused by cell swelling. However, the physiological mechanisms associated with such changes remain uncertain. Aquaporins (AQPs) facilitate water diffusion through the plasma membrane and provide a unique opportunity to examine the molecular mechanisms underlying water mobility. Because of this critical role and the recognition that brain AQP4 is distributed within astrocytic cell membranes, we hypothesized that AQP4 contributes to the regulation of water diffusion and variations in its expression would alter ADC values in normal brain. Using RNA interference in the rodent brain, we acutely knocked down AQP4 expression and observed that a 27% AQP4-specific silencing induced a 50% decrease in ADC values, without modification of tissue histology. Our results demonstrate that ADC values in normal brain are modulated by astrocytic AQP4. These findings have major clinical relevance as they suggest that imaging changes seen in acute neurologic disorders such as stroke and trauma are in part due to changes in tissue AQP4 levels.


Assuntos
Aquaporina 4/antagonistas & inibidores , Astrócitos/metabolismo , Água Corporal/metabolismo , Encéfalo/metabolismo , Interferência de RNA , Animais , Aquaporina 4/genética , Difusão , Embrião de Mamíferos , Masculino , Camundongos , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Esferoides Celulares , Técnicas de Cultura de Tecidos , Transfecção
8.
J Cereb Blood Flow Metab ; 29(7): 1305-16, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19436315

RESUMO

We report a new clinically relevant model of neonatal hypoxic-ischemic injury in a 10-day-old rat pup. Bilateral carotid artery occlusion and 8% hypoxia (1 to 15 mins, BCAO-H) was induced with varying degrees of injury (mild, moderate, severe), which was quantified using magnetic resonance imaging including diffusion-weighted and T2-weighted imaging at 24 h and 21/28 days. We developed a magnetic resonance imaging-based rat pup severity score and compared 3D ischemic injury volumes/rat pup severity score with histology and behavioral testing. At 24 h, hypoxic-ischemic injury was observed in 17/27 animals; long-term survival was 81%. Magnetic resonance imaging lesion volumes did not correlate with hypoxia duration but correlated with rat pup severity score, which was used to classify animals into mild (n=21), moderate (n=6), and severe (n=10) groups with average brain lesion volumes of 0.9%, 33.2%, and 56.3%, respectively. Histology confirmed lesion location and histologic scoring correlated with the rat pup severity score. We also found excellent correlation between injury severity and multiple behavioral tasks. Bilateral carotid artery occlusion and hypoxia in the P10 rat pup is an excellent model of neonatal hypoxic-ischemic injury because it induces diffuse global injury similar to the term infant. This model can produce graded injury severity, similar to that seen in human neonates, but manipulation with hypoxia duration is unpredictable.


Assuntos
Doenças das Artérias Carótidas/patologia , Modelos Animais de Doenças , Hipóxia-Isquemia Encefálica/patologia , Animais , Animais Recém-Nascidos , Humanos , Hipóxia , Imageamento por Ressonância Magnética , Ratos , Índice de Gravidade de Doença , Taxa de Sobrevida , Fatores de Tempo
9.
Pediatr Res ; 62(3): 261-6, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17622953

RESUMO

Human serum albumin therapy confers neurobehavioral and histopathologic neuroprotection in adult stroke models. We investigated whether albumin might also be neuroprotective in ischemic brain injury using a transient filament middle cerebral artery occlusion (tfMCAO) model in 10-d-old rat pups treated with 0.25% albumin or saline 1 h after reperfusion. We performed serial neurobehavioral and magnetic resonance imaging (MRI) assessments immediately after tfMCAO (day 0) and on 1, 3, 7, 14, and 28 d. IgG staining to assess blood-brain barrier (BBB) integrity and standard histology was obtained on 1, 3, and 28 d. Hemispheric infarct volumes from MRI were similar in saline and albumin groups (0 h: 39% and 44%; d 1: 46% and 55%; and d 28:10% and 24%) as were neurobehavioral assessments. IgG staining at 3 d post-ischemia showed loss of BBB integrity that was significantly reduced after albumin. Elevated T2 values suggesting vasogenic edema was seen in albumin compared with saline-treated animals, as was increased water mobility (i.e. increased apparent diffusion coefficient (ADC) reflecting cytotoxic edema. The reasons why albumin was not neuroprotective in neonatal stroke compared with adults remain uncertain. Effective strategies in adult models need to be reassessed in the neonate.


Assuntos
Barreira Hematoencefálica/fisiologia , Infarto Cerebral/patologia , Fármacos Neuroprotetores/metabolismo , Albumina Sérica/metabolismo , Acidente Vascular Cerebral/patologia , Adulto , Animais , Animais Recém-Nascidos , Comportamento Animal/fisiologia , Humanos , Infarto da Artéria Cerebral Média , Imageamento por Ressonância Magnética , Permeabilidade , Ratos , Ratos Endogâmicos SHR , Acidente Vascular Cerebral/fisiopatologia
10.
Pediatr Res ; 62(3): 248-54, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17622964

RESUMO

Edema formation can be observed using magnetic resonance imaging (MRI) in patients with stroke. Recent studies have shown that aquaporin-4 (AQP4), a water channel, is induced early after stroke and potentially participates in the development of brain edema. We studied whether induction of AQP4 correlated with edema formation in a rat pup filament stroke model using high field (11.7-Tesla) MRI followed by immunohistochemical investigation of AQP4 protein expression. At 24 h, we observed increased T2 values and decreased apparent diffusion coefficients (ADC) within injured cortical and striatal regions that reflected the edema formation. Coincident with these MR changes were significant increases in AQP4 expression on astrocytic end-feet in the border regions of injured tissues. Striatal imaging findings were still present at 72 h with a slow normalization of AQP4 expression in the border regions. At 28 d, AQP4 expression normalized in the border while in this region ADC values increased. We show that induction of AQP4 is increased during the period of active edema formation in the border region without regional correlation with edema. Finally, induction of AQP4 on astrocyte end-feet could participate in tissue preservation after ischemia in the immature rat brain.


Assuntos
Animais Recém-Nascidos , Aquaporina 4/metabolismo , Edema Encefálico , Acidente Vascular Cerebral , Animais , Astrócitos/metabolismo , Astrócitos/ultraestrutura , Encéfalo/citologia , Encéfalo/metabolismo , Encéfalo/patologia , Edema Encefálico/etiologia , Edema Encefálico/metabolismo , Edema Encefálico/patologia , Modelos Animais de Doenças , Humanos , Imageamento por Ressonância Magnética , Ratos , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Fatores de Tempo
11.
Pediatr Res ; 61(1): 9-14, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17211133

RESUMO

Using an 11.7-Tesla magnetic resonance imaging (MRI) scanner in 10-d-old rat pups we report on the evolution of injury over 28 d in a model of neonatal stroke (transient filament middle cerebral artery occlusion, tfMCAO) and a model of hypoxic-ischemic injury (Rice-Vannucci model, RVM). In both models, diffusion-weighted imaging (DWI) was more sensitive in the early detection of ischemia than T2-weighted imaging (T2WI). Injury volumes in both models were greater on d 1 for DWI and d 3 for T2WI, decreased over time and by d 28 T2WI injury volumes (tfMCAO 10.3% of ipsilateral hemisphere; RVM 23.9%) were definable. The distribution of injury with tfMCAO was confined to the vascular territory of the middle cerebral artery and a definable core and penumbra evolved over time. Ischemic injury in the RVM was more generalized and greater in cortical regions. Contralateral hemispheric involvement was only observed in the RVM. Our findings demonstrate that high-field MRI over extended periods of time is possible in a small animal model of neonatal brain injury and that the tfMCAO model should be used for studies of neonatal stroke and that the RVM does not reflect the vascular distribution of injury seen with focal ischemia.


Assuntos
Modelos Animais de Doenças , Hipóxia-Isquemia Encefálica/fisiopatologia , Imageamento por Ressonância Magnética , Acidente Vascular Cerebral/fisiopatologia , Animais , Animais Recém-Nascidos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA