Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Med Sci Monit ; 25: 1291-1300, 2019 Feb 17.
Article in English | MEDLINE | ID: mdl-30772888

ABSTRACT

BACKGROUND The phosphatase actin regulator-1 (PHACTR-1) gene on chromosome 6 encodes an actin and protein phosphatase 1 (PP1) binding protein, Phactr-1, which is highly expressed in brain tissues. Phactr-1 expression is involved in physiological and pathological cerebral microvascular events. This study aimed to investigate the role of expression of Phactr-1 in a mouse brain capillary endothelial cell line, bEnd.3, by knockdown the PHACTR-1 gene. MATERIAL AND METHODS Three bEnd.3 cell groups were studied, CON (normal control cells), NC (control scramble transfected cells), and KD (cells with PHACTR-1 gene knockdown). The PHACTR-1 gene was knocked down using transfection with small hairpin RNA (shRNA). In the three cell groups cell proliferation, migration, and apoptosis were studied by MTT and colony formation assays, transwell and scratch assays, and flow cytometry. The related cell pathways of associated with Phactr-1 knockdown were studied by Western blot. RESULTS Phactr-1 knockdown suppressed bEnd.3 cell proliferation and migration, promoted cell apoptosis, and downregulated the expressions of migration-associated proteins, including matrix metalloproteinase (MMP)-2 and MMP-9 and upregulated apoptosis-associated proteins, including Bax, Bcl-2, cleaved caspase-3, and caspase-3. CONCLUSIONS Phactr-1 was shown to have a role in the inhibition of endothelial cell proliferation and migration, promoted cell apoptosis, and regulated matrix metalloproteinases and apoptosis-associated proteins. These findings indicate that the expression of the Phactr-1 should be studied further in the cerebral microvasculature, both in vitro and in vivo, regarding its potential as a diagnostic and therapeutic target for cerebral microvascular disease.


Subject(s)
Brain/blood supply , Endothelial Cells/metabolism , Microfilament Proteins/genetics , Actins/metabolism , Animals , Apoptosis/genetics , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation , Endothelial Cells/cytology , Gene Knockdown Techniques , Mice , Microfilament Proteins/biosynthesis , Microfilament Proteins/deficiency , Phosphoric Monoester Hydrolases/metabolism , Signal Transduction , Transfection
2.
Med Sci Monit ; 23: 4241-4251, 2017 Sep 02.
Article in English | MEDLINE | ID: mdl-28865235

ABSTRACT

BACKGROUND This study aimed to evaluate the effects of electro-acupuncture (EA) on neuroplasticity associated with the expressions of neurotrophic factors (NTFs) and their receptors in rats subjected to spinal cord transection (SCT). MATERIAL AND METHODS A total of 144 rats were randomly divided into 3 groups (n=48 per group): sham-operated group, SCT group, and EA (electro-acupuncture) group. Rats in SCT and EA groups received spinal cord transection at T10-T11 vertebral levels. Then, EA group rats received EA treatment. Reverse transcription polymerase chain reaction was used to detect NTFs and receptors at the mRNA level. In situ hybridization (ISH) and immunohistochemistry (IHC) were used to detect the expression of NTFs and their receptors. Basso, Beattie, Bresnahan (BBB) scores and cortical somato-sensory evoked potentials (CSEP) were evaluated to assess the recovery of motor and sensory functions. We also measured BDA (Biotinylated dextran amine) axonal tracing, CGRP (Calcitonin gene-related peptide), GAP-43 (Growth-associated protein), and synaptophysin immunohistochemistry (IHC). RESULTS EA treatment led to obvious improvement in hindlimb locomotor and sensory functions. CNTF, FGF-2, and TrkB mRNA were significantly upregulated, while NGF, PDGF, TGF-b1, IGF-1, TrkA, and TrkC mRNA were concomitantly downregulated in the caudal spinal segment (CSS) following EA. Immunohistochemistry demonstrated an increased number of CGRP fibers, GAP-43, and synaptophysin profiles in the CSS in the EA rats. CONCLUSIONS EA may promote the recovery of neuroplasticity in rats subjected to SCT. This could be attributed to the systematic regulation of NTFs and their receptors after EA.


Subject(s)
Electroacupuncture/methods , Neuronal Plasticity/drug effects , Spinal Cord Injuries/therapy , Animals , Nerve Growth Factors/analysis , Nerve Growth Factors/drug effects , Nerve Regeneration/physiology , Neuronal Plasticity/genetics , Rats , Rats, Sprague-Dawley , Recovery of Function
3.
World J Surg Oncol ; 14(1): 267, 2016 Oct 19.
Article in English | MEDLINE | ID: mdl-27756319

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most malignant nervous system tumor with an almost 100 % recurrence rate. Thymopoietin (TMPO) has been demonstrated to be upregulated in various tumors, including lung cancer, breast cancer, and so on, but its role in GBM has not been reported. This study was aimed to determine the role of TMPO in GBM. METHODS: Publicly available Oncomine dataset analysis was used to explore the expression level of TMPO in GBM specimens. Then the expression of TMPO was knocked down in GBM cells using lentiviral system, and the knockdown efficacy was further validated by real-time quantitative PCR and western blot analysis. Furthermore, the effects of TMPO silencing on GBM cell proliferation and apoptosis were examined by MTT, colony formation, and flow cytometry analysis. Meanwhile, the expression of apoptotic markers caspase-3 and poly(ADP-ribose) polymerase (PARP) were investigated by western blot analysis. RESULTS: This study observed that the expression of TMPO in GBM specimens was remarkably higher than that in normal brain specimens. Moreover, knockdown of TMPO could significantly inhibit cell proliferation and arrest cell cycle progression at the G2/M phase. It also found that TMPO knockdown promoted cell apoptosis by upregulation of the cleavage of caspase-3 and PARP protein levels which are the markers of apoptosis. CONCLUSIONS: The results suggested TMPO might be a novel therapeutic target for GBM.


Subject(s)
Apoptosis , Brain Neoplasms/pathology , Cell Cycle Checkpoints , Glioblastoma/pathology , Nuclear Proteins/metabolism , Thymopoietins/metabolism , Apoptosis/genetics , Brain Neoplasms/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation , Gene Knockdown Techniques , Glioblastoma/metabolism , Humans , Neoplasm Recurrence, Local , Nuclear Proteins/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , RNA Interference , Thymopoietins/genetics , Up-Regulation
4.
Neural Regen Res ; 18(7): 1578-1583, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36571365

ABSTRACT

Studies have found that the phosphatase actin regulatory factor 1 expression can be related to stroke, but it remains unclear whether changes in phosphatase actin regulatory factor 1 expression also play a role in traumatic brain injury. In this study we found that, in a mouse model of traumatic brain injury induced by controlled cortical impact, phosphatase actin regulatory factor 1 expression is increased in endothelial cells, neurons, astrocytes, and microglia. When we overexpressed phosphatase actin regulatory factor 1 by injection an adeno-associated virus vector into the contused area in the traumatic brain injury mice, the water content of the brain tissue increased. However, when phosphatase actin regulatory factor 1 was knocked down, the water content decreased. We also found that inhibiting phosphatase actin regulatory factor 1 expression regulated the nuclear factor kappa B signaling pathway, decreased blood-brain barrier permeability, reduced aquaporin 4 and intercellular adhesion molecule 1 expression, inhibited neuroinflammation, and neuronal apoptosis, thereby improving neurological function. The findings from this study indicate that phosphatase actin regulatory factor 1 may be a potential therapeutic target for traumatic brain injury.

5.
Brain Inj ; 26(10): 1211-6, 2012.
Article in English | MEDLINE | ID: mdl-22571813

ABSTRACT

PURPOSE: To discuss the repeated CT scanning in patients with traumatic brain injury (TBI) and to identify the conditions under which this approach is necessary. METHODS: One hundred and seventy-one patients who suffered TBI but were not surgically treated were divided into two groups: the routine-repeat CT group (n = 89) and the non-routine-repeat CT group (n = 82). The patients' clinical characteristics were compared. T-tests and stepwise logistic regression were used for analysis. Patients in the routine-repeat CT group were divided into three groups according to GCS scores to determine the need for routinely repeated CT scans. RESULTS: The results revealed statistically significant differences between the two groups in terms of neuro-ICU-LOS and LOS (p < 0.01). No significant differences emerged with respect to hospital charges and GCS scores at discharge (p > 0.05). AGE, international normalized ratio (INR), D-dimer concentration (DD), GCS scores and number of hours between the first CT scan and the injury (HCT1) were influential factors of developing progressive haemorrhage. CONCLUSION: The routine-repeat CT group fared better than did the non-routine-repeat CT group. Routinely repeated CTs were minimally effective among those with mild TBI, whereas this procedure demonstrated a significant effect on patients with moderate and severe TBI.


Subject(s)
Brain Injuries/diagnostic imaging , Hematoma, Epidural, Cranial/diagnostic imaging , Hematoma, Subdural, Acute/diagnostic imaging , Tomography, X-Ray Computed , Brain Injuries/physiopathology , China , Diagnostic Tests, Routine , Female , Humans , Male , Middle Aged , Prospective Studies , Risk Assessment
6.
Front Neurosci ; 14: 925, 2020.
Article in English | MEDLINE | ID: mdl-33013306

ABSTRACT

Traumatic brain injury (TBI) could highly induce coagulopathy through breaking the dynamic balance between coagulation and fibrinolysis systems, which may be a major contributor to the progressive secondary injury cascade that occurs after TBI. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) inhibition is reported to exert neuroprotection in TBI, making it a potential regulatory target involved in TBI-induced coagulation disorder. PTEN level is controlled in a major way by E3 ligase-mediated degradation through the ubiquitin-proteasome system. The C terminus of Hsc70-interacting protein (CHIP) has been shown to regulate proteasomal degradation and ubiquitination level of PTEN. In the present study, CHIP was overexpressed and knocked down in mouse brain microvascular endothelial cells (bEnd.3) and tissues during the early phase of TBI. In vitro cell proliferation, cell apoptosis, migration capacity, and invasion capacity were determined. The changes of procoagulant and apoptosis molecules after TBI were also detected as well as the micrangium density and blood-brain barrier permeability after in vivo TBI. In vitro results demonstrated that CHIP overexpression facilitated bEnd.3 cell proliferation, migration, and invasion and downregulated cell apoptosis and the expressions of procoagulant molecules through promoting PTEN ubiquitination in a simulated TBI model with stretch-induced injury treatment. In vivo experiments also demonstrated that CHIP overexpression suppressed post-TBI apoptosis and procoagulant protein expressions, as well as increased microvessel density, reduced hemorrhagic injury, and blood-brain barrier permeability. These findings suggested that the upregulation of CHIP may attenuate apoptosis and procoagulant activity, facilitate brain repair, and thus exerts neuroprotective effects in TBI.

7.
J Alzheimers Dis ; 68(4): 1687-1697, 2019.
Article in English | MEDLINE | ID: mdl-30958378

ABSTRACT

Traumatic brain injury (TBI), a brain disorder that causes death and long-term disability in humans, is increasing in prevalence, though there is a lack of protective or therapeutic strategies for mitigating the damage after TBI and for preserving neurological functionality. Microglia cells play a key role in neuroinflammation following TBI, but their regulation and polarization by a member of the vascular endothelial growth factor (VEGF) family, VEGF-C, is unknown. Here, we show that VEGF-C induced M2 polarization in a murine microglia cell line, BV-2, in vitro, by a mechanism that required signaling from its unique receptor, VEGF receptor 3 (VEGFR3). Moreover, in a TBI model in rats, VEGF-C administration induced M2 polarization of microglia cells, significantly improved motor deficits after experimental TBI, and significantly improved neurological function following TBI, likely through a reduction in cell apoptosis. Together, our data reveal a previously unknown role of VEGF-C/VEGFR3 signaling in the regulation of post-TBI microglia cell polarization, which appears to be crucial for recovery from TBI.


Subject(s)
Brain Injuries, Traumatic/metabolism , Microglia/drug effects , Vascular Endothelial Growth Factor C/pharmacology , Vascular Endothelial Growth Factor Receptor-3/metabolism , Animals , Cell Line , Cell Polarity/drug effects , Disease Models, Animal , Male , Mice , Microglia/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
8.
Mol Inform ; 35(6-7): 262-7, 2016 07.
Article in English | MEDLINE | ID: mdl-27492240

ABSTRACT

Rho-kinase dimerization is essential for its kinase activity and biological function; disruption of the dimerization has recently been established as a new and promising therapeutics strategy for cerebrovascular malformation (CM). Based on Rho-kinase dimer crystal structure we herein combined in silico analysis and in vitro assay to rationally derive self-inhibitory peptides from the dimerization interface. Three peptides namely Hlp1, Hlp2 and Hlp3 were successfully designed that have potential capability to rebind at the dimerization domain of Rho-kinase. Molecular dynamics (MD) simulations revealed that these peptides are helically structured when bound to Rho-kinase, but exhibit partially intrinsic disorder in unbound state. Binding free energy (BFE) analysis suggested that the peptides have a satisfactory energetic profile to interact with Rho-kinase. The computational findings were then substantiated by fluorescence anisotropy assays, conforming that the helical peptides can bind tightly to Rho-kinase with affinity KD at micromolar level. These designed peptides are considered as lead molecular entities that can be further modified and optimized to obtain more potent peptidomimetics as self-competitors to disrupt Rho-kinase dimerization in CM.


Subject(s)
Protein Kinase Inhibitors/chemistry , rho-Associated Kinases/chemistry , Drug Evaluation, Preclinical , Fluorescence Polarization , Intracranial Arteriovenous Malformations/drug therapy , Intracranial Arteriovenous Malformations/enzymology , Molecular Dynamics Simulation , Peptides/chemistry , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Protein Multimerization , Thermodynamics
9.
Biomed Res Int ; 2013: 402375, 2013.
Article in English | MEDLINE | ID: mdl-24089677

ABSTRACT

Recent studies have suggested that endogenous angiogenesis inhibitor endostatin/collagen XVIII might play an important role in the secondary brain injury following traumatic brain injury (TBI). In this study, we measured endostatin/collagen XVIII concentrations serially for 1 week after hospitalization by using the enzyme-linked immunosorbent assay method in the cerebrospinal fluid (CSF) of 30 patients with TBI and a Glasgow Coma Scale (GCS) score of 8 or less on admission. There was a significant trend toward increased CSF levels of endostatin after TBI versus control from 72 h after injury. In patients with GCS score of 3-5, CSF endostatin concentration was substantially higher at 72 h after injury than that in patients with GCS score of 6-8 (P < 0.05) and peaked rapidly at day 5 after injury, but decreased thereafter. The CSF endostatin concentration in 12 patients with an unfavorable outcome was significantly higher than that in 18 patients with a favorable outcome at day 5 (P = 0.043) and day 7 (P = 0.005) after trauma. Receiver operating characteristic curve analysis suggested a reliable operating point for the 7-day CSF endostatin concentration predicting poor prognosis to be 67.29 pg/mL. Our preliminary findings provide new evidence that endostatin/collagen XVIII concentration in the CSF increases substantially in patients with sTBI. Its dynamic change may have some clinical significance on the judgment of brain injury severity and the assessment of prognosis. This trial is registered with the ClinicalTrials.gov Identifier: NCT01846546.


Subject(s)
Brain Injuries/cerebrospinal fluid , Brain Injuries/physiopathology , Collagen Type XVIII/cerebrospinal fluid , Prognosis , Aged , Collagen Type XVIII/isolation & purification , Enzyme-Linked Immunosorbent Assay , Female , Glasgow Coma Scale , Humans , Male , Middle Aged , ROC Curve
10.
Asian Pac J Cancer Prev ; 13(6): 2837-40, 2012.
Article in English | MEDLINE | ID: mdl-22938469

ABSTRACT

We investigated whether IFN-ß inhibits the growth of human malignant glioma and induces glioma cell apoptosis using the human IFN-ß gene transfected into glioma cells. A eukaryonic expression vector (pSV2IFNß) for IFN-ß was transfected into the glioma cell line SHG44 using liposome transfection. Stable transfection and IFN-ß expression were confirmed using an enzyme-linked immunosorbent assay (ELISA). Cell apoptosis was also assessed by Hoechst staining and electron microscopy. In vivo experiments were used to establish a SHG44 glioma model in nude mice. Liposomes containing the human IFN-ß gene were injected into the SHG44 glioma of nude mice to observe glioma growth and calculate tumor size. Fas expression was evaluated using immunohistochemistry. The IFN-ß gene was successfully transfected and expressed in the SHG44 glioma cells in vitro. A significant difference in the number of apoptotic cells was observed between transfected and non- transfected cells. Glioma growth in nude mice was inhibited in vivo, with significant induction of apoptosis. Fas expression was also elevated. The IFN-ß gene induces apoptosis in glioma cells, possibly through upregulation of Fas. The IFN-ß gene modulation in the Fas pathway and apoptosis in glioma cells may be important for the treatment of gliomas.


Subject(s)
Apoptosis , Glioma/metabolism , Glioma/pathology , Interferon-beta/genetics , fas Receptor/biosynthesis , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Gene Transfer Techniques , Genetic Therapy , Glioma/genetics , Humans , Mice , Mice, Nude , Up-Regulation , fas Receptor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL