Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
Brain Res ; 1834: 148844, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38432260

RESUMEN

Periventricular leukomalacia (PVL) is a neurological condition observed in premature infants, characterized by hypomyelination and activation of microglia. Maternal inflammation-induced brain injury in offspring significantly contributes to the development of PVL. Currently, there are no clinical pharmaceutical interventions available for pregnant women to prevent maternal inflammation-mediated brain injury in their offspring. Inosine has been shown to modulate the immune response in diverse stressful circumstances, such as injury, ischemia, and inflammation. The aim of this investigation was to examine the potential prophylactic impact of inosine on offspring PVL induced by maternal inflammation. This was accomplished by administering a 1 mg/ml inosine solution (40 ml daily) to pregnant Sprague-Dawley (SD) rats for 16 consecutive days prior to their intraperitoneal injection of lipopolysaccharide (350 µg/kg, once a day, for two days). The results showed that maternal inosine pretreatment significantly reversed the reduction in MBP and CNPase (myelin-related markers), CC-1 and Olig2 (oligodendrocyte-related markers) in their PVL pups (P7), suggesting that inosine administration during pregnancy could improve hypomyelination and enhance the differentiation of oligodendrocyte precursor cells (OPCs) in their PVL pups. Furthermore, the protective mechanism of inosine against PVL is closely associated with the activation and polarization of microglia. This is evidenced by a notable reduction in the quantity of IBA 1-positive microglia, a decrease in the level of CD86 (a marker for M1 microglia), an increase in the level of Arg 1 (a marker for M2 microglia), as well as a decrease in the level of pro-inflammatory factors TNF-α, IL-1ß, and IL-6, and an increase in the level of anti-inflammatory factors IL-4 and IL-10 in the brain of PVL pups following maternal inosine pretreatment. Taken together, inosine pretreatment of pregnant rats can improve hypomyelination in their PVL offspring by triggering the M1/M2 switch of microglia.


Asunto(s)
Inflamación , Inosina , Microglía , Ratas Sprague-Dawley , Animales , Femenino , Embarazo , Microglía/efectos de los fármacos , Microglía/metabolismo , Ratas , Inosina/farmacología , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Leucomalacia Periventricular/metabolismo , Vaina de Mielina/metabolismo , Vaina de Mielina/efectos de los fármacos , Animales Recién Nacidos , Efectos Tardíos de la Exposición Prenatal
2.
Neurochem Res ; 49(1): 129-141, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37642893

RESUMEN

Periventricular leukomalacia (PVL), a predominant form of brain injury in preterm survivors, is characterized by hypomyelination and microgliosis and is also the major cause of long-term neurobehavioral abnormalities in premature infants. Receptor-interacting protein kinase 1 (RIPK1) plays a pivotal role in mediating cell death and inflammatory signaling cascade. However, very little is known about the potential effect of RIPK1 in PVL and the underlying mechanism. Herein, we found that the expression level of RIPK1 was drastically increased in the brain of PVL neonatal mice models, and treatment of PVL neonatal mice with Necrostatin-1s (Nec-1s), an inhibitor of RIPK1, greatly ameliorated cerebral ischemic injury, exhibiting an increase of body weights, reduction of cerebral infarct size, neuronal loss, and occurrence of necrosis-like cells, and significantly improved the long-term abnormal neurobehaviors of PVL. In addition, Nec-1s significantly suppressed hypomyelination and promoted the differentiation of oligodendrocyte precursor cells (OPCs), as demonstrated by the increased expression levels of MBP and Olig2, the decreased expression level of GPR17, a significant increase in the number of CC-1-positive cells, and suppression of myelin ultrastructure impairment. Moreover, the mechanism of neuroprotective effects of Nec-1s against PVL is closely associated with its suppression of the RIPK1-mediated necrosis signaling molecules, RIPK1, PIPK3, and MLKL. More importantly, inhibition of RIPK1 could reduce microglial inflammatory injury by triggering the M1 to M2 microglial phenotype, appreciably decreasing the levels of M1 marker CD86 and increasing the levels of M2 markers Arg1 or CD206 in PVL mice. Taken together, inhibition of RIPK1 markedly ameliorates the brain injury and long-term neurobehavioral abnormalities of PVL mice through the reduction of neural cell necroptosis and reversing neuroinflammation.


Asunto(s)
Lesiones Encefálicas , Leucomalacia Periventricular , Humanos , Recién Nacido , Lactante , Ratones , Animales , Leucomalacia Periventricular/tratamiento farmacológico , Leucomalacia Periventricular/metabolismo , Animales Recién Nacidos , Necroptosis , Necrosis , Inflamación/tratamiento farmacológico , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas del Tejido Nervioso/metabolismo
3.
Ther Hypothermia Temp Manag ; 13(3): 134-140, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36862528

RESUMEN

Periventricular leukomalacia (PVL), characterized by distinctive form of white matter injury, often arises after neonatal cardiac surgery. Proven therapies for PVL are absent. In this study, we designed to quest therapeutic effects of delayed mild hypothermia on PVL and its mechanism in a neonatal rat brain slice model. With the increase of delayed mild hypothermia-treating time, the reduced expression of myelin basic protein and loss of preoligodendrocytes were significantly attenuated after oxygen-glucose deprivation. In addition, the proportion of ionized calcium binding adapter molecule 1 (Iba-1)-positive cells and the expression of Iba-1 were apparently reduced with the increased duration of mild hypothermia treatment. Furthermore, the levels of tumor necrosis factor alpha and interleukin-6 reduced after the mild hypothermia treatment relative to the control. Inhibition of microglial activation with prolonged mild hypothermia may be a potential strategy for white matter protection during cardiopulmonary bypass and hypothermic circulatory arrest.


Asunto(s)
Hipotermia Inducida , Hipotermia , Leucomalacia Periventricular , Células Precursoras de Oligodendrocitos , Ratas , Animales , Animales Recién Nacidos , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/patología , Microglía/metabolismo , Microglía/patología , Hipotermia/metabolismo , Leucomalacia Periventricular/terapia , Leucomalacia Periventricular/metabolismo , Leucomalacia Periventricular/patología , Encéfalo/patología
4.
Bioengineered ; 13(4): 10224-10234, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35436416

RESUMEN

Studies have shown that periventricular leukomalacia (PVL) is a distinctive form of cerebral white matter injury that pertains to myelination disturbances. Maternal inflammation is a main cause of white matter injury. Intrauterine inflammation cellular will be propagated to the developing brain by the entire maternal-placental-fetal axis, and triggers neural immune injury. As a low-affinity receptor, adenosine A2B receptor (A2BAR) requires high concentrations of adenosine to be significantly activated in pathological conditions. We hypothesized that in the maternal inflammation-induced PVL model, a selective A2BAR antagonist PSB0788 had the potential to prevent the injury. In this work, a total of 18 SD pregnant rats were divided into three groups, and treated with intraperitoneal injection of phosphate buffered saline (PBS), lipopolysaccharide (LPS), or LPS+PSB0788. Placental infection was determined by H&E staining and the inflammatory condition was determined by ELISA. Change of MBP, NG2 and CC-1 in the brain of the rats' offspring were detected by western blot and immunohistochemistry. Furthermore, LPS-induced maternal inflammation reduced the expression of MBP, which related to the decrease in the numbers of OPCs and mature oligodendrocytes in neonate rats. After treatment with PSB0788, the levels of MBP proteins increased in the rats' offspring, improved the remyelination. In conclusion, our study shows that the selective A2BAR antagonist PSB0788 plays an important role in promoting the normal development of OPCs in vivo by the maternal inflammation-induced PVL model. Future studies will focus on the mechanism of PSB0788 in this model.


Asunto(s)
Lesiones Encefálicas , Leucomalacia Periventricular , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Recién Nacido , Inflamación , Leucomalacia Periventricular/tratamiento farmacológico , Leucomalacia Periventricular/metabolismo , Leucomalacia Periventricular/patología , Lipopolisacáridos/toxicidad , Placenta/metabolismo , Placenta/patología , Embarazo , Ratas
5.
Bioengineered ; 12(1): 7314-7324, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34569901

RESUMEN

The G protein-coupled receptor-17 (GPR17) plays an important role in regulating the differentiation of oligodendrocytes and remyelination, which is a key negative regulator of oligodendrocyte differentiation. The present study aimed to investigate the function of GPR17 in the white matter of periventricular leukomalacia (PVL) neonatal rats. The PVL model was established in 2-day old neonatal rats by intracerebral injection of LPS (1 mg/kg). Compared to sham, GPR17 was significantly upregulated, while Olig1 was significantly downregulated in the PVL group at 1 d, 3 days, and 7 days post-modeling. Compared to the negative control (NC) group, the expression of GPR17 was suppressed, while that of Olig1 was elevated in the siRNA-GPR17 group as time progressed; the opposite results were observed in the GPR17-overexpressed group. Decreased formation of myelin sheaths as well as poor structure and loose arrangement were observed in the PVL group. Similar observations were found in the PVL + siRNA-GPR17 group at 1 d and 3 days post-modeling. However, on day 7 post-modeling, a dramatic increase in the formation of myelin sheath as well as thicker myelin sheaths were observed in the PVL + siRNA-GPR17 group. The migration ability of oligodendrocyte progenitor cells (OPCs) isolated from animals was found to be significantly suppressed in the GPR17-overexpressed group, accompanied by the downregulation of Olig1. Taken together, the regeneration and repair of myelin sheaths post-PVL white matter injury were induced by downregulating the GPR17 gene, which elevated the expression of Olig1.


Asunto(s)
Técnicas de Silenciamiento del Gen , Leucomalacia Periventricular , Vaina de Mielina/metabolismo , Receptores Acoplados a Proteínas G/genética , Regeneración/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Modelos Animales de Enfermedad , Leucomalacia Periventricular/genética , Leucomalacia Periventricular/metabolismo , Leucomalacia Periventricular/patología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Transgénicas , Receptores Acoplados a Proteínas G/metabolismo
6.
J Neuroinflammation ; 17(1): 365, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33261624

RESUMEN

BACKGROUND: Periventricular leukomalacia (PVL), a devastating brain injury affecting premature infants, is the most common cause of cerebral palsy. PVL is caused by hypoxia ischemia (HI) and is characterized by white matter necrotic lesions, microglial activation, upregulation of NF-κB, and neuronal death. The microglia is the main cell involved in PVL pathogenesis. The goal of this study was to investigate the role of microglial NF-κB activity and its prophylactic inhibition in a neonate mouse model of HI. METHODS: Transgenic mice with specific knockout NF-κB in microglia and colony stimulating factor 1 receptor Cre with floxed IKKß (CSF-1R Cre + IKKßflox/wt ) were used. Postnatal day 5 (P5) mice underwent sham or bilateral temporary carotid artery ligation followed by hypoxia. After HI insult, inflammatory cytokines, volumetric MRI, histopathology, and immunohistochemistry for oligodendroglia and microglial activation markers were analyzed. Long-term neurobehavioral assessment, including grip strength, rotarod, and open field testing, was performed at P60. RESULTS: We demonstrate that selective inhibition of NF-κB in microglia decreases HI-induced brain injury by decreasing microglial activation, proinflammatory cytokines, and nitrative stress. Rescue of oligodendroglia is evidenced by immunohistochemistry, decreased ventriculomegaly on MRI, and histopathology. This selective inhibition leads to attenuation of paresis, incoordination, and improved grip strength, gait, and locomotion. CONCLUSION: We conclude that NF-κb activation in microglia plays a major role in the pathogenesis of hypoxic ischemic injury of the immature brain, and its prophylactic inhibition offers significant neuroprotection. Using a specific inhibitor of microglial NF-κB may offer a new prophylactic or therapeutic alternative in preterm infants affected by HI and possibly other neurological diseases in which microglial activation plays a role.


Asunto(s)
Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , Microglía/metabolismo , FN-kappa B/metabolismo , Animales , Leucomalacia Periventricular/metabolismo , Leucomalacia Periventricular/patología , Ratones , Ratones Noqueados
7.
Neuropharmacology ; 162: 107803, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31580838

RESUMEN

The most common injury of preterm infants is periventricular leukomalacia (PVL) and to date there is still no safe and effective treatment. In our previous studies, leptin has been found to have neuroprotective effects on the preterm ischemia-hypoxia brain damage model rats in animal behavior. To gain insight into the neuroprotective mechanisms of leptin on preterm brain damage model rats, we constructed a comparative peptidomic profiling of hippocampal tissue between leptin-treated after model and preterm ischemia-hypoxia brain damage model rats using a stable isobaric labeling strategy involving tandem mass tag reagents, followed by nano liquid chromatography tandem mass spectrometry. We identified and quantified 4164 peptides, 238 of which were differential expressed in hippocampal tissue in the two groups. A total of 150 peptides were up regulated and 88 peptides were down regulated. These peptides were imported into the Ingenuity Pathway Analysis (IPA) and identified putative roles in nervous system development, function and diseases. We concluded that the preterm ischemia-hypoxia brain damage model with leptin treatment induced peptides changes in hippocampus, and these peptides, especially for the peptides associated "microtubule-associated protein 1b (MAP1b), Elastin (Eln), Piccolo presynaptic cytomatrix protein (Pclo), Zinc finger homeobox 3(Zfhx3), Alpha-kinase 3(Alpk3) and Myosin XVA(Myo15a) ", could be candidate bio-active peptides and participate in neuroprotection of leptin. These may advance our current understanding of the mechanism of leptin's neuroprotective effect on preterm brain damage and may be involved in the etiology of preterm brain damage. Meanwhile, we found that repression of ILK signaling pathway plays a significant role in neuroprotection of leptin. A better understanding of the role of ILK signaling pathway in neuroprotective mechanisms will help scientists and researchers to develop selective, safe and efficacious drug for therapy against human nervous system disorders.


Asunto(s)
Hipocampo/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Leptina/farmacología , Fármacos Neuroprotectores/farmacología , Péptidos/metabolismo , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Arteria Carótida Común , Proteínas del Citoesqueleto/efectos de los fármacos , Proteínas del Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Elastina/efectos de los fármacos , Elastina/metabolismo , Hipocampo/efectos de los fármacos , Proteínas de Homeodominio/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Hipoxia-Isquemia Encefálica/fisiopatología , Leucomalacia Periventricular/metabolismo , Leucomalacia Periventricular/fisiopatología , Ligadura , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Miosinas/efectos de los fármacos , Miosinas/metabolismo , Neuropéptidos/efectos de los fármacos , Neuropéptidos/metabolismo , Péptidos/efectos de los fármacos , Proteínas Serina-Treonina Quinasas , Ratas , Transducción de Señal
8.
Pediatr Neonatol ; 60(4): 435-440, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30559060

RESUMEN

BACKGROUND: To investigate the clinical values of serum melatonin and αII spectrin cleavage products (SBDPs) in assessing the severity of brain injury in preterm infants. METHODS: Sixty-four premature infants in total were selected and classified into the brain injury group (BI, n = 30) and the non-brain injury group (CON, n = 34) according to cranial imaging examination. The serum melatonin and SBDPs were detected by ELISA. All the preterm infants were received NBNA testing at 40 weeks of corrected gestational age. RESULTS: The levels of melatonin and SBDPs in the BI group were significantly higher than the CON group (p < 0.05) and the levels in the infants with severe brain injury were significantly higher than those with mild brain injury (p < 0.05), as well as exhibiting a negative correlation with the NBNA score at 40 weeks of corrected gestational age (p < 0.05). CONCLUSIONS: Detecting melatonin and SBDPs has clinical value in diagnosing and assessing the severity of brain injury in preterm infants.


Asunto(s)
Encéfalo/diagnóstico por imagen , Infarto Cerebral/sangre , Hemorragia Cerebral Intraventricular/sangre , Hemorragias Intracraneales/sangre , Leucomalacia Periventricular/sangre , Melatonina/sangre , Espectrina/sangre , Lesiones Encefálicas/sangre , Lesiones Encefálicas/diagnóstico por imagen , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Estudios de Casos y Controles , Infarto Cerebral/diagnóstico por imagen , Infarto Cerebral/metabolismo , Infarto Cerebral/fisiopatología , Hemorragia Cerebral Intraventricular/diagnóstico por imagen , Hemorragia Cerebral Intraventricular/metabolismo , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/sangre , Enfermedades del Prematuro/diagnóstico por imagen , Enfermedades del Prematuro/metabolismo , Enfermedades del Prematuro/fisiopatología , Hemorragias Intracraneales/diagnóstico por imagen , Hemorragias Intracraneales/metabolismo , Hemorragias Intracraneales/fisiopatología , Leucomalacia Periventricular/diagnóstico por imagen , Leucomalacia Periventricular/metabolismo , Leucomalacia Periventricular/fisiopatología , Imagen por Resonancia Magnética , Masculino , Índice de Severidad de la Enfermedad , Espectrina/metabolismo
9.
J Thorac Cardiovasc Surg ; 156(6): 2271-2280, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30121135

RESUMEN

BACKGROUND: Periventricular leukomalacia is a common white-matter injury after neonatal cardiac surgery; however, its potential cellular mechanism remains uncertain. There is limited study regarding periventricular leukomalacia treatment. METHODS: A neonatal rat brain slice perfusion model was used for reproducing the condition of cardiopulmonary bypass, and oxygen glucose deprivation simulated circulatory arrest. Seven-day-old Sprague-Dawley rats were randomly divided into 7 groups: (1) control group with 36°C; (2) 60 minutes of oxygen glucose deprivation group on 15°C, 25°C, 36°C, respectively; and (3) 60 minutes of oxygen glucose deprivation group on 15°C, 25°C, 36°C, plus minocycline (10 µmol/L), respectively. Immunohistochemistry, Western blot, and inflammatory mediators were compared after the perfusion procedures in the different groups. RESULTS: This neonatal rat brain slice perfusion with oxygen glucose deprivation model could replicate the pathophysiologic process and injury after cardiopulmonary bypass and hypothermic circulatory arrest. With the increase of oxygen glucose deprivation perfusion temperature, we found that both microglia activation and preoligodendrocyte loss increased. The application of minocycline can significantly inhibit microglial activation and preoligodendrocyte cells loss in the normothermic (36°C) and moderate hypothermia (25°C) oxygen glucose deprivation groups (P < .05), with accompanying significant decreasing microglial inflammatory productions; however, no significant improvement was found in the deep hypothermia (15°C) group. CONCLUSIONS: The microglial activation may play a key role in preoligodendrocyte injury in the ex vivo neonatal rat brain slice perfusion and circulatory arrest model. Inhibition of microglial activation with minocycline may be an attractive target for white-matter protection during cardiopulmonary bypass and hypothermic circulatory arrest.


Asunto(s)
Leucomalacia Periventricular/prevención & control , Microglía/efectos de los fármacos , Minociclina/farmacología , Fármacos Neuroprotectores/farmacología , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Animales , Animales Recién Nacidos , Puente Cardiopulmonar/efectos adversos , Hipoxia de la Célula , Supervivencia Celular/efectos de los fármacos , Femenino , Glucosa/deficiencia , Paro Cardíaco Inducido/efectos adversos , Hipotermia Inducida , Técnicas In Vitro , Interleucina-6/metabolismo , Leucomalacia Periventricular/etiología , Leucomalacia Periventricular/metabolismo , Leucomalacia Periventricular/patología , Masculino , Microglía/metabolismo , Microglía/patología , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/patología , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo
10.
Mol Med Rep ; 17(4): 5940-5949, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29436652

RESUMEN

As research into periventricular leukomalacia (PVL) gradually increases, concerns are emerging about long­term neuron injury. The present study aimed to investigate neuronal injury and the relevant alterations in apoptosis and autophagy in a PVL model established previously. A rat model of hypoxia­ischemia­induced PVL was established. In the model group, Sprague­Dawley (SD) rats [postnatal day 3 (P3)] were subjected to right common carotid artery ligation followed by suturing and exposed to 6­8% oxygen for 2 h; in the control group, SD rats (P3) were subjected to right common carotid artery dissection followed by suturing, without ligation and hypoxic exposure. At 1, 3, 7 and 14 days following modeling, brain tissue samples were collected and stained with hematoxylin and eosin. Cellular apoptosis was detected by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and the protein and mRNA expression alterations of neuronal nuclei (NeuN), caspase­3 and Beclin 1 in the model group were detected by western blot analysis and reverse transcription­quantitative polymerase chain reaction (RT­qPCR) analyses. Compared with the control group, the protein and mRNA expression levels of NeuN (a marker of mature neurons) were markedly reduced, the number of positive cells was increased as detected by TUNEL, and the protein and mRNA expression levels of caspase­3 and Beclin 1 were elevated in the model group. In the rat model of hypoxia­ischemia­induced PVL, oligodendrocyte injury and myelinization disorders were observed, in addition to neuron injury, a decrease in mature neurons and the co­presence of apoptosis and autophagy. However, apoptosis and autophagy exist in different phases: Apoptosis is involved in neuron injury, while autophagy is likely to have a protective role.


Asunto(s)
Apoptosis , Autofagia , Hipoxia-Isquemia Encefálica/complicaciones , Leucomalacia Periventricular/etiología , Leucomalacia Periventricular/metabolismo , Neuronas/metabolismo , Animales , Animales Recién Nacidos , Beclina-1/genética , Beclina-1/metabolismo , Biomarcadores , Biopsia , Caspasa 3/genética , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Inmunohistoquímica , Leucomalacia Periventricular/patología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Ratas
11.
Am J Pathol ; 188(3): 757-767, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29248460

RESUMEN

Brain injury in premature infants, especially periventricular leukomalacia, is an important cause of neurologic disabilities. Inflammation contributes to perinatal brain injury development, but the essential mediators that lead to early-life brain injury remain largely unknown. Neonates have reduced capacity for mounting conventional αßT-cell responses. However, γδT cells are already functionally competent during early development and are important in early-life immunity. We investigated the potential contribution of γδT cells to preterm brain injury using postmortem brains from human preterm infants with periventricular leukomalacia and two animal models of preterm brain injury-the hypoxic-ischemic mouse model and a fetal sheep asphyxia model. Large numbers of γδT cells were observed in the brains of mice, sheep, and postmortem preterm infants after injury, and depletion of γδT cells provided protection in the mouse model. The common γδT-cell-associated cytokines interferon-γ and IL-17A were not detectable in the brain. Although there were increased mRNA levels of Il17f and Il22 in the mouse brains after injury, neither IL-17F nor IL-22 cytokines contributed to preterm brain injury. These findings highlight unique features of injury in the developing brain, where, unlike injury in the mature brain, γδT cells function as initiators of injury independently of common γδT-cell-associated cytokines. This finding will help to identify therapeutic targets for preventing or treating preterm infants with brain injury.


Asunto(s)
Encéfalo/patología , Hipoxia-Isquemia Encefálica/patología , Linfocitos Intraepiteliales/patología , Leucomalacia Periventricular/patología , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Hipoxia-Isquemia Encefálica/metabolismo , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Linfocitos Intraepiteliales/metabolismo , Leucomalacia Periventricular/metabolismo , Masculino , Ratones , Ovinos
12.
Neurosci Lett ; 650: 12-17, 2017 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-28359933

RESUMEN

Recombinant human erythropoietin (rh-EPO) has been reported to have protective effects against brain injury. The purpose of this study was to evaluate the levels of erythropoietin receptor (EPOR) and neuroglobin (Ngb) in a neonatal rat model of periventricular white matter damage (PWMD), and to identify the relationship between the two proteins. On postnatal day 3 (P3), rats underwent permanent ligation of the right common carotid artery followed by 6% O2 for 4h (HI) or sham operation and normoxic exposure (sham). Immediately after HI, rats received a single intraperitoneal injection of rh-EPO (5U/g) or saline. We assessed the expression level of Ngb and EPOR on postnatal days 5, 7, 10 and 14. EPOR in the HI rats was initially increased as compared to the sham rats at P5. Subsequently, EPOR expression decreased, but was maintained at a higher level than in sham rats from P7 to P14. In rh-EPO treated rats, the increase in EPOR was greater than in HI rats at P5. However, EPOR levels decreased sharply from P7 to P14. In HI rats, Ngb was increased compared to the sham rats from P5 to P14. Ngb levels were further upregulated after rh-EPO administration from P5 to P10 compared to HI rats. However, this upregulation decreased at P14. In conclusion, this study shows that EPOR and Ngb were upregulated, and both of them act as important coordinated neuroprotectors in rh-EPO treatment of PWMD. However, the two proteins exhibit different expression patterns.


Asunto(s)
Eritropoyetina/administración & dosificación , Globinas/metabolismo , Leucomalacia Periventricular/tratamiento farmacológico , Leucomalacia Periventricular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Receptores de Eritropoyetina/metabolismo , Animales , Animales Recién Nacidos , Relación Dosis-Respuesta a Droga , Eritropoyetina/genética , Femenino , Leucomalacia Periventricular/patología , Masculino , Neuroglobina , Neuroprotección/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes , Resultado del Tratamiento
13.
PLoS One ; 12(4): e0175438, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28406931

RESUMEN

Periventricular leukomalacia (PVL), a brain injury affecting premature infants is commonly associated with cerebral palsy. PVL results from hypoxia-ischemia (HI) with or without infection and is characterized by white matter necrotic lesions, hypomyelination, microglial activation, astrogliosis, and neuronal death. It is important to study a PVL mouse model that mimics human PVL in symptomatology, anatomic and molecular basis. In our neonate mice model, bilateral carotid arteries were temporary ligated at P5 followed by hypoxic exposure (FiO2 of 8% for 20 min.). At P5 in mice, the white matter is more vulnerable to HI injury than the grey matter. In our PVL model, mice suffer from significant hind limb paresis, incoordination and feeding difficulties. Histologically they present with ventriculomegally, white matter loss, microglial activation and neuronal apoptosis. HI injury increases proinflammtory cytokines, activates NF-kB, activates microglia and causes nitrative stress. All these inflammatory mediators lead to oligodendroglial injury and white matter loss. Neurobehavioral analysis in the PVL mice model at P60 showed that the HI group had a significant decrease in hind limb strength, worsening rotarod testing and worsening performance in the open field test. This new PVL model has great advantages far beyond just mimicking human PVL in clinical features and histopathology. Long term survival, the development of cerebral palsy and the ability of using this model in transgenic animals will increase our understanding of the mechanistic pathways underlying PVL and defining specific targets for the development of suitable therapeutics.


Asunto(s)
Conducta Animal , Parálisis Cerebral , Hipoxia , Mediadores de Inflamación/metabolismo , Leucomalacia Periventricular , Paresia , Animales , Animales Recién Nacidos , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Parálisis Cerebral/metabolismo , Parálisis Cerebral/patología , Parálisis Cerebral/fisiopatología , Modelos Animales de Enfermedad , Miembro Posterior/metabolismo , Miembro Posterior/patología , Miembro Posterior/fisiopatología , Humanos , Hipoxia/metabolismo , Hipoxia/patología , Hipoxia/fisiopatología , Leucomalacia Periventricular/metabolismo , Leucomalacia Periventricular/patología , Leucomalacia Periventricular/fisiopatología , Ratones , Paresia/patología , Paresia/fisiopatología
15.
BMC Neurosci ; 17: 2, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26733225

RESUMEN

BACKGROUND: Periventricular leukomalacia (PVL) is the leading cause of neurological disabilities including motor and cognitive deficits in premature infants. Periventricular leukomalacia is characterized by damage to the white matter in the immature brain, but the mechanisms by which damage to immature white matter results in widespread deficits of cognitive and motor function are unclear. The thalamocortical system is crucial for human consciousness and cognitive functions, and impaired development of the cortico-thalamic projections in the neonatal period is implicated to contribute importantly to abnormalities of cognitive function in children with PVL. RESULTS: In this study, using a mouse model of PVL, we sought to test the hypothesis that PVL-like injury affects the different components of the thalamocortical circuitry that can be defined by vesicular glutamate transporters 1 and 2 (vGluT1 and vGluT2), both of which are required for glutamatergic synaptic transmission in the central nervous system. We combined immunocytochemistry and immuno-electron microscopy to investigate changes in cortico-thalamic synapses which were specifically identified by vGluT1 immunolabeling. We found that a drastic reduction in the density of vGluT1 labeled profiles in the somatosensory thalamus, with a reduction of 72-74 % in ventroposterior (VP) nucleus and a reduction of 42-82 % in thalamic reticular nucleus (RTN) in the ipsilateral side of PVL mice. We further examined these terminals at the electron microscopic level and revealed onefold-twofold decrease in the sizes of vGluT1 labeled corticothalamic terminals in VP and RTN. The present study provides anatomical and ultrastructural evidence to elucidate the cellular mechanisms underlying alteration of thalamic circuitry in a mouse model of PVL, and reveals that PVL-like injury has a direct impact on the corticothalamic projection system. CONCLUSIONS: Our findings provide the first set of evidence showing that the thalamocortical circuitry is affected and vulnerable in PVL mice, supporting a working model in which vGluT1 defined corticothalamic synapses are altered in PVL mice, and vGluT2 defined thalamocortical synapses are associated with such changes, leading to the compromised thalamocortical circuitry in the PVL mice. Our study demonstrates that the thalamocortical circuitry is highly vulnerable to hypoxia-ischemia in the PVL model, thus identifying a novel target site in PVL pathology.


Asunto(s)
Corteza Cerebral/ultraestructura , Modelos Animales de Enfermedad , Hipoxia-Isquemia Encefálica/complicaciones , Leucomalacia Periventricular/patología , Sinapsis/ultraestructura , Tálamo/ultraestructura , Animales , Corteza Cerebral/metabolismo , Leucomalacia Periventricular/etiología , Leucomalacia Periventricular/metabolismo , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/metabolismo , Vías Nerviosas/ultraestructura , Sinapsis/metabolismo , Tálamo/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo
16.
Adv Neonatal Care ; 15(2): 94-103; quiz E1-2, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25822515

RESUMEN

BACKGROUND: Neonatal resuscitation continues to be challenged by evolving research on the best approach to resuscitating preterm infants while minimizing potential health risks. The actions of the resuscitation team in the first minutes of transition to extrauterine life can have a lasting impact on the growth and development of the preterm infant. PURPOSE: This article reviews the most current literature on the use of oxygen in the delivery room and discusses the implications related to nursing and the multidisciplinary care team. FINDINGS: Oxygen saturation monitoring in the delivery room through the use of pulse oximetry in conjunction with oxygen titration via a blended oxygen source is an appropriate intervention to decrease the risk of free radical damage to the tissues. IMPLICATIONS FOR PRACTICE: Ensure delivery room providers are educated to resuscitation standards and ensure delivery rooms are appropriately supplied with a compressed air source, oxygen blenders, and pulse oximeters to minimize the free radical damage to the tissues. IMPLICATIONS FOR RESEARCH: Future studies should be focused on pulse oximetry use in the delivery room and its effect on long-term outcomes for preterm infants, safe oxygen saturation target ranges for the preterm infant in the delivery room, and effective resuscitation procedures for extremely preterm infants.


Asunto(s)
Hemorragia Cerebral/prevención & control , Leucomalacia Periventricular/prevención & control , Enfermedades Pulmonares/prevención & control , Estrés Oxidativo , Terapia por Inhalación de Oxígeno/métodos , Especies Reactivas de Oxígeno/metabolismo , Retinopatía de la Prematuridad/prevención & control , Hemorragia Cerebral/metabolismo , Salas de Parto , Humanos , Recién Nacido , Recien Nacido Prematuro , Leucomalacia Periventricular/metabolismo , Enfermedades Pulmonares/metabolismo , Enfermería Neonatal , Oximetría , Terapia por Inhalación de Oxígeno/efectos adversos , Retinopatía de la Prematuridad/metabolismo
17.
Invest Ophthalmol Vis Sci ; 56(3): 1830-41, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25698709

RESUMEN

PURPOSE: To characterize concurrent retinal vessel pathologies reminiscent to retinopathy of prematurity (ROP) in a rat model of periventricular leukomalacia (PVL), in order to identify uniform damage pathways in both organs, the eye and the brain. METHODS: Ischemia was induced in Long Evans rat pups on postnatal day 6 (P6) with unilateral (left side) carotid ligation (UCL) followed by exposure to different oxygen concentrations. Four different groups were studied: group A, hypoxia/ischemia (UCL + 6% O2, 1 hour); group B, hyperoxia (80% O2, 24 hours); group C, hypoxia/ischemia + hyperoxia (UCL + 6% O2, 1 hour + 80% O2, 24 hours); and group D, normoxia. In groups A and C, both retinae were examined separately (left retina, group A [A-L], right retina, group A [A-R]; left retina, group C [C-L], right retina, group C [C-R]). Morphologic analysis of vessel development based on flatmounts and cryosections was performed at P11 and P21. Quantitative (q)PCR was performed at P7, P11, and P21 (VEGF-A164, HIF-1α, EpoR, TNFα, iNOS, BMP-9, and IGF-1). RESULTS: On flatmounts, distinct retardation in deeper vascular plexus development was observed, most prominent in A-L and C-L. Retinae of groups A-L and C-L displayed reduced capillary-free zones and an increased number of branching points at P11. Quantitative PCR analysis showed significantly different expression profiles of IGF-1 in A-L and B compared with D over the time course of the experiment. CONCLUSIONS: This is the first report on concurring damage to the retina that was evaluated in a rat model of white matter injury in the developing brain. The relatively mild damage to the retinal vessel system may represent the basis for a model of moderate forms of ROP and to study vascular remodeling.


Asunto(s)
Animales Recién Nacidos , Leucomalacia Periventricular/patología , Vasos Retinianos/patología , Retinopatía de la Prematuridad/patología , Animales , Modelos Animales de Enfermedad , Proteínas del Ojo/biosíntesis , Proteínas del Ojo/genética , Regulación de la Expresión Génica , Inmunohistoquímica , Leucomalacia Periventricular/genética , Leucomalacia Periventricular/metabolismo , Microscopía Fluorescente , ARN/genética , Ratas , Ratas Long-Evans , Retinopatía de la Prematuridad/genética , Retinopatía de la Prematuridad/metabolismo
18.
J Perinat Med ; 43(3): 367-71, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25222588

RESUMEN

BACKGROUND: Periventricular white matter damage (PWMD), also termed periventricular leukomalacia, is the predominant neurologic lesion in preterm infants. It appears to relate in part to the development of the vascular supply to the cerebral white matter. We investigated whether, in case of severe hypoxia-ischemia, the vascular system would be subject to severe damage or remodeled. AIMS: To evaluate microvessel density (MVD) and the use of ephrinB2 and its receptor EphB4 to mark arterioles and venules to establish the correct anatomic assignment of the remodeled vessels in a hypoxia-induced PWMD rat model. METHODS: Postnatal day 3 rats underwent permanent ligation of the right common carotid artery followed by 6% O2 for 4 h (hypoxia-ischemia) or sham operation and normoxic exposure (sham). MVD and levels of ephrinB2 and EphB4, which are respectively regarded as relatively specific molecular markers of arteries and veins, were determined at postnatal day 7. RESULTS: Compared with sham rats, MVD, ephrinB2 and EphB4 levels were higher in the brains of hypoxic-ischemic rats. Similar percentages of vessels expressed ephrinB2 and EphB4 in sham rats, but expression of ephrinB2 was greater in brains injured by hypoxia-ischemia. CONCLUSIONS: Following hypoxic-ischemic injury to the rat brain, microvessels were remodeled and more arterioles than venules were acquired.


Asunto(s)
Arteriolas/patología , Efrina-B2/metabolismo , Leucomalacia Periventricular/patología , Microvasos/patología , Receptor EphB4/metabolismo , Vénulas/patología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Leucomalacia Periventricular/metabolismo , Embarazo , Ratas Sprague-Dawley
19.
Mol Med Rep ; 11(4): 2379-86, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25435330

RESUMEN

OLIG1 is an oligodendrocyte (OL) transcription factor, which can contribute to the proliferation and differentiation of OLs, and the maturation of myelin. The aim of this study was to clarify the role of OLIG1 in neonatal Sprague Dawley rats with periventricular leukomalacia (PVL), induced by hypoxia­ischemia (HI). Newborn rats in the HI group were subjected to ligation of the right carotid artery, followed by 8% oxygen delivery for 2 h, while rats in the normoxia group were only subjected to isolation of the right carotid artery, without exposure to hypoxia. Samples of brain tissue from rats in both groups were collected at 1, 3, 7, 14 and 21 days. In the HI group, observation by transmission electron microscopy (TEM) revealed OLs with a damaged nuclear membrane, cellular atrophy, deformation and necrosis, and cells in myelin with a high number of small vacuoles. A double­label immunofluorescence assay revealed the translocation of OLIG1 from the cytoplasm to the nucleus, while western blot and reverse transcription­quantitative polymerase chain reaction assays showed that there is a significant decrease, followed by an increase, in the gene and protein expression levels of OLIG1 and myelin basic protein (MBP). Despite the increase at the late stages of HI, the final levels of these proteins remained lower than the corresponding levels in the normoxia group. In conclusion, the decreased protein expression of OLIG1 following HI plays an important role in inhibiting the development and maturation of OLs and myelin. Although OLIG1 may, via its nuclear translocation, promote the growth and development of myelin to a certain extent, this factor fails to fully repair injured myelin.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hipoxia-Isquemia Encefálica/complicaciones , Leucomalacia Periventricular/etiología , Leucomalacia Periventricular/metabolismo , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/metabolismo , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/ultraestructura , Femenino , Expresión Génica , Inmunohistoquímica , Leucomalacia Periventricular/patología , Masculino , Proteína Básica de Mielina/metabolismo , Proteínas del Tejido Nervioso/genética , Oligodendroglía/ultraestructura , ARN Mensajero , Ratas
20.
Cereb Cortex ; 25(2): 482-95, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24046081

RESUMEN

The pathophysiology of perinatal brain injury is multifactorial and involves hypoxia-ischemia (HI) and inflammation. N-methyl-d-aspartate receptors (NMDAR) are present on neurons and glia in immature rodents, and NMDAR antagonists are protective in HI models. To enhance clinical translation of rodent data, we examined protein expression of 6 NMDAR subunits in postmortem human brains without injury from 20 postconceptional weeks through adulthood and in cases of periventricular leukomalacia (PVL). We hypothesized that the developing brain is intrinsically vulnerable to excitotoxicity via maturation-specific NMDAR levels and subunit composition. In normal white matter, NR1 and NR2B levels were highest in the preterm period compared with adult. In gray matter, NR2A and NR3A expression were highest near term. NR2A was significantly elevated in PVL white matter, with reduced NR1 and NR3A in gray matter compared with uninjured controls. These data suggest increased NMDAR-mediated vulnerability during early brain development due to an overall upregulation of individual receptors subunits, in particular, the presence of highly calcium permeable NR2B-containing and magnesium-insensitive NR3A NMDARs. These data improve understanding of molecular diversity and heterogeneity of NMDAR subunit expression in human brain development and supports an intrinsic prenatal vulnerability to glutamate-mediated injury; validating NMDAR subunit-specific targeted therapies for PVL.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Sustancia Gris/crecimiento & desarrollo , Receptores de N-Metil-D-Aspartato/metabolismo , Sustancia Blanca/crecimiento & desarrollo , Adulto , Encéfalo/embriología , Encéfalo/metabolismo , Niño , Preescolar , Femenino , Sustancia Gris/embriología , Sustancia Gris/metabolismo , Humanos , Lactante , Recién Nacido , Leucomalacia Periventricular/metabolismo , Masculino , Persona de Mediana Edad , Sustancia Blanca/embriología , Sustancia Blanca/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...