Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 181: 106114, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37023830

ABSTRACT

The neurovascular unit (NVU) plays an essential role in regulating neurovascular coupling, which refers to the communication between neurons, glia, and vascular cells to control the supply of oxygen and nutrients in response to neural activity. Cellular elements of the NVU coordinate to establish an anatomical barrier to separate the central nervous system from the milieu of the periphery system, restricting the free movement of substances from the blood to the brain parenchyma and maintaining central nervous system homeostasis. In Alzheimer's disease, amyloid-ß deposition impairs the normal functions of NVU cellular elements, thus accelerating the disease progression. Here, we aim to describe the current knowledge of the NVU cellular elements, including endothelial cells, pericytes, astrocytes, and microglia, in regulating the blood-brain barrier integrity and functions in physiology as well as alterations encountered in Alzheimer's disease. Furthermore, the NVU functions as a whole, therefore specific labeling and targeting NVU components in vivo enable us to understand the mechanism mediating cellular communication. We review approaches including commonly used fluorescent dyes, genetic mouse models, and adeno-associated virus vectors for imaging and targeting NVU cellular elements in vivo.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/genetics , Endothelial Cells , Blood-Brain Barrier/physiology , Brain/diagnostic imaging , Brain/blood supply , Astrocytes/physiology
2.
Zhongguo Dang Dai Er Ke Za Zhi ; 25(4): 431-435, 2023 Apr 15.
Article in Chinese | MEDLINE | ID: mdl-37073851

ABSTRACT

The male neonate in this case study was admitted to the hospital at 15 hours of age due to respiratory distress for 15 hours and poor response for 3 hours after resuscitation from asphyxia. The neonate was highly unresponsive, with central respiratory failure and seizures. Serum ammonia was elevated (>1 000 µmol/L). Blood tandem mass spectrometry revealed a significant decrease in citrulline. Rapid familial whole genome sequencing revealed OTC gene mutations inherited from the mother. Continuous hemodialysis filtration and other treatments were given. Neurological assessment was performed by cranial magnetic resonance imaging and electroencephalogram. The neonate was diagnosed with ornithine transcarbamylase deficiency combined with brain injury. He died at 6 days of age after withdrawing care. This article focuses on the differential diagnosis of neonatal hyperammonemia and introduces the multidisciplinary management of inborn error of metabolism.


Subject(s)
Hyperammonemia , Ornithine Carbamoyltransferase Deficiency Disease , Humans , Infant, Newborn , Male , Citrulline , Electroencephalography , Ornithine Carbamoyltransferase Deficiency Disease/diagnosis , Ornithine Carbamoyltransferase Deficiency Disease/therapy , Seizures
3.
Front Cell Neurosci ; 16: 976002, 2022.
Article in English | MEDLINE | ID: mdl-36204450

ABSTRACT

Hypoxic-ischemic (HI) induced perinatal white matter injury (PWMI) is a major cause of neurologic disabilities characterized by selective oligodendroglial death and myelin disruption. Galectin-3 (Gal-3) modulates postnatal subventricular zone gliogenesis and attenuates ischemic injury. However, the association between Gal-3 and myelin formation still remains unclear. In this study, we first perform Gal-3 knockdown (KD) to identify the importance of Gal-3 on myelin formation. Our results show impeded myelin formation, manifested by Olig2/CC1 (+) mature oligodendrocytes number, expression of oligodendroglial maturation-associated markers (MBP and CNPase), and myelin thickness and integrity. Then we perform recombinant Gal-3 (rGal-3) administration by intracerebroventricular injection. Notably, although rGal-3 administration shows no beneficial effect on oligodendrogenesis and myelin formation under normal condition, our results show that rGal-3 administration attenuates cognitive deficits and drives remyelination after PWMI, which are coupled to signs of enhanced myelin resiliency and cognition. Also, our results indicates that the significant increases in substrates for remyelination of rGal-3 administration are accompanied by enhanced Iba-1 (microglia marker)/ Mrc1 (M2 marker) (+) microglia and decreased Iba-1/ iNOS (M1 marker) (+) microglia. Altogether, our data in this research confirm the association between Gal-3 and myelin formation, underscore its position for the capacity for remyelination and restoration of function, and unveils the efficacy of rGal-3 administration with anti-inflammatory phenotype microglia (M2 microglia) activation. Thus, the findings suggest that Gal-3 plays a significant role in myelin formation and remyelination restoration.

4.
Transl Pediatr ; 11(7): 1074-1084, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35957997

ABSTRACT

Background: The increasing incidence of preterm birth has led to a global problem of adverse neurodevelopmental outcomes in preterm neonates as a result of brain injury. There is still a lack of models mimicking diffuse white matter injury (WMI) in preterm neonates that can be applied to transgenic mice. Methods: The right common carotid artery of the neonatal mouse was ligated on postnatal day 3 (P3) C57BL/6 mice and followed by 80, 90, or 100 min of hypoxia using a mixture of 10%±0.2% oxygen-nitrogen. The most suitable model was chosen by characterizing the effects of this hypoxic-ischemic insult on development of myelin, glial cell conditions, and neurological outcomes by hematoxylin-eosin (HE) staining performed at postnatal day 17 (P17), western blot measuring myelin basic protein (MBP) at postnatal day 10 (P10) and P17, immunofluorescence staining of MBP-neurofilament protein heavy chain (NFH), oligodendrocyte transcription factor-2 (Olig2)-adenomatous polyposis coli clone (CC1), glial fibrillary acidic protein (GFAP) and ionic calcium linker protein (Iba-1) at P17, electron microscopy observing myelin microstructure at postnatal day 52 (P52) and behavioral testing at postnatal day 45-50 (P45-P50). Results: The 90-min group showed neuroanatomical changes in the ipsilateral side of the brain, the 80-min group showed minor changes, and the 100-min group showed severe injury. Mice in the 90-min group subsequently showed marked activation of astrocytes, augmentation of microglia, a notable decrease in expression of MBP with a normal level of NFH, long-term cognitive dysfunction, and impairment of the myelin ultrastructure in adulthood. Conclusions: In conclusion, a mouse model of preterm diffuse WMI rather than cystic periventricular leukomalacia was successfully achieved by ligating one of the common carotid arteries on P3 followed by 90 min of hypoxia in a mixture of 10%±0.2% oxygen-nitrogen. The attempt provides an adequate translational animal model for elucidating the underlying mechanism.

5.
Transl Pediatr ; 11(1): 20-32, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35242649

ABSTRACT

BACKGROUND: The underlying cause of neurological sequelae after immature cerebral hypoxia-ischaemia (HI) white matter injury is impaired myelination. Previous studies have indicated that astrocyte activation is closely related to impaired myelination. However, the mechanism of reactive gliosis in white matter injury post-HI remains poorly understood. METHODS: Studies using adult ischaemic animal models demonstrated that hypoxia inducible factor-1α (HIF-1α) expression was involved in the formation of reactive astrocytes. Here, we investigated the temporal expression of HIF-1α and its impact on reactive gliosis and further myelination using a perinatal HI white matter injury model induced in rats at postnatal day 3. The temporal pattern of HIF-1α expression post-HI injury was tested by western blotting and immunofluorescence. Rats were treated with a HIF-1α inhibitor at 72 hours post-HI injury. Reactive gliosis and myelination were assessed with western blotting, immunofluorescence and electron microscopy, and neurological functions were examined by behavioural testing. RESULTS: Our results showed that the expression of HIF-1α was upregulated in neurons at 24 hours and in astrocytes at 7 days post-HI. Inhibiting delayed HIF-1α expression post-HI injury could restrain reactive gliosis, ameliorate hypomyelination, and improve the performance of rats in the Morris water maze test. CONCLUSIONS: Our findings suggest that a delayed increase in HIF-1α in astrocytes is involved in glial scar formation and leads to arrested oligodendrocyte maturation, impaired myelination, and long-term neurological function after experimental white matter injury in immature rats.

6.
Stem Cells Dev ; 31(3-4): 53-66, 2022 02.
Article in English | MEDLINE | ID: mdl-34913742

ABSTRACT

Bronchopulmonary dysplasia (BPD) remains the most important respiratory morbidity of preterm infants with few effective preventive strategies. Administration of mesenchymal stem cells (MSC) was considered effective to prevent BPD via paracrine extracellular vesicles (EVs), while appropriate regimens of MSC-EVs and the mechanism remain unclear. Therefore, we established a hyperoxia-induced rat BPD model, and examined the effect of early intraperitoneal MSC-EVs with different doses on BPD. We found that MSC-EVs ameliorated hyperoxia-induced lung injury in a dose-dependent manner, and high-dose MSC-EVs ameliorated alveolar simplification and fibrosis. Also, MSC-EVs showed its beneficial effects on vascular growth and pulmonary hypertension. Primary AT2 cells were observed to transdifferentiate into AT1 cells when exposure to hyperoxia in vitro. Administration of MSC-EVs at the first-day culture significantly delayed the transdifferentiation of AT2 cells induced by hyperoxia. We further found that exposure to hyperoxia led to elevated expression of WNT5a mRNA and protein, a key agent in AT2 transdifferentiation, while MSC-EVs administration decreased it. Further study is warranted that MSC-EVs may delay the transdifferentiation of AT2 cells via WNT5a. These studies provide key preclinical evidence of MSC-EVs therapeutics on BPD and highlight the effect of MSC-EVs on suppressing the transdifferentiation of AT2 cells and its possible mechanism through downregulation of WNT5a.


Subject(s)
Bronchopulmonary Dysplasia , Extracellular Vesicles , Hyperoxia , Mesenchymal Stem Cells , Alveolar Epithelial Cells , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/metabolism , Cell Transdifferentiation , Disease Models, Animal , Extracellular Vesicles/metabolism , Humans , Infant, Newborn , Infant, Premature , Rats
7.
Cell Biochem Biophys ; 79(2): 311-320, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33683657

ABSTRACT

The proliferation of fetal alveolar type II cells (FATIICs) was impaired in bronchopulmonary dysplasia (BPD), which is modulated by hyperoxia and inflammatory response. Interleukin 24 (IL-24), a cytokine produced by certain cell types, plays an essential role in inflammation and host protection against infection. However, the ability of FATIICs to produce IL-24 remains unclear, and the role of IL-24 in BPD progression is yet to be determined. With reverse transcription quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay, the authors evaluated whether FATIICs produce IL-24 in physiological conditions. The authors quantified IL-24 expression in the lungs of newborn rat pups exposed to hyperoxia (70% oxygen) and in FATIICs isolated on embryonic day 19 that were exposed to 95% oxygen or lipopolysaccharide (LPS). The role of IL-24 in FATIICs, cell proliferation, cell apoptosis, and cell cycle were further evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and flow cytometric analysis. Also, they assessed caspase-3 and SOCS3 mRNA in IL-24 siRNA-treated cells by using RT-qPCR. During culture, IL-24 mRNA and protein levels in FATIICs gradually decreased with FATIIC differentiation. IL-24 expression increased significantly in rat lungs exposed to hyperoxia and FATIICs exposed to oxygen or LPS. Recombinant IL-24 enhanced cell proliferation by decreasing the proportion of apoptotic cells and increasing the proportion of cells in the S phase. The IL-24 siRNA-treated cells expressed more caspase-3 mRNA. Furthermore, suppressor of cytokine signaling 3 (SOCS3) mRNA was significantly decreased in rats and FATIICs exposed to oxygen, whereas it dramatically increased in FATIICs exposed to LPS. The IL-24 siRNA-treated cells expressed more SOCS3 mRNA. These studies suggest IL-24 is a pulmonary target cytokine in BPD, and may possibly regulate SOCS3 in oxidative stress and inflammation of the lung.


Subject(s)
Bronchopulmonary Dysplasia/pathology , Interleukins/metabolism , Animals , Bronchopulmonary Dysplasia/metabolism , Cell Proliferation/drug effects , Down-Regulation/drug effects , Female , Hyperoxia , Interleukins/antagonists & inhibitors , Interleukins/genetics , Lipopolysaccharides/pharmacology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , S Phase Cell Cycle Checkpoints/drug effects , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Up-Regulation/drug effects
8.
Front Pediatr ; 8: 452, 2020.
Article in English | MEDLINE | ID: mdl-32850559

ABSTRACT

Background: Transdifferentiation of type II alveolar epithelial cells (AECII) into type I alveolar epithelial cells (AECI) is involved in neonatal respiratory distress syndrome (NRDS). Different ligands of the Notch pathway could have different effects on AECII transdifferentiation. Objective: To investigate the effects of Dlk1 and Jagged1 on the proliferation and transdifferentiation of AECII. Methods: Fetal AECIIs (19 days of gestation) were divided: control group, Dlk1 group, rhNF-κB group. Proliferation was tested using the MTT assay. Expression of surfactant protein C (SP-C) and aquaporin 5 (AQP5) was examined by immunofluorescence. mRNA and protein levels of SP-C, AQP5, Nortch1, Dlk1, Jagged1, and Hes1 were examined by RT-PCR and western blot. Results: In response to Dlk1, cell number and proliferation were increased (P < 0.05), and mRNA and protein levels of SP-C, Dlk1, Notch1, and Hes1 were up-regulated, while AQP and Jagged1 were decreased. In response to rhNF-κB, the cell number and proliferation were reduced, and mRNA and protein levels of Jagged1 and Notch1 were up-regulated, while Dlk1, and SP-C were downregulated. In the Dlk1 group, SP-C, and AQP5 expression patterns suggested that the cells were still transdifferentiating by 96 h, while in the rhNF-κB group, most cells had transdifferentiated by 72 h and were close to apoptosis by 96 h. Conclusion: These results suggest that Dlk1 promoted proliferation of AECIIs and inhibited cell transdifferentiation, while Jagged1 treatment inhibited proliferation of AECIIs and promoted transdifferentiation to AECIs. These results provide some clue for the eventual management of NDRS.

9.
Neurosci Lett ; 711: 134439, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31425825

ABSTRACT

In this research, we intended to evaluate the expression pattern, distribution and sources of Galectin-3 (Gal-3) in perinatal hypoxic-ischemic brain injury rat model. Postnatal day 3 Sprague-Dawley rat pups were subjected to right carotid artery ligation followed by 2.5 h of hypoxia (6% oxygen). Expression and distribution of Gal-3 were evaluated by western blotting and immunofluorescence. Sources of Gal-3 were evaluated by double staining with neuronic, oligodendrocytic, astrocytic, microglial and endotheliocytic markers. Our results indicated Gal-3 significantly upregulated from 12 h and maintained an increasing tendency within 72 h post injury. Although the relative expression of Gal-3 decreased after 72 h, we detected significant differences until 14d. We found Gal-3 started to distribute in cortex and thalamus area and maintained an increasing tendency. Gal-3 could be detected in cortex, thalamus, corpus callosum and hippocampus area at 72 h post injury. After that, expression of Gal-3 in cortex and thalamus area downregulated, the expression in corpus callosum and hippocampus area vanished. We found astrocyte, microglia, neuron and endotheliocyte were sources of Gal-3 in cortex area; astrocyte, microglia and endotheliocyte were sources of Gal-3 in thalamus area; oligodendrocyte precursor cell and endotheliocyte were sources of Gal-3 in corpus callosum; neuron, microglia and endotheliocyte were sources of Gal-3 in hippocampus. In conclusion, we demonstrated spatiotemporal expression patterns of Galectin-3 post perinatal hypoxic-ischemic brain injury in this research.


Subject(s)
Galectin 3/metabolism , Hypoxia-Ischemia, Brain/metabolism , Animals , Animals, Newborn , Brain/metabolism , Disease Models, Animal , Rats , Rats, Sprague-Dawley
10.
Neuroreport ; 29(7): 517-523, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29271834

ABSTRACT

Chondroitin sulfate proteoglycans (CSPGs) are pluripotent components of the extracellular matrix in the brain. Although previous studies have examined the developmental change in certain CSPGs in the whole brain, no known systematic studies have been carried out on the temporal or spatial expression of CSPGs. Here, we used quantitative real-time PCR to examine the CSPG mRNAs expression in the postnatal developing rat brain starting from postnatal day 5-42, mainly focusing on the parietal cortex, hippocampus, and corpus callosum. Results were further verified by immunohistochemistry. Our results showed that aggrecan, brevican, phosphacan, and NG2 generally showed upregulation across developmental stages in all three regions. Neurocan showed a rapid increase until postnatal day 10 in all three regions. Versican, however, showed a sharp decrease until postnatal day 10. Cross-region analysis showed higher expression of most CSPG members in the corpus callosum during later stages of development. Further immunohistochemistry staining confirmed our results by showing prominent CSPGs protein expression in the corpus callosum. In summary, our study reported specific temporal-expression and spatial-expression patterns of the CSPGs species. These results are consistent with the known roles of these members in neurodevelopment. The current study provided clues for the development of CSPGs as potential treatment targets in neurodevelopmental disorders.


Subject(s)
Brain/growth & development , Brain/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Animals , Corpus Callosum/growth & development , Corpus Callosum/metabolism , Female , Hippocampus/growth & development , Hippocampus/metabolism , Parietal Lobe/growth & development , Parietal Lobe/metabolism , RNA, Messenger , Rats, Sprague-Dawley , Up-Regulation
11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(12): 1623-1626, 2016 Dec.
Article in Chinese | MEDLINE | ID: mdl-27916093

ABSTRACT

Objective To study the protective effect of interleukin-22 (IL-22) against the hyperoxia-induced lung injury in neonatal SD rats. Methods The neonatal SD rats were randomized into control group, hyperoxia group and IL-22 (10 ng/g) treatment group. Each group was randomly divided into three subgroups of 1, 3, 7 days (n=9). Body mass in every group was detected; lung pathological changes were observed by HE staining; tumor necrosis factor α (TNF-α) in lung tissues was tested by quantitative real-time PCR; and IL-1ß in plasma was measured by ELISA. Results After exposure to hyperoxia for 1 day, the body mass in the three groups showed no significant difference, and the structure of lung tissues were normal. After exposure to hyperoxia for 3 or 7 days, the body mass in the hyperoxia group was significantly lower, IL-1ß in plasma was significantly enhanced, and the structure of lung tissues were destroyed, which aggravated with the time of hyperoxia exposure. TNF-α mRNA level increased obviously in the hyperoxia group. However, when treated with IL-22, TNF-α mRNA was significantly down-regulated in the treatment group. Conclusion IL-22 may play an protective role in hyperoxia-induced lung injury.


Subject(s)
Hyperoxia/complications , Interleukins/pharmacology , Interleukins/therapeutic use , Lung Injury/drug therapy , Lung Injury/etiology , Animals , Animals, Newborn , Disease Models, Animal , Hyperoxia/metabolism , Interleukin-1beta/blood , RNA, Messenger , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Interleukin-22
12.
Apoptosis ; 21(11): 1315-1326, 2016 11.
Article in English | MEDLINE | ID: mdl-27658784

ABSTRACT

Angiotensin II (AngII) is an important factor that promotes the proliferation of cancer cells, whereas celastrol exhibits a significant antitumor activity in various cancer models. Whether celastrol can effectively suppress AngII mediated cell proliferation remains unknown. In this study, we studied the effect of celastrol on AngII-induced HepG2 cell proliferation and evaluated its underlying mechanism. The results revealed that AngII was able to significantly promote HepG2 cell proliferation via up-regulating AngII type 1 (AT1) receptor expression, improving mitochondrial respiratory function, enhancing nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity, increasing the levels of reactive oxygen species (ROS) and pro-inflammatory cytokines. The excess ROS from mitochondrial dysfunction is able to cause the apoptosis of tumor cells via activating caspase3 signal pathway. In addition, the reaction between NO and ROS results in the formation of peroxynitrite (ONOO-), and then promoting cell damage. celastrol dramatically enhanced ROS generation, thereby causing cell apoptosis through inhibiting mitochodrial respiratory function and boosting the expression levels of AngII type 2 (AT2) receptor without influencing NADPH oxidase activity. PD123319 as a special inhibitor of AT2R was able to effectively decreased the levels of inflammatory cytokines and endothelial nitric oxide synthase (eNOS) activity, but only partially attenuate the effect of celastrol on AnII mediated HepG2 cell proliferation. Thus, celastrol has the potential for use in liver cancer therapy. ROS derived from mitochondrial is an important factor for celastrol to suppress HepG2 cell proliferation.


Subject(s)
Angiotensin II/metabolism , Apoptosis/drug effects , Mitochondria/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Triterpenes/pharmacology , Angiotensin II/genetics , Cell Proliferation/drug effects , Hep G2 Cells , Humans , Mitochondria/drug effects , NADPH Oxidases/genetics , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Pentacyclic Triterpenes , Signal Transduction/drug effects
13.
In Vitro Cell Dev Biol Anim ; 51(5): 479-87, 2015 May.
Article in English | MEDLINE | ID: mdl-25515249

ABSTRACT

Retinoic acid (RA) plays an important role in lung development and maturation. Many stimuli can induce alveolar epithelial cell damage which will result in the injury of lung parenchyma. The aim of this study was to observe the effect of RA on the proliferation and differentiation of primary fetal alveolar epithelial type II cells (fAECIIs). Primary fAECIIs were isolated from fetal rats at 19 d of gestation and purified by a differential centrifugation and adhesion method. The cells were randomly divided into control (dimethyl sulfoxide, DMSO) and RA groups. Cell proliferation, viability, apoptosis, cycle, and expression of target protein were examined at 24, 48, and 72 h. We found that the proliferation and viability of cells in the RA-exposed group significantly increased compared with the DMSO control group. The proportion (%) of cells in the G2 and S phases in the RA group was significantly higher than that in control group cells. The proportion (%) of both early apoptotic cells and late apoptotic cells decreased significantly in cells exposed to RA compared with cells exposed to DMSO. RA significantly enhanced the expression of aquaporin 5 (AQP5). The expression level of pulmonary surfactant C (SPC) was elevated after cells were exposed to RA for 24 and 72 h but was inhibited when cells were exposed to RA for 48 h. These results suggest that RA promotes fAECII proliferation by improving cell viability, promoting S phase entry and inhibiting apoptosis and RA promotes fAECIIs differentiation to alveolar epithelial type I cells (AECIs).


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Fetus/cytology , Pulmonary Alveoli/cytology , Respiratory Mucosa/cytology , Tretinoin/pharmacology , Analysis of Variance , Animals , Apoptosis/drug effects , Aquaporin 5/metabolism , Blotting, Western , Cell Cycle/drug effects , Cell Differentiation/physiology , Cell Proliferation/physiology , Cell Survival/drug effects , Dimethyl Sulfoxide , Epithelial Cells/cytology , Epithelial Cells/physiology , Molecular Structure , Rats , Rats, Sprague-Dawley , Time Factors , Tretinoin/chemistry , Tretinoin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...