Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters











Publication year range
1.
Neuroscience ; 551: 103-118, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38810691

ABSTRACT

Monosialoganglioside GM1 (GM1) has long been used as a therapeutic agent for neurological diseases in the clinical treatment of ischemic stroke. However, the mechanism underlying the neuroprotective function of GM1 is still obscure until now. In this study, we investigated the effects of GM1 in ischemia and reperfusion (I/R) brain injury models. Middle cerebral artery occlusion and reperfusion (MCAO/R) rats were treated with GM1 (60 mg·kg-1·d-1, tail vein injection) for 2 weeks. The results showed that GM1 substantially attenuated the MCAO/R-induced neurological dysfunction and inhibited the inflammatory responses and cell apoptosis in ischemic parietal cortex. We further revealed that GM1 inhibited the activation of NFκB/MAPK signaling pathway induced by MCAO/R injury. To explore its underlying mechanism of the neuroprotective effect, transcriptome sequencing was introduced to screen the differentially expressed genes (DEGs). By function enrichment and PPI network analyses, Sptbn1 was identified as a node gene in the network regulated by GM1 treatment. In the MCAO/R model of rats and oxygen-glucose deprivation and reperfusion (OGD/R) model of primary culture of rat cortical neurons, we first found that SPTBN1 was involved in the attenuation of I/R induced neuronal injury after GM1 administration. In SPTBN1-knockdown SH-SY5Y cells, the treatment with GM1 (20 µM) significantly increased SPTBN1 level. Moreover, OGD/R decreased SPTBN1 level in SPTBN1-overexpressed SH-SY5Y cells. These results indicated that GM1 might achieve its potent neuroprotective effects by regulating inflammatory response, cell apoptosis, and cytomembrane and cytoskeleton signals through SPTBN1. Therefore, SPTBN1 may be a potential target for the treatment of ischemic stroke.


Subject(s)
G(M1) Ganglioside , Neurons , Neuroprotective Agents , Rats, Sprague-Dawley , Reperfusion Injury , Signal Transduction , Animals , Reperfusion Injury/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , G(M1) Ganglioside/pharmacology , Male , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Brain Ischemia/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Rats , Apoptosis/drug effects , Apoptosis/physiology , Spectrin/metabolism
2.
Sheng Wu Gong Cheng Xue Bao ; 40(2): 321-336, 2024 Feb 25.
Article in Chinese | MEDLINE | ID: mdl-38369824

ABSTRACT

Triple-negative breast cancer (TNBC) is the most malignant subtype of breast cancer. Currently, chemotherapy remains to be the primary treatment for TNBC, but drug resistance is common while patient prognosis is poor. With the development of proteomics technology, phosphoproteomics research has made great progress and has been widely used in the study of tumor mechanism, diagnosis and treatment. Similarly, phosphoproteomics plays a significant role in the studies of the occurrence, development, targeted therapy, and drug resistance mechanisms of TNBC. This article summarizes the research progress of phosphoproteomics in TNBC, with the aim to facilitate the research on the mechanism and treatment of TNBC based on phosphoproteomics.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Prognosis , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/drug therapy , Female
3.
Open Biol ; 12(4): 210315, 2022 04.
Article in English | MEDLINE | ID: mdl-35382569

ABSTRACT

A precise guiding signal is crucial to orchestrate directional migration and patterning of the complex vascular network and neural system. So far, limited studies have reported the discovery and functions of microRNAs (miRNAs) in guiding vascular and neural pathfinding. Currently, we showed that the deficiency of miRNA-22a, an endothelial-enriched miRNA, caused dramatic pathfinding defects both in intersegmental vessels (ISVs) and primary motor neurons (PMNs) in zebrafish embryos. Furthermore, we found the specific inhibition of miR-22a in endothelial cells (ECs) resulted in patterning defects of both ISVs and PMNs. Neuronal block of miR-22a mainly led to axonal defects of PMN. Sema4c was identified as a potential target of miR-22a through transcriptomic analysis and in silico analysis. Additionally, a luciferase assay and EGFP sensor assay confirmed the binding of miR-22a with 3'-UTR of sema4c. In addition, downregulation of sema4c in the miR-22a morphants significantly neutralized the aberrant patterning of vascular and neural networks. Then we demonstrated that endothelial miR-22a regulates PMNs axonal navigation. Our study revealed that miR-22a acted as a dual regulatory cue coordinating vascular and neuronal patterning, and expanded the repertoire of regulatory molecules, which might be of use therapeutically to guide vessels and nerves in the relevant diseases.


Subject(s)
MicroRNAs , Semaphorins , Zebrafish Proteins , Zebrafish , Animals , Axon Guidance , Endothelial Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Motor Neurons , Semaphorins/genetics , Semaphorins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
Materials (Basel) ; 14(23)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34885359

ABSTRACT

In this study, TiN-TiB2-hBN composite ceramics were prepared via reactive hot pressing using TiN and amorphous B powders as raw materials. Different sintering temperatures and composition ratios were studied. The results show that the 70 vol% TiN-17.6 vol% TiB2-12.4 vol% hBN ceramic composites obtained ideal comprehensive properties at 1600 °C. The relative density, Vickers hardness, bending strength, and fracture toughness were 99%, 11 GPa, 521 MPa, and 4.22 MPa·m1/2, respectively. Densification was promoted by the highly active reaction product TiB2, and the structural defects formed in the grains. Meanwhile, the good interfacial bonding between TiN and TiB2 grains and the uniform dispersion of ultrafine hBN in the matrix contributed to the excellent bending strength. Moreover, the toughening mechanism of crack deflection and grain pull-out improved the fracture toughness.

6.
ACS Chem Neurosci ; 12(21): 3994-4006, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34637270

ABSTRACT

C5a receptor 1 (C5aR1) can induce a strong inflammatory response to an injury. Targeting C5aR1 has emerged as a novel anti-inflammatory therapeutic method. However, the role of C5aR1 in cerebral ischemia and reperfusion (I/R) injury and the definitive mechanism have not been elucidated clearly. Here, we determined whether C5aR1 signaling was essential to the post-ischemic inflammation and brain injury and whether it is a valid target for therapeutic blockade by using soluble receptor antagonist PMX53 in the early stage after I/R injury. In an in vitro model (oxygen and glucose deprivation and reperfusion, OGD/R) and in vivo model (middle cerebral artery occlusion and reperfusion, MCAO/R) of I/R, the neuronal cells of rats showed significantly up-regulated gene expression of C5aR1, and a notable inflammatory response was demonstrated with elevated tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and IL-6. Inhibition of C5aR1 by PMX53 treatment significantly reduced cell injury and inflammation and promoted brain function recovery. Further mechanism studies showed that inhibiting C5aR1 by PMX53 protected the rats from MCAO/R injury, decreased cell inflammation, and apoptosis via inhibiting the TLR4 and NF-κB signaling pathway and reducing the production of TNF-α, IL-1ß, and IL-6 in MCAO/R rats. In addition, manipulation of the C5aR1 gene expression in vitro displayed that the inflammatory cascade signals including TLR4, TNF-α, IL-1ß, and IL-6 were coincidently regulated with the regulation of C5aR1 expression levels. Thus, our results demonstrated a pathogenic role for C5aR1 in the progression of brain injury and inflammation response following I/R injury. Our study clearly demonstrated that C5aR1 inhibition might be an effective treatment strategy for ischemic stroke.


Subject(s)
Brain Ischemia , Reperfusion Injury , Animals , Brain/metabolism , Infarction, Middle Cerebral Artery , Inflammation , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Anaphylatoxin C5a , Reperfusion
7.
Front Neurosci ; 15: 555543, 2021.
Article in English | MEDLINE | ID: mdl-33633530

ABSTRACT

Inflammatory response contributes to brain injury after ischemia and reperfusion (I/R). Our previous literature has shown isoquercetin plays an important role in protecting against cerebral I/R injury. The present study was conducted to further investigate the effect of isoquercetin on inflammation-induced neuronal injury in I/R rats with the involvement of cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) and inhibitor of NF-κB (I-κB)/nuclear factor-kappa B (NF-κB) signaling pathway mediated by Toll-like receptor 4 (TLR4) and C5a receptor 1 (C5aR1). In vivo middle cerebral artery occlusion and reperfusion (MCAO/R) rat model and in vitro oxygen-glucose deprivation and reperfusion (OGD/R) neuron model were used. MCAO/R induced neurological deficits, cell apoptosis, and release of cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6 in ischemic brain in rats. Simultaneously, the expression of TLR4 and C5aR1 was significantly up-regulated in both MCAO/R rats and OGD/R neurons, accompanied with the inhibition of cAMP/PKA signaling and activation of I-κB/NF-κB signaling in the cortex of MCAO/R rats. Over-expression of C5aR1 in neurons induced decrease of cell viability, exerting similar effects with OGD/R injury. Isoquercetin acted as a neuroprotective agent against I/R brain injury to suppress inflammatory response and improve cell recovery by inhibiting TLR4 and C5aR1 expression, promoting cAMP/PKA activation, and inhibiting I-κB/NF-κB activation and Caspase 3 expression. TLR4 and C5aR1 contributed to inflammation and apoptosis via activating cAMP/PKA/I-κB/NF-κB signaling during cerebral I/R, suggesting that this signaling pathway may be a potent therapeutic target in ischemic stroke. Isoquercetin was identified as a neuroprotective agent, which maybe a promising therapeutic agent used for the treatment of ischemic stroke and related diseases.

8.
Int J Biol Macromol ; 165(Pt B): 1783-1792, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33045296

ABSTRACT

The widespread use of nanomaterials poses a great threat to human living environments. Among them, biomass-derived cellulose nanoparticle (CN) is one of the widely used nanomaterials. To date, the toxicity of CNs during embryonic development remains undetermined. In this study, we exposed zebrafish embryos to cellulose nanofibrils (CNFs) and cellulose nanocrystals (CNCs) to evaluate the toxicity of these CNs. Exposure to CNFs or CNCs below 30 mg/ml exhibited no dose-dependent increases in malformation and mortality in zebrafish embryos. Then we demonstrated that CNs were highly enriched in zebrafish embryo via imaging analyses of embryos treated with FITC-coupled CNCs. In addition, we found that CNF or CNC exposure resulted in compromised motor ability of zebrafish larva. Furthermore, it was revealed that the differentiation and the morphogenesis of motor neurons were significantly interrupted. While, blood vessels were normally patterned, suggesting the specific neurotoxicity of these nanomaterials. Transcriptome sequencing assay showed that the neurotoxicity of CNs in the motor neurons might be attributed to the expression alteration of neural genes. In summary, we discovered the neurotoxicity of CNs for the first time.


Subject(s)
Biomass , Cellulose/toxicity , Nanoparticles/toxicity , Neurotoxins/toxicity , Toxicity Tests , Zebrafish/physiology , Animals , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/drug effects , Gene Ontology , Larva/drug effects , Motor Activity/drug effects , Motor Neurons/drug effects , Motor Neurons/pathology , Nanofibers/toxicity , Particle Size , RNA, Messenger/genetics , RNA, Messenger/metabolism , Zebrafish/embryology
9.
J Ophthalmol ; 2020: 5905269, 2020.
Article in English | MEDLINE | ID: mdl-32655941

ABSTRACT

Choroidal neovascularization (CNV) is a leading cause of blindness in the elderly in developed countries and is particularly associated with age-related macular degeneration (AMD). Cabozantinib (CBZ) hinders the activation of multiple receptor tyrosine kinases involved in tumor angiogenesis, such as hepatocyte growth factor receptor (MET) and vascular endothelial growth factor receptor 2 (VEGFR2). We aimed to investigate the role and mechanism of CBZ in a mouse laser-induced CNV model. In zebrafish embryos, CBZ perturbed intersegmental vessel (ISV) formation without obvious neurodevelopment impairment. In the mouse laser-induced CNV model, phosphorylated hepatocyte growth factor receptor (p-MET) and phosphorylated vascular endothelial growth factor receptor 2 (p-VEGFR2) were increased in the CNV region. CBZ intravitreal injection or oral gavage alleviated CNV leakage and the CNV lesion area without obvious intraocular toxicity, as well as disturbed the phosphorylation of MET and VEGFR2. Additionally, CBZ downregulated the expression of the hepatocyte growth factor (HGF) with no effect on the expression of the vascular endothelial growth factor (VEGF). CBZ downregulated HGF, p-MET, and p-VEGFR2 expressions in vitro, as well as inhibited the proliferation, migration, and tube formation of b-End3 cells. In summary, CBZ alleviates mouse CNV formation possibly via inhibiting the activation of MET and VEGFR2. The findings provide a novel potential therapy method for CNV patients.

10.
Neurol Res ; 42(8): 693-702, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32657248

ABSTRACT

It has been established that poor outcomes in ischemic stroke patients are associated with the post-reperfusion inflammatory response and up-regulation of TLR4. Therefore, suppression of the TLR4 signaling pathway constitutes a potential neuroprotective therapeutic strategy. Schisandrin B, a compound extracted from Schisandra chinensis, has been shown to possess anti-inflammatory and neuroprotective properties. However, the mechanism remains unclear. In the present study, the therapeutic effect of schisandrin B was assessed following cerebral ischemia and reperfusion (I/R) injury in a model of middle cerebral artery occlusion and reperfusion (MCAO/R) in rats. The effects of schisandrin B were investigated with particular emphasis on TLR4 signal transduction and on the inflammatory response. Schisandrin B treatment conferred significant protection against MCAO/R injury, as evidenced by decreases in infarct volume, neurological score, and the number of apoptotic neurons and inflammatory signaling molecules. ABBREVIATIONS: I/R: schemia/reperfusion; IL: interleukin; MCAO/R: middle cerebral artery occlusion and reperfusion; NF-κB: nuclear; TLR4: Toll-like receptor 4; TNF-α: tumor necrosis factor-α.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Brain Ischemia/metabolism , Lignans/administration & dosage , Polycyclic Compounds/administration & dosage , Reperfusion Injury/metabolism , Signal Transduction/drug effects , Animals , Brain Ischemia/complications , Brain Ischemia/prevention & control , Cyclooctanes/administration & dosage , Male , NF-kappa B/metabolism , Rats, Sprague-Dawley , Reperfusion Injury/complications , Reperfusion Injury/prevention & control , Toll-Like Receptor 4/metabolism
11.
J Cell Physiol ; 235(2): 1259-1273, 2020 02.
Article in English | MEDLINE | ID: mdl-31270802

ABSTRACT

In age-related macular degeneration (AMD), choroidal neovascularization (CNV), a major pathologic feature of neovascular AMD (nAMD), affects 10% of patients, potentially causing serious complications, including vision loss. Vascular endothelial growth factor receptor 2 (VEGFR2) and fibroblast growth factor receptor 1 (FGFR1) contribute to the pathogenesis of CNV. Brivanib is an oral selective dual receptor tyrosine kinase (RTK) inhibitor of FGFRs and VEGFRs, especially VEGFR2 and FGFR1. In this study, brivanib inhibited zebrafish embryonic angiogenesis without impairing neurodevelopment. In a mouse CNV model, brivanib intravitreal injection blocked phosphorylation of FGFR1 and VEGFR2 and reduced CNV leakage, area, and formation without causing intraocular toxicity. Moreover, brivanib oral gavage reduced CNV leakage and area. Accordingly, brivanib remained at high concentrations (above 14,000 ng/ml) in retinal/choroidal/scleral tissues following intravitreal injection. Similarly, brivanib remained at high concentrations (over 10,000 ng/ml) in retinal/choroidal/scleral tissues following oral gavage. Finally, in vitro cell experiments demonstrated that brivanib inhibited the proliferation, migration and tube formation of microvascular endothelial cells. In conclusion, our study suggested that brivanib treatment could be a novel therapeutic strategy for nAMD.


Subject(s)
Alanine/analogs & derivatives , Choroidal Neovascularization/pathology , Endothelial Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Triazines/pharmacology , Wet Macular Degeneration/pathology , Alanine/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Choroidal Neovascularization/metabolism , Disease Models, Animal , Lasers , Male , Mice , Mice, Inbred C57BL , Wet Macular Degeneration/metabolism , Zebrafish
12.
J Cardiovasc Pharmacol ; 74(1): 71-79, 2019 07.
Article in English | MEDLINE | ID: mdl-31274845

ABSTRACT

Ganglioside has been implicated to play important roles in modulating various cell signaling and biological functions. However, the functional analysis of a single ganglioside in a zebrafish model is so far lacking. In this study, we investigated the angiogenic effects of 2 monosialoganglioside compounds isolated from GM1 in zebrafish embryos. First, we showed the tested compounds are adequate safe. Then, we found that these compounds exhibited significant proangiogenic effect through enhancement of endothelial cell proliferation, migration, and differentiation. Furthermore, the 2 compounds were proved to promote angiogenesis through, at least partially, modulating the level of Notch signaling. This study provides the novel insights into the clinical application of the 2 ganglioside compounds and GM1.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Embryo, Nonmammalian/blood supply , Endothelial Cells/drug effects , G(M1) Ganglioside/pharmacology , Neovascularization, Physiologic/drug effects , Angiogenesis Inducing Agents/toxicity , Animals , Animals, Genetically Modified , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/metabolism , G(M1) Ganglioside/toxicity , Gene Expression Regulation, Developmental , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
J Org Chem ; 84(3): 1228-1237, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30609359

ABSTRACT

A challenging problem in natural product discovery is to rapidly dereplicate known compounds and expose novel ones from complicated components. Herein, integrating the LC-MS/MS-dependent molecular networking and 1H NMR techniques efficiently and successfully enabled the targeted identification of seven new cyclohexadepsipeptides, chrysogeamides A-G (1-7), from the coral-derived fungus Penicillium chrysogenum (CHNSCLM-0003) which was targeted from a library of marine-derived Penicillium fungi. Compound 4 features a rare 3-hydroxy-4-methylhexanoic acid (HMHA) moiety which was first discovered from marine-derived organisms. Interestingly, isotope-labeling feeding experiments confirmed that 13C1-l-Leu was transformed into 13C1-d-Leu moiety, indicating that d-Leu could be isomerized from l-Leu. Compounds 1 and 2 obviously promoted angiogenesis in zebrafish at 1.0 µg/mL with nontoxic to embryonic zebrafish at 100 µg/mL. Combining molecular networking with 1H NMR as a discovery tool will be implemented as a systematic strategy, not only for known compounds dereplication but also for untapped reservoir discovery.


Subject(s)
Biological Products/chemistry , Fungi/chemistry , Penicillium/chemistry , Tandem Mass Spectrometry/methods , Aquatic Organisms , Proton Magnetic Resonance Spectroscopy
14.
Angiogenesis ; 22(2): 211-221, 2019 05.
Article in English | MEDLINE | ID: mdl-30656567

ABSTRACT

MicroRNAs (miRNAs) are single-stranded small non-coding RNAs, generally 18-25 nucleotides in length, that act as repressors of gene expression. miRNAs are encoded by independent genes or processed from a variety of different RNA species. So far, there is no evidence showing that the ribosomal DNA-hosted microRNA is implicated in vertebrate development. Currently, we found a highly expressed small RNA hosted in ribosomal DNA was predicted as a novel miRNA, named miR-ntu1, in zebrafish endothelial cells by deep sequencing analysis. The miRNA was validated by custom-designed Taqman PCR, Northern Blot, and in silico analysis. Furthermore, we demonstrated that miR-ntu1 played a crucial role in zebrafish angiogenesis via modulation of Notch signaling. Our findings provide a notable case that a miRNA hosted in ribosomal DNA is involved in vertebrate development.


Subject(s)
DNA, Ribosomal/genetics , Endothelium, Vascular/embryology , MicroRNAs/physiology , Neovascularization, Physiologic/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Animals, Genetically Modified , Cloning, Molecular , Embryo, Nonmammalian/blood supply , Embryonic Development/genetics , Endothelium, Vascular/physiology , MicroRNAs/genetics
15.
Oncogene ; 37(21): 2873-2889, 2018 05.
Article in English | MEDLINE | ID: mdl-29520105

ABSTRACT

Benefiting from more precise imaging and radiotherapy, patients with locoregionally nasopharyngeal carcinoma (NPC) have a significantly higher survival rate. Nonetheless, distant metastasis is still the predominant mode of failure. Advances in cancer research have highlighted that pathological angiogenesis is necessary for tumor metastasis by offering oxygen, nutrients, or cell metastatic conduits. MicroRNAs (miRNAs), a class of small noncoding RNAs, are increasingly implicated in modulation of angiogenesis in physiological and pathological conditions. Currently, we detected that miR-23a was highly enriched in NPC tissues at the metastatic or premetastatic stage, and its levels in NPC were associated with microvessel density. Subsequently, we proved that alteration of miR-23a expression modulated the growth, migration, and tube formation of HUVECs in vitro and affected the blood vessel outgrowth in the zebrafish model. Considering the possibility that extracellular miR-23a was horizontally transferred from CNE2 cells to HUVECs, we analyzed miR-23a encapsulated in exosomes, showing that overexpression of exosomal miR-23a in NPC promoted angiogenesis both in vitro and in vivo. Moreover, we provided evidences that miR-23a regulated angiogenesis by directly targeting testis-specific gene antigen (TSGA10). Taken together, our findings revealed that metastasis-associated miR-23a from NPC-derived exosomes plays an important role in mediating angiogenesis by targeting TSGA10.


Subject(s)
Exosomes/genetics , MicroRNAs/genetics , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology , Neovascularization, Pathologic/genetics , Proteins/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Cytoskeletal Proteins , Disease Progression , Exosomes/pathology , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , Mice , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , Neoplasm Metastasis , Up-Regulation , Zebrafish
16.
Front Mol Neurosci ; 10: 274, 2017.
Article in English | MEDLINE | ID: mdl-28894416

ABSTRACT

Insulinoma-associated1a (insm1a) is a zinc-finger transcription factor playing a series of functions in cell formation and differentiation of vertebrate central and peripheral nervous systems and neuroendocrine system. However, its roles on the development of motor neuron have still remained uncovered. Here, we provided evidences that insm1a was a vital regulator of motor neuron development, and provided a mechanistic understanding of how it contributes to this process. Firstly, we showed the localization of insm1a in spinal cord, and primary motor neurons (PMNs) of zebrafish embryos by in situ hybridization, and imaging analysis of transgenic reporter line Tg(insm1a: mCherry)ntu805 . Then we demonstrated that the deficiency of insm1a in zebrafish larvae lead to the defects of PMNs development, including the reduction of caudal primary motor neurons (CaP), and middle primary motor neurons (MiP), the excessive branching of motor axons, and the disorganized distance between adjacent CaPs. Additionally, knockout of insm1 impaired motor neuron differentiation in the spinal cord. Locomotion analysis showed that swimming activity was significantly reduced in the insm1a-null zebrafish. Furthermore, we showed that the insm1a loss of function significantly decreased the transcript levels of both olig2 and nkx6.1. Microinjection of olig2 and nkx6.1 mRNA rescued the motor neuron defects in insm1a deficient embryos. Taken together, these data indicated that insm1a regulated the motor neuron development, at least in part, through modulation of the expressions of olig2 and nkx6.1.

17.
Front Physiol ; 8: 214, 2017.
Article in English | MEDLINE | ID: mdl-28443032

ABSTRACT

The monolayer of endothelial cells (ECs) lining the intima of all blood vessel wall forms a semipermeable barrier that regulates tissue-fluid homeostasis, transport of nutrients, and migration of blood cells across the barrier. A number of signaling pathways and molecules mediate endothelial permeability, which plays important roles in a variety of the physiological and pathological conditions. Fatty acid binding proteins (FABPs) are able to bind various hydrophobic molecules, such as long-chain fatty acids, prostaglandins and eicosanoids. FABP4, a member of the family of FABPs, plays an important role in maintenance of glucose and lipid homeostasis as well as angiogenesis. In the present study, we found that fabp11a, the ortholog of mammalian FABP4, was highly expressed in developing brain vessels of zebrafish. Knockout of fabp11a gene caused hemorrhage in zebrafish brain. Morpholino mediated fabp11a gene knockdown phenocopied the hemorrhage in mutants. Furthermore, we demonstrated permeability of brain vessels in fabp11a mutant is significantly higher than that of control. In addition, COX and LOX inhibition partially rescued the brain vessel integrity defects caused by fabp11a loss-of-function, suggesting the integrity defect was relevant to the Fatty Acid function.

18.
J Cell Biochem ; 118(10): 3495-3510, 2017 10.
Article in English | MEDLINE | ID: mdl-28338241

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disease, involving resting tremor and bradykinesia, for which no recognized therapies or drugs are available to halt or slow progression. In recent years, natural botanic products have been considered relatively safe, with limited side effects, and are expected to become an important source for clinical mediation of PD in the future. Our study focuses on the ability of loganin, a compound derived from fruits of cornus, to mediate neuroprotection in a mouse model of PD. Mice were administered 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) with a dosage of 30 mg/kg daily for 5 days to establish a subacute PD model and treated with loganin. Locomotor activity was assessed by a pole test, then mice were euthanized at 1 and 3 days after the last treatment, and brain tissue was prepared for subsequent assays. Loganin rescued decrease of dopamine levels and tyrosine hydroxylase (TH) expression in the striatum, and shortened total locomotor activity (TLA) time of mice. Furthermore, loganin alleviated microglia and astrocyte activation, and suppressed TNF-α and caspase-3 expression through a c-Abl-p38-NFκB pathway. Loganin also downregulated LC3-II and Drp1 expression, and decreased the level of acidic vesicular organelles (AVOs). Loganin exerts neuroprotective effects on MPTP-induced PD mice by decreasing inflammation, autophagy, and apoptosis, suggesting that loganin could serve as a therapeutic drug to ameliorate PD. J. Cell. Biochem. 118: 3495-3510, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Astrocytes/metabolism , Corpus Striatum/metabolism , Iridoids/pharmacology , MPTP Poisoning/prevention & control , Microglia/metabolism , Parkinson Disease, Secondary/prevention & control , Animals , Astrocytes/pathology , Corpus Striatum/pathology , Dopamine/metabolism , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Male , Mice , Microglia/pathology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Tyrosine 3-Monooxygenase/metabolism
19.
Mol Neurobiol ; 54(3): 2126-2142, 2017 04.
Article in English | MEDLINE | ID: mdl-26924319

ABSTRACT

Ischemic stroke is a major disability and cause of death worldwide due to its narrow therapeutic time window. Neuroprotective agent is a promising strategy to salvage acutely ischemic brain tissue and extend the therapeutic time window for stroke treatment. In this study, we aimed to evaluate the neuroprotective effects of isoquercetin in (1) primary culture of rat hippocampal neurons exposure on oxygen and glucose deprivation and reperfusion (OGD/R) injury and (2) rats subjected to transient middle cerebral artery occlusion and reperfusion (MCAO/R) injury. The results showed that isoquercetin post-treatment reduced the infarct size, number of apoptotic cells, oxidative stress, and inflammatory response after ischemia and reperfusion injury. The underlying mechanism study indicated that the neuroprotective effects of isoquercetin were elicited via suppressing the activation of toll-like receptor 4 (TLR4), nuclear factor-kappa B (NF-κB) and caspase-1; the phosphorylation of ERK1/2, JNK1/2, and p38 mitogen-activated protein kinase (MAPK); and the secretion of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and IL-6. In addition, isoquercetin also effectively alleviated hippocampus neuron apoptosis by regulation of cyclic AMP responsive element-binding protein (CREB), Bax, Bcl-2, and caspase-3. Our report provided new considerations into the therapeutic action and the underlying mechanisms of isoquercetin to improve brain injury in individuals who have suffered from ischemic stroke. As a potent anti-inflammatory and anti-oxidative compound with neuroprotective capacities, the beneficial effects of isoquercetin when used to treat ischemic stroke and related diseases in humans warrant further studies.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Brain Ischemia/drug therapy , CA1 Region, Hippocampal/drug effects , Neurons/drug effects , Oxidative Stress/drug effects , Quercetin/analogs & derivatives , Animals , Brain Ischemia/pathology , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Cells, Cultured , Inflammation/drug therapy , Ischemia/drug therapy , Male , Neurons/metabolism , Neuroprotective Agents/pharmacology , Quercetin/pharmacology , Rats, Sprague-Dawley , Reperfusion Injury/pathology
SELECTION OF CITATIONS
SEARCH DETAIL