Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Anticancer Drugs ; 33(10): 1081-1090, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35946567

ABSTRACT

Brain metastasis is a devastating clinical condition globally as one of the most common central nervous system malignancies. The current study aimed to assess the effect of defibrotide, an Food and Drug Administration-approved drug, against brain metastasis and the underlying molecular mechanisms. Two tumor cell lines with high brain metastasis potential, PC-9 and 231-BR, were subjected to defibrotide treatment of increasing dosage. The metastasis capacity of the tumor cells was evaluated by cell invasion and migration assays. Western blotting was employed to determine the levels of tight junction proteins in the blood-brain barrier (BBB) including Occludin, Zo-1, and Claudin-5, as well as metastasis-related proteins including CXCR4, MMP-2, and MMP-9. The in-vitro observations were further verified in nude mice, by monitoring the growth of xenograft tumors, mouse survival and brain metastasis foci following defibrotide treatment. Defibrotide inhibited proliferation, migration, invasion, and promotes lactate dehydrogenase release of brain metastatic tumor cells, elevated the levels of BBB tight junction proteins and metastasis-related proteins. Such beneficial role of defibrotide was mediated by its inhibitory action on the SDF-1/CXCR4 signaling axis both in vitro and in vivo , as CXCR4 agonist SDF1α negated the anti-tumoral effect of defibrotide on mouse xenograft tumor growth, mouse survival and brain metastasis. Defibrotide inhibits brain metastasis through activating the adenosine A2A receptors, which in turn inhibits the SDF-1/CXCR4 signaling axis. Our study hereby proposes defibrotide as a new and promising candidate drug against brain metastasis of multiple organ origins.


Subject(s)
Brain Neoplasms , Receptor, Adenosine A2A , Animals , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement , Chemokine CXCL12/metabolism , Chemokine CXCL12/pharmacology , Claudin-5/pharmacology , Humans , Lactate Dehydrogenases , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Mice , Mice, Nude , Neoplasm Metastasis , Occludin/pharmacology , Polydeoxyribonucleotides , Receptor, Adenosine A2A/metabolism , Receptors, CXCR4/metabolism
2.
Cerebrovasc Dis ; 51(5): 678-685, 2022.
Article in English | MEDLINE | ID: mdl-35421860

ABSTRACT

BACKGROUND: Recent studies have shown that curcumin can reduce the symptoms of hydrocephalus. However, the underlying mechanisms remain unclear. Our previous studies demonstrated that E2F transcription factor 4 (E2F4) protein plays an important role in hydrocephalus; hence, we hypothesized that E2F4 may involve in curcumin mediated anti-hydrocephalus benefits. METHODS: E2F4 expression and functions in different human tissues and cell lines were determined and analyzed using the all RNA-seq and ChIP-seq sample and signature search database and ChIP-atlas database. Hydrocephalus mouse model was established through stereotactic injection of shE2F4 into frontal cortex. Mice were treated with curcumin, and then hydrocephalus severity, the expression of E2F4, and downstream targets were analyzed. RESULTS: E2F4 was highly expressed in the nervous system, which was downregulated in the bran of hydrocephalus patients. Knockdown E2F4 in mice could mimic the phenotype of human hydrocephalus. Upon curcumin administration, E2F4 expression level was increased, and the hydrocephalus severity score was significantly decreased in mouse model. Mechanistically, curcumin attenuated hydrocephalus through activating E2F4 signaling pathway. CONCLUSION: Curcumin suppresses hydrocephalus progression via activation of E2F4, which could be a target for hydrocephalus treatment.


Subject(s)
Curcumin , Animals , Cell Line , Curcumin/pharmacology , E2F4 Transcription Factor/genetics , E2F4 Transcription Factor/metabolism , Humans , Mice
3.
Neuroimmunomodulation ; 29(4): 439-449, 2022.
Article in English | MEDLINE | ID: mdl-35609523

ABSTRACT

BACKGROUND: Neuroinflammation subsequent to traumatic brain injury (TBI) is important for the recovery of patients and is associated with neurodegenerative changes post-TBI. The tripartite motif containing 44 (TRIM44) protein is an E3 ligase involved in the regulation of immune function with no previously known link to TBI. This study explores the connection between TRIM44 and TBI. METHODS: After induction of TBI in rats by control cortex injury, TRIM44 expressions were determined with quantitative real-time reverse transcription polymerase chain reaction and Western blot, and Toll-like receptor 4 (TLR4)-NF-κB signaling was examined by the expression of TLR4, p65 phosphorylation, and the specific NF-κB transcription activity. The effects of TRIM44 knockdown on inflammation, neurological function, and TLR4-NF-κB signaling in TBI rats were revealed by the detection of proinflammatory cytokines and TLR4-NF-κB signaling molecules, modified neurological severity score, brain water content, and Evans blue permeability. RESULTS: We found that TRIM44 expression was significantly increased following TBI induction along with TLR4-NF-κB activation. Silencing of TRIM44 suppressed proinflammatory cytokine production, improved neurological outcomes, alleviated brain edema, and inhibited TLR4-NF-κB signaling in TBI rats. CONCLUSION: Our findings suggest that suppressing TRIM44 or modulation of relevant pathways may be a therapeutic strategy for TBI.


Subject(s)
Brain Injuries, Traumatic , Inflammation , Tripartite Motif Proteins , Animals , Rats , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/immunology , Inflammation/genetics , Inflammation/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Rats, Sprague-Dawley , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/immunology
4.
Metab Brain Dis ; 36(1): 123-132, 2021 01.
Article in English | MEDLINE | ID: mdl-32880813

ABSTRACT

This study aimed to characterize the expression status and potentially mechanistic involvement of SNHG7 in pituitary adenoma. Relative expression of SNHG7 and miR-449a was analyzed by real-time PCR. Cell viability was measured with Cell Counting Kit-8 (CCK-8). Cell apoptosis was determined by PI/Annexin V double staining followed by flow cytometry analysis. Cell invasion and migration were analyzed by wound healing and transwell assays, respectively. The regulatory action of miR-449a on SNHG7 was interrogated by luciferase reporter assay. We also investigated the pro-tumor activity of SNHG7 with the MMQ xenograft tumor mouse model. We identified the aberrant up-regulation of SNHG7 in pituitary adenoma both in vivo and in vitro, which associated with poor survival outcome. siRNA-mediated SNHG7-knockdown decreased cell viability, increased apoptosis and compromised migration and invasion. We further predicted and validated that SNHG7 negatively regulated miR-449a via sponging. Concurrent inhibition of miR-449a restored cell viability, apoptosis, migration and invasion influenced by SNHG7-deficiency. Most importantly, we demonstrated that SNHG7-silencing delayed xenograft tumor progression, which was accompanied with increased miR-449a and decreased Ki67 intensity. Our study highlighted the essential oncogenic properties of the SNHG7/miR-449a axis in pituitary adenoma.


Subject(s)
Adenoma/metabolism , MicroRNAs/metabolism , Pituitary Gland/metabolism , Pituitary Neoplasms/metabolism , RNA, Long Noncoding/metabolism , Adenoma/genetics , Adenoma/pathology , Animals , Apoptosis/physiology , Cell Line , Cell Movement/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Disease Progression , Humans , MicroRNAs/genetics , Pituitary Gland/pathology , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , RNA, Long Noncoding/genetics , Rats
5.
J Pharm Pharm Sci ; 21(1): 318-325, 2018.
Article in English | MEDLINE | ID: mdl-30070971

ABSTRACT

Purpose To investigate the role of ICA II in subarachnoid hemorrhage (SAH)-  related chronic hydrocephalus. Methods A two hemorrhage injection model of SAH was created in Sprague Dawley rats (6-8 weeks). A total of 125 rats were randomly assigned into five groups: Sham group, SAH group, SAH+ ICA II (1 mg/kg) group, SAH + ICA II (5 mg/kg) group, and SAH + ICA II (10 mg/kg) group. TGF-ß1, phospho-Smad2/3, connective tissue growth factor (CTGF), and procollagen type I carboxy-terminal propeptide (PICP) were assessed via real-time PCR, Western blotting, and enzyme-linked immunosorbent assay. Lateral ventricular index, Masson staining, and Morris water maze tests were employed to evaluate subarachnoid fibrosis, hydrocephalus, and long term neurological function following SAH. Results ICA II (1, 5, 10 mg/kg) inhibited subarachnoid fibrosis, attenuated ventriculomegaly, and effectively suppressed SAH related chronic hydrocephalus. In addition, parallel reduced expression of members of the TGF-ß1/Smad/CTGF signaling pathway were observed. Importantly, ICA II may improve long term neurocognitive deficits after SAH. Conclusion ICA II might suppress fibrosis via inhibition of TGF-ß1/Smad/CTGF pathway, prevent the development of SAH related chronic hydrocephalus, and improve long term neurocognitive defects following SAH.


Subject(s)
Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Flavonoids/pharmacology , Hydrocephalus/drug therapy , Subarachnoid Hemorrhage/drug therapy , Administration, Oral , Animals , Chronic Disease , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/administration & dosage , Flavonoids/administration & dosage , Male , Maze Learning/drug effects , Rats , Rats, Sprague-Dawley
6.
Gynecol Oncol ; 143(3): 655-663, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27726922

ABSTRACT

OBJECTIVE: Ovarian cancer is one of the most lethal gynecologic malignancies worldwide and with poor prognosis and survival rate in women. Identifying sensitive and specific molecular in carcinogenesis may improve diagnostic and therapeutic strategies for this malignancy and achieve a better clinical outcome. METHODS: miR-760 expression in ovarian cancer cell lines and patient tissues were determined using Real-time PCR. 145 human ovarian cancer tissue samples were analyzed by RT-PCR to investigate the association between miR-760expression and the clinicopathological characteristics of ovarian cancer patients. Functional assays, such as MTT, anchorage-independent growth, colony formation and BRDU assay were used to determine the oncogenic role of miR-760 in human ovarian cancer progression. Furthermore, western blotting and luciferase assay were used to determine the mechanism of miR-760 promotes proliferation in ovarian cancer cells. RESULT: The expression of miR-760 was markedly upregulated in ovarian cancer cell lines and tissues, and high miR-760 expression was associated with an aggressive phenotype and poor prognosis with ovarian cancer patients. Upregulation of miR-760 promoted, whereas downregulation of miR-760 inhibited the proliferation of ovarian cancer cells in vitro. Additionally, we identified PHLPP2 as a direct target of miR-760, and silencing the expression of PHLPP2 is the essential biological function of miR-760 during ovarian cancer cell proliferation. Finally, we showed a significant correlation between miR-760 and PHLPP2 expression in ovarian cancer tissues. CONCLUSION: Our findings suggest that miR-760 represents a potential onco-miR and participates in ovarian cancer carcinogenesis, which highlight its potential as a target for ovarian cancer therapy.


Subject(s)
Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Ovarian Neoplasms/genetics , Phosphoprotein Phosphatases/metabolism , Blotting, Western , Carcinogenesis/genetics , Cell Line, Tumor , Down-Regulation , Female , HEK293 Cells , Humans , In Vitro Techniques , Ovarian Neoplasms/metabolism , Real-Time Polymerase Chain Reaction , Up-Regulation
7.
Chin Med J (Engl) ; 132(11): 1344-1353, 2019 Jun 05.
Article in English | MEDLINE | ID: mdl-30939485

ABSTRACT

BACKGROUND: The increased permeability of the blood-brain barrier (BBB) induced by ischemia/hypoxia is generally correlated with alteration of tight junctions (TJs). DL-3-n-butylphthalide (NBP) has been shown to exert neuroprotective effects after ischemic injury. However, few studies have assessed the correlation between NBP and TJs. This study aimed to investigate the potential effect of NBP on the TJ proteins claudin-5, zonula occludens-1 (ZO-1), and occludin during brain ischemia. METHODS: A chronic cerebral hypoperfusion (CCH) Sprague-Dawley rat model was established, and NBP (20, 40, or 80 mg/kg, gavage, once a day) treatment was performed for 14 days. NBP (0.1 or 1.0 µmol/L) pre-treatment was applied to an in vitro hypoxia microvascular endothelial cell model (1% O2, 24 h). BBB permeability was assessed by performing the Evans blue assay. The expressions and localization of claudin-5, ZO-1, occludin, phosphorylated/total protein kinase B (p-Akt/Akt), phosphorylated/total glycogen synthase kinase 3ß (GSK-3ß)/GSK-3ß, and ß-catenin/ß-actin were evaluated by Western blotting or immunofluorescence. Reactive oxygen species (ROS) generation was measured by flow cytometry analysis. TJ ultrastructure was observed by transmission electron microscopy. RESULTS: In CCH rats, treatment with 40 and 80 mg/kg NBP decreased the Evans blue content in brain tissue (9.0 ±â€Š0.9 µg/g vs. 12.3 ±â€Š1.9 µg/g, P = 0.005; 6.7 ±â€Š0.6 µg/g vs. 12.3 ±â€Š1.9 µg/g, P < 0.01), increased the expression of claudin-5 (0.79 ±â€Š0.08 vs. 0.41 ±â€Š0.06, P < 0.01; 0.97 ±â€Š0.07 vs. 0.41 ±â€Š0.06, P < 0.01), and elevated the ZO-1 protein level (P < 0.05) in brain microvascular segments in a dose-dependent manner in comparison with the corresponding values in the model group. There was no significant difference in occludin expression (P > 0.05). In the hypoxia cell model, NBP pre-treatment improved TJ ultrastructure, decreased intracellular ROS level, and increased the expression of claudin-5 (P < 0.01) and ZO-1 (P < 0.01) in comparison with the corresponding values in the hypoxia group. NBP treatment also elevated the relative expression levels of p-Akt/Akt, p-GSK-3ß/GSK-3ß, and ß-catenin/ß-actin in comparison with the corresponding values in the hypoxia group (all P < 0.05). CONCLUSION: NBP improves the barrier function of BBB against ischemic injury by upregulating the expression of TJ proteins, possibly by reducing oxidative stress and activating the Akt/GSK-3ß/ß-catenin signaling pathway.


Subject(s)
Benzofurans/therapeutic use , Blood-Brain Barrier/drug effects , Tight Junction Proteins/metabolism , Animals , Blood-Brain Barrier/ultrastructure , Blotting, Western , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Cells, Cultured , Claudin-5/metabolism , Immunohistochemistry , Male , Microscopy, Electron, Transmission , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Zonula Occludens-1 Protein/metabolism
8.
J Neurol Surg A Cent Eur Neurosurg ; 80(6): 498-502, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31075809

ABSTRACT

BACKGROUND: Cranioplasty is a common procedure in neurosurgery. It is usually performed following decompressive craniectomy (DC). However, complications may occur after the operation, such as massive brain swelling. Up to now, far too little attention has been given to this severe complication. We report one case of fatal cerebral swelling after cranioplasty and analyze the possible mechanism of this complication. CASE DESCRIPTION: The patient was a 40-year-old man who had a severe right basal ganglia cerebral hemorrhage and underwent DC ∼ 2 months before. One day before scheduled cranioplasty, a lumbar cerebrospinal fluid drainage was placed. The cranioplasty itself was uneventful. However, he gradually fell into a coma, and his right pupil was moderately dilated 20 hours after the surgery. A brain computed tomography (CT) scan indicated massive right cerebral edema with compressed right midbrain. The patient did not regain consciousness, and he remained quadriplegic. CONCLUSION: It is necessary to increase awareness of complications of cranioplasty in high-risk patients. The lessons learned from this case include avoiding excessive drainage of cerebrospinal fluid. Patients with low-density lesions in the brain need to be treated with caution. Once the CT scan shows massive cerebral swelling, the patient has a poor prognosis.


Subject(s)
Basal Ganglia Hemorrhage/surgery , Brain Edema/etiology , Decompressive Craniectomy/adverse effects , Plastic Surgery Procedures/adverse effects , Adult , Basal Ganglia Hemorrhage/diagnostic imaging , Brain Edema/diagnostic imaging , Drainage , Humans , Male , Postoperative Complications/diagnostic imaging , Postoperative Complications/etiology , Tomography, X-Ray Computed
10.
Oncol Rep ; 31(5): 2279-85, 2014 May.
Article in English | MEDLINE | ID: mdl-24626771

ABSTRACT

Although implicated in a number of tumor types, the role of N-myc downstream regulated gene 1 (NDRG1) in ovarian cancer (OC) is unclear. In the present study, we used short hairpin RNA (shRNA) to silence NDRG1 in the OC cell line OVCAR3 and assessed the effect of its knockdown on cell morphology, proliferation, colony formation, migration and invasion. To complement these knockdown studies, we overexpressed NDRG1 in the same cell line. We found that NDRG1 knockdown significantly enhanced OVCAR3 proliferation, migration and invasion; however, there were no apparent changes in cell morphology. We also examined the effect in vivo and found that NDRG1 depletion promoted OVCAR3 xenograft growth in nude mice. In accordance with these data, we found that NDRG1 overexpression decreased proliferation, adhesion and apoptosis, and induced G0/G1 cell cycle arrest in OVCAR3 cells; expression of p21 and p53 was also increased. In conclusion, we demonstrated that NDRG1 acts as a tumor suppressor in ovarian carcinogenesis and may be a potential therapeutic target in this disease.


Subject(s)
Cell Cycle Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Neoplasm Invasiveness/genetics , Ovarian Neoplasms/pathology , Animals , Apoptosis/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Female , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , RNA Interference , RNA, Small Interfering , Transplantation, Heterologous , Tumor Suppressor Protein p53/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL