Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
Aging (Albany NY) ; 16(5): 4191-4203, 2024 Mar 09.
Article En | MEDLINE | ID: mdl-38461458

Glioblastoma is the most common malignant tumor in the central nervous system and its occurrence and development is involved in various molecular abnormalities. C-X-C chemokine ligand 10 (CXCL10), an inflammatory chemokine, has been reported to be related to the pathogenesis of cancer while it has not yet been linked to glioma. Calycosin, a bioactive compound derived from Radix astragali, has demonstrated anticancer properties in several malignancies, including glioma. Nonetheless, its underlying mechanisms are not fully understood. This study explores CXCL10 as a potential therapeutic target for calycosin in the suppression of glioblastoma. We observed that CXCL10 expression correlates positively with glioma malignancy and inversely with patient prognosis, highlighting its potential as a glioblastoma treatment target. Furthermore, we found that calycosin inhibited proliferation, migration, and invasion in U87 and U251 glioma cells, and decreased CXCL10 expression in a dose-dependent manner, along with its downstream effectors such as NLRP3, NF-κB, and IL-1ß. Additionally, molecular docking experiments demonstrated that calycosin exhibits a notable binding affinity to CXCL10. Overexpression of CXCL10 counteracted the inhibitory effects of calycosin on cell proliferation, migration, and invasion, while CXCL10 knockdown enhanced these effects. Finally, we verified that calycosin inhibited glioma growth in a xenograft mouse model and downregulated CXCL10 and its downstream molecules. These findings suggest that targeting CXCL10 may be an effective strategy in glioblastoma treatment, and calycosin emerges as a potential therapeutic agent.


Glioblastoma , Glioma , Isoflavones , Humans , Mice , Animals , Glioblastoma/pathology , Molecular Docking Simulation , Ligands , Cell Line, Tumor , Glioma/pathology , Cell Proliferation , Disease Models, Animal , Signal Transduction , Cell Movement , Chemokine CXCL10/genetics
2.
J Med Chem ; 67(3): 2095-2117, 2024 Feb 08.
Article En | MEDLINE | ID: mdl-38236416

Epoxyeicosatrienoic acids with anti-inflammatory effects are inactivated by soluble epoxide hydrolase (sEH). Both sEH and histone deacetylase 6 (HDAC6) inhibitors are being developed as neuropathic pain relieving agents. Based on the structural similarity, we designed a new group of compounds with inhibition of both HDAC6 and sEH and obtained compound M9. M9 exhibits selective inhibition of HDAC6 over class I HDACs in cells. M9 shows good microsomal stability, moderate plasma protein binding rate, and oral bioavailability. M9 exhibited a strong analgesic effect in vivo, and its analgesic tolerance was better than gabapentin. M9 improved the survival time of mice treated with lipopolysaccharide (LPS) and reversed the levels of inflammatory factors induced by LPS in mouse plasma. M9 represents the first sEH/HDAC6 dual inhibitors with in vivo antineuropathic pain and anti-inflammation.


Lipopolysaccharides , Neuralgia , Animals , Mice , Analgesics/pharmacology , Analgesics/therapeutic use , Epoxide Hydrolases/antagonists & inhibitors , Gabapentin , Histone Deacetylase 6/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Neuralgia/chemically induced , Neuralgia/drug therapy , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology
3.
J Clin Neurosci ; 120: 196-203, 2024 Feb.
Article En | MEDLINE | ID: mdl-38277995

BACKGROUND: We retrospectively analyzed the effects of low-dose bevacizumab (BEV) combined with temozolomide (TMZ) on health-related quality of life (HRQL) in patients with recurrent high-grade glioma (rHGG). METHODS: A total of 129 patients with rHGG were included in this study. Patients were divided into a combination group and TMZ group based on the treatment they received. The Quality of Life Questionnaire Core 30 (QLQ-C30) and EORTC Brain Cancer Module (QLQ-BN20) were used to evaluate HRQL in all patients before and after treatment. Categorical variables were compared using the chi-squared test. The data for all continuous variables were first tested for a normal distribution. If the data conformed to a normal distribution, a T test was used for comparison. If the data did not conform to a normal distribution, the rank-sum test was used. RESULTS: There were differences in PFS and PFS-6 between the BEV + TMZ and TMZ groups (P<0.05). However, there was no difference in the OS between the two groups (P>0.05). The BEV + TMZ group performed better than the TMZ group in both the QLQ-C30 and QLQ-BN20. In addition, the KPS score was higher in the BEV + TMZ group than in the TMZ group. Steroid doses given were lower in the BEV + TMZ group than in the TMZ group (P < 0.05). CONCLUSIONS: Low-dose BEV + TMZ can relieve the clinical symptoms of rHGG patients, reduce their steroid dose, improve HRQL, and prolong PFS, but does not bear any benefit on OS.


Brain Neoplasms , Glioblastoma , Glioma , Humans , Bevacizumab/therapeutic use , Retrospective Studies , Quality of Life , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Glioma/drug therapy , Temozolomide/therapeutic use , Brain Neoplasms/drug therapy , Steroids/therapeutic use , Glioblastoma/therapy
4.
Bioorg Chem ; 143: 107064, 2024 Feb.
Article En | MEDLINE | ID: mdl-38150937

Alzheimer's disease, the commonest cause of dementia, is a growing global health concern with huge implications for individuals and society. Stroke has still been a significant challenge in clinics for a long time, which is the second leading cause of death in the world, especially ischemic stroke. Both Alzheimer's disease and stroke are closely related to oxidative stress and HIF-1 signaling pathways in nerve cells. Herein, we describe our structure-based design, synthesis, and biological evaluation of a new class of 8-biaryl-2,2-dimethylbenzopyranamide derivatives as natural product derivatives. Our efforts have resulted in the discovery of highly potent neuroprotective agents, as exemplified by compound D13 as a HIF-1α inhibitor, which significant improvement in the behavior of Alzheimer's disease mice and shows great potential improvement of brain infarct volume in pMCAO model rats, improves the increase of blood-brain barrier permeability after cerebral ischemia in rats, neuroprotective effect, reduce the level of apoptotic cells in rats after cerebral ischemia, better than Edaravone.


Alzheimer Disease , Benzopyrans , Brain Ischemia , Ischemic Stroke , Neuroprotective Agents , Stroke , Animals , Mice , Rats , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Blood-Brain Barrier , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Ischemic Stroke/drug therapy , Ischemic Stroke/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/metabolism , Stroke/drug therapy , Stroke/metabolism , Benzopyrans/chemistry , Benzopyrans/pharmacology
5.
J Med Chem ; 66(8): 5685-5702, 2023 04 27.
Article En | MEDLINE | ID: mdl-37021456

In recent years, it has been proposed that G9a/EZH2 dual inhibition is a promising cancer treatment strategy. Herein, we present the discovery of G9a/EZH2 dual inhibitors that merge the pharmacophores of G9a and EZH2 inhibitors. Among them, the most promising compound 15h displayed potent inhibitory activities against G9a (IC50 = 2.90 ± 0.05 nM) and EZH2 (IC50 = 4.35 ± 0.02 nM), superior antiproliferative profiles against RD (CC50 = 19.63 ± 0.18 µM) and SW982 (CC50 = 19.91 ± 0.50 µM) cell lines. In vivo, 15h achieved significant antitumor efficacy in a xenograft mouse model of human rhabdoid tumor with a tumor growth inhibitory rate of 86.6% without causing observable toxic effects. The on-target activity assays illustrated that compound 15h can inhibit tumor growth by specifically inhibiting EZH2 and G9a. Therefore, 15h is a potential anticancer drug candidate for the treatment of malignant rhabdoid tumor.


Antineoplastic Agents , Rhabdoid Tumor , Humans , Mice , Animals , Rhabdoid Tumor/drug therapy , Lysine/pharmacology , Histone-Lysine N-Methyltransferase , Enzyme Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein
6.
J Med Chem ; 66(4): 2979-3009, 2023 02 23.
Article En | MEDLINE | ID: mdl-36689364

Soluble epoxide hydrolase (sEH) has been identified as an attractive target for anti-inflammatory drug design in recent years. Picomolar level compound G1 against sEH was obtained by introducing the hydrophilic group homopiperazine and hydrophobic fragment propionyl onto the structure of lead compound A. G1 showed good microsomal stability, a moderate plasma protein binding rate, and good oral bioavailability and was well tolerated in rats. G1 has significant analgesic effects on CFA-induced AIA mice, ameliorated the pancreatic injury in acute pancreatitis induced by l-arginine, reversed pancreatic injury, edema, and neutrophil infiltration, and increased the survival time of C57BL/6 mice in a lipopolysaccharide (LPS)-induced sepsis model. Moreover the expression levels of sEH, COX-2, NOS-2, vascular cell adhesion molecule (VCAM), IL-6, MCP-5, and tumor necrosis factor α (TNF-α) were measured by Western blot or enzyme-linked immunosorbent assay (ELISA), with varying degrees of decrease. These results suggested that G1 is a drug candidate worthy of further evaluation for the treatment of inflammation-induced diseases such as arthritis, acute pancreatitis, and sepsis.


Epoxide Hydrolases , Pancreatitis , Mice , Rats , Animals , Pancreatitis/drug therapy , Acute Disease , Mice, Inbred C57BL , Anti-Inflammatory Agents/therapeutic use
7.
Proc (Bayl Univ Med Cent) ; 34(6): 726-728, 2021.
Article En | MEDLINE | ID: mdl-34733003

The conventional treatment for the resection of cervical spinal tumors comprises anterior, posterior, and combined surgical approaches. However, these approaches have certain limitations when tumors invade the vertebrae, vertebral artery, or spinal nerves. Herein, we report an interesting case where a 45-year-old patient was admitted for neck pain. An invasive cervical spinal tumor was discovered and approached in two stages: stage 1 was open biopsy with posterior instrumentation, which was followed by stage 2 with an anterolateral approach for definitive surgical resection. A series of preoperative tests including angiography as well as a balloon occlusion test of the vertebral artery facilitated stage 2 surgical planning for gross total resection of the tumor while minimizing surgical complications.

8.
J Cell Biochem ; 120(8): 13985-13993, 2019 08.
Article En | MEDLINE | ID: mdl-30957285

Matrix attachment regions (MARs) can enhance transgene expression levels and maintain stability. However, the consensus sequence from MARs and its functional analysis remains to be examined. Here, we assessed a possible consensus sequence from MARs and assessed its activity in stably transfected Chinese hamster ovary (CHO) cells. First, we analyzed the effects of 10 MARs on transfected CHO cells and then analyzed the consensus motifs from these MARs using a bioinformatics method. The consensus sequence was synthesized and cloned upstream or downstream of the eukaryotic vector. The constructs were transfected into CHO cells and the expression levels and stability of enhanced green fluorescent protein were detected by flow cytometry. The results indicated that eight of the ten MARs increased transgene expression in transfected CHO cells. Three consensus motifs were found after bioinformatics analyses. The consensus sequence tandemly enhanced transgene expression when it was inserted into the eukaryotic expression vector; the effect of the addition upstream was stronger than that downstream. Thus, we found a MAR consensus sequence that may regulate the MAR-mediated increase in transgene expression.


Consensus Sequence/genetics , Matrix Attachment Regions/genetics , Transfection , Animals , Base Sequence , CHO Cells , Cricetinae , Cricetulus , Gene Dosage , Gene Expression , Green Fluorescent Proteins/metabolism , Humans , Recombinant Proteins/metabolism , Transgenes
9.
Brain Res ; 1692: 154-162, 2018 08 01.
Article En | MEDLINE | ID: mdl-29782850

Histone deacetylase 1 (HDAC1) plays a crucial role in cancer progression and development. This enzyme has been confirmed to be a key regulator of tumor biology functions, such as tumor cell proliferation, migration and invasion. However, HDAC1 expression in glioma remains controversial, and its specific function and molecular mechanism in glioblastoma is poorly understood. In this study, our findings demonstrated that protein and mRNA levels of HDAC1 were increased in glioma cell lines and glioma tissues compared to normal glial cell lines and non-neoplastic brain tissues, respectively. Furthermore, HDAC1 knockdown cells displayed decreased proliferation and invasion capabilities, whereas HDAC1 overexpressing glioblastoma cells displayed more proliferation and invasion capabilities in vitro. These novel outcomes suggested that knockdown of HDAC1 possibly suppressed the expression of phosphorylated AKT (p-AKT) and phosphorylated ERK (p-ERK) proteins, while overexpression of HDAC1 significantly increased p-AKT and p-ERK protein in glioblastoma cells. In addition, knockdown of HDAC1 repressed subcutaneous tumor growth in vivo, and led to down-regulation of p-AKT and p-ERK protein in U87 MG xenograft tumors. For the first time, we have demonstrated that HDAC1 promotes proliferation and invasion in glioblastoma cells by activating PI3K/AKT and MEK/ERK signaling pathways in vitro and in vivo. These results suggest that HDAC1 may be a novel biomarker and potential therapeutic target in glioblastoma.


Brain Neoplasms/pathology , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/pathology , Histone Deacetylase 1/metabolism , Signal Transduction/physiology , Adolescent , Adult , Aged , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase 1/genetics , Humans , Ki-67 Antigen/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred BALB C , Middle Aged , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Transfection , Young Adult
10.
Neurochem Res ; 42(8): 2191-2207, 2017 Aug.
Article En | MEDLINE | ID: mdl-28397068

Transplantation of human amniotic mesenchymal stem cells (hAM-MSCs) seems to be a promising strategy for the treatment of neurodegenerative disorders, including Alzheimer's disease (AD). However, the clinical therapeutic effects of hAM-MSCs and their mechanisms of action in AD remain to be determined. Here, we used amyloid precursor protein (APP) and presenilin1 (PS1) double-transgenic mice to evaluate the effects of hAM-MSC transplantation on AD-related neuropathology and cognitive dysfunction. We found that hAM-MSC transplantation into the hippocampus dramatically reduced amyloid-ß peptide (Aß) deposition and rescued spatial learning and memory deficits in APP/PS1 mice. Interestingly, these effects were associated with increasing in Aß-degrading factors, elevations in activated microglia, and the modulation of neuroinflammation. Furthermore, enhanced hippocampal neurogenesis in the subgranular zone (SGZ) of the dentate gyrus (DG) and enhanced synaptic plasticity following hAM-MSC treatment could be another important factor in reversing the cognitive decline in APP/PS1 mice. Instead, the mechanism underlying the improved cognition apparently involves a robust increase in hippocampal synaptic density and neurogenesis that is mediated by brain-derived neurotrophic factor (BDNF). In conclusion, our data suggest that hAM-MSCs may be a new and effective therapy for the treatment of AD.


Amniotic Fluid/physiology , Amyloid beta-Peptides/metabolism , Memory Disorders/metabolism , Memory Disorders/therapy , Memory/physiology , Mesenchymal Stem Cell Transplantation/trends , Amniotic Fluid/cytology , Amyloid beta-Protein Precursor/genetics , Animals , Cells, Cultured , Male , Maze Learning/physiology , Memory Disorders/genetics , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/genetics
11.
Neurochem Res ; 41(10): 2708-2718, 2016 Oct.
Article En | MEDLINE | ID: mdl-27351200

Human amniotic membrane mesenchymal stem cells (hAMSCs) are considered ideal candidate stem cells for cell-based therapy. In this study, we assessed whether hAMSCs transplantation promotes neurological functional recovery in rats after traumatic spinal cord injury (SCI). In addition, the potential mechanisms underlying the possible benefits of this therapy were investigated. Female Sprague-Dawley rats were subjected to SCI using a weight drop device and then hAMSCs, or phosphate-buffered saline (PBS) were immediately injected into the contused dorsal spinal cord at 2 mm rostral and 2 mm caudal to the injury site. Our results indicated that transplanted hAMSCs migrated in the host spinal cord without differentiating into neuronal or glial cells. Compared with the control group, hAMSCs transplantation significantly decreased the numbers of ED1+ macrophages/microglia and caspase-3+ cells. In addition, hAMSCs transplantation significantly increased the levels of brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF) in the injured spinal cord, and promoted both angiogenesis and axonal regeneration. These effects were associated with significantly improved neurobehavioral recovery in the hAMSCs transplantation group. These results show that transplantation of hAMSCs provides neuroprotective effects in rats after SCI, and could be candidate stem cells for the treatment of SCI.


Cell Movement/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Spinal Cord Injuries/therapy , Animals , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Female , Humans , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Microglia/metabolism , Neuroglia/metabolism , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord Injuries/physiopathology , Vascular Endothelial Growth Factor A/metabolism
12.
Oncol Lett ; 12(1): 125-131, 2016 Jul.
Article En | MEDLINE | ID: mdl-27347112

It has been reported previously that the expression of glucose transporter member 3 (GLUT3) is increased in malignant glioma cells compared with normal glial cells. However, the regulating mechanism that causes this phenomenon remains unknown. The present study investigated the regulating role of transcription factor specific protein 1 (Sp1) in GLUT3 expression in a human glioma cell line. In the present study, Sp1 was identified to directly bind to the GLUT3 5'-untranslated region in human glioma U251 cells. Small interfering RNA- and the Sp1-inhibitor mithramycin A-mediated Sp1 knockdown experiments revealed that Sp1 depletion decreased glucose uptake and inhibited cell growth and invasion of U251 cells by downregulating GLUT3 expression. Therefore Sp1 is an important positive regulator for the expression of GLUT3 in human glioma cells, and may explain the overexpression of GLUT3 in U251 cells. These results suggest that Sp1 may have a role in glioma treatment.

13.
PLoS One ; 10(9): e0137211, 2015.
Article En | MEDLINE | ID: mdl-26352672

Induced neural stem cells (iNSCs) can be directly transdifferentiated from somatic cells. One potential clinical application of the iNSCs is for nerve regeneration. However, it is unknown whether iNSCs function in disease models. We produced transdifferentiated iNSCs by conditional overexpressing Oct4, Sox2, Klf4, c-Mycin mouse embryonic fibroblasts. They expanded readily in vitro and expressed NSC mRNA profile and protein markers. These iNSCs differentiated into mature astrocytes, neurons and oligodendrocytes in vitro. Importantly, they reduced lesion size, promoted the recovery of motor and sensory function as well as metabolism status in middle cerebral artery stroke rats. These iNSCs secreted nerve growth factors, which was associated with observed protection of neurons from apoptosis. Furthermore, iNSCs migrated to and passed through the lesion in the cerebral cortex, where Tuj1+ neurons were detected. These findings have revealed the function of transdifferentiated iNSCs in vivo, and thus provide experimental evidence to support the development of personalized regenerative therapy for CNS diseases by using genetically engineered autologous somatic cells.


Cell Transdifferentiation , Cerebral Cortex/growth & development , Infarction, Middle Cerebral Artery/therapy , Nerve Regeneration , Neural Stem Cells/transplantation , Animals , Astrocytes/transplantation , Cell Differentiation/genetics , Cerebral Cortex/pathology , Humans , Induced Pluripotent Stem Cells/transplantation , Kruppel-Like Factor 4 , Mice , Neural Stem Cells/cytology , Neurons/transplantation , Oligodendroglia/transplantation , Rats
14.
Neurochem Res ; 40(8): 1583-92, 2015 Aug.
Article En | MEDLINE | ID: mdl-26068144

Alzheimer's disease (AD) is associated with the inflammatory response in response to amyloid ß-peptide (Aß). Previous studies have suggested that paeoniflorin (PF) shows anti-inflammatory and neuroprotective effects in inflammation-related diseases. However, the impacts of PF on AD have not been investigated. In the present study, we showed that a 4-week treatment with PF could significantly inhibit Aß burden, Aß-induced over activation of astrocytes and microglia, downregulation of proinflammatory cytokines, and upregulation of anti-inflammatory cytokines in the brain. In addition, we demonstrated that chronic treatment with PF inhibited the activation of glycogen synthase kinase 3ß (GSK-3ß) and reversed neuroinflammtory-induced activation of nuclear factor-kappa B (NF-κB) signaling pathways. Moreover, PF exerted inhibitory effects on NALP3 inflammasome, caspase-1, and IL-1ß. Collectively, in the present study, we demonstrated that PF exhibits neuroprotective effects in amyloid precursor protein (APP) and presenilin 1 (PS1) double-transgenic (APP/PS1) mice via inhibiting neuroinflammation mediated by the GSK-3ß and NF-κB signaling pathways and nucleotide-binding domain-like receptor protein 3 inflammasome. Thus, these results suggest that PF might be useful to intervene in development or progression of neurodegeneration in AD through its anti-inflammatory and anti-amyloidogenic effects.


Alzheimer Disease/drug therapy , Disease Models, Animal , Glucosides/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Monoterpenes/therapeutic use , Paeonia , Plaque, Amyloid/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/metabolism , Male , Memory Disorders/drug therapy , Memory Disorders/genetics , Memory Disorders/metabolism , Mice , Mice, Transgenic , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Presenilin-1/genetics
15.
J Alzheimers Dis ; 46(4): 863-76, 2015.
Article En | MEDLINE | ID: mdl-25854934

Lamotrigine (LTG), a broad-spectrum anti-epileptic drug widely used in treatment for seizures, shows potential efficacy in Alzheimer's disease (AD) therapy. Chronic LTG treatment rescues the suppressed long-term potentiation, loss of spines and cognitive deficits in AßPP/PS1 mice, known to overexpress a chimeric mouse/human mutant amyloid-ß protein precursor (AßPP) and a mutant human presenilin 1 (PS1). These changes are accompanied by reduction of amyloid-ß (Aß) plaques density and of levels of ß-C-terminal fragment of AßPP (ß-CTF), a fragment of AßPP cleaved by ß-secretase. These results suggest LTG treatment reduces Aß production, possibly through modulation of cleavage of AßPP by ß-secretase. However, the underlying mechanisms still remain unclear. In this study, decreased protein levels, but not mRNA levels of ß-site AßPP-cleaving enzyme 1 (BACE1), were observed in cultured HEK293 cells and the brains of AßPP/PS1 transgenic mice upon LTG treatment. Moreover, LTG treatment suppressed mammalian target of rapamycin (mTOR) signaling, while enhancing activation of cAMP response element binding protein (CREB), two signaling pathways essential for autophagy induction. LTG treatment increased the numbers of LC3-GFP + puncta and LC3-II levels in HEK293 cells, indicating an induction of autophagy. The downregulation of BACE1 by LTG treatment was prevented by the autophagy inhibitor 3-Methyladenine. Therefore, this study shows that LTG treatment reduces the protein levels of BACE1 through activation of autophagy, possibly via inhibition of mTOR signaling and activation of CREB.


Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Autophagy/drug effects , Triazines/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Autophagy/genetics , CHO Cells , CREB-Binding Protein/metabolism , Cricetulus , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Humans , Lamotrigine , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Presenilin-1/genetics , Triazines/therapeutic use
16.
Exp Mol Pathol ; 98(2): 192-9, 2015 Apr.
Article En | MEDLINE | ID: mdl-25617528

Glioma is an aggressive tumor with poor prognosis. Identification of precise prognostic marker and effective therapeutic target is important in the treatment of glioma. HTATIP2 is a novel tumor suppressor gene, which is frequently silenced by epigenetic mechanisms in many caners. However, the expression of HTATIP2 and how it is regulated in glioma are unknown. Hence, we assessed whether loss of HTATIP2 expression occurs in glioma, and, if so, what is the mechanism of such loss. We found that HTATIP2 expression was absent or diminished in primary gliomas compared with normal brain tissue. In vitro experiments showed that HTATIP2 expression could be restored via 5-aza-2'deoxycytidine treatment in U87 and U251 cell lines. Methyl-specific PCR indicated that the two cell lines and 60% primary gliomas carried aberrant methylated HTATIP2 alleles while normal brain tissue did not. Pyrosequencing confirmed these results and showed a higher density of methylation in the minimal promoter element, which contains four Sp1 binding sites in primary gliomas, than in normal brain tissue. Finally, we found that the overall survival was significantly higher in patients with positive HTATIP2 expression than those with loss of HTATIP2 expression. Overexpression of HTATIP2 inhibited glioma proliferation and growth in vitro. Taken together, the present study showed that loss of HTATIP2 expression was a frequent event in glioma and is associated with poor prognosis. Promoter methylation may be an underlying mechanism.


Acetyltransferases/biosynthesis , Brain Neoplasms/genetics , DNA Methylation/genetics , Glioma/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/biosynthesis , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Binding Sites , Brain Neoplasms/mortality , Cell Line, Tumor , Decitabine , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Glioma/mortality , HEK293 Cells , Humans , Male , Middle Aged , Prognosis , Sp1 Transcription Factor/metabolism
17.
Neurobiol Aging ; 35(12): 2713-2725, 2014 Dec.
Article En | MEDLINE | ID: mdl-25044076

Hyperactivity and its compensatory mechanisms may causally contribute to synaptic and cognitive deficits in Alzheimer's disease (AD). Blocking the overexcitation of the neural network, with levetiracetam (LEV), a sodium channel blocker applied in the treatment of epilepsy, prevented synaptic and cognitive deficits in human amyloid precursor protein (APP) transgenic mice. This study has brought the potential use of antiepileptic drugs (AEDs) in AD therapy. We showed that the chronic treatment with lamotrigine (LTG), a broad-spectrum AED, suppressed abnormal spike activity, prevented the loss of spines, synaptophysin immunoreactivity, and neurons, and thus attenuated the deficits in synaptic plasticity and learning and memory in APP and presenilin 1 (PS1) mice, which express human mutant APP and PS1. In contrast with LEV, which failed to reduce the generation of amyloid ß, the chronic LTG treatment reduced the cleavage of APP by ß-secretase and thus the numbers and the size of amyloid plaques in the brains of APP and PS1 mice. Moreover, the levels of brain-derived neurotrophic growth factor (BDNF) and nerve growth factor (NGF) were enhanced in the brains of APP and PS1 mice by the chronic LTG treatment. Therefore, these observations demonstrate that LTG attenuates AD pathology through multiple mechanisms, including modulation of abnormal network activity, reduction of the generation of amyloid beta and upregulation of BDNF and NGF.


Amyloid beta-Peptides/metabolism , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Brain/metabolism , Neuronal Plasticity/drug effects , Plaque, Amyloid/metabolism , Triazines/pharmacology , Triazines/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Humans , Lamotrigine , Learning/drug effects , Memory/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Nerve Growth Factor/metabolism , Up-Regulation/drug effects
18.
Plant Cell Physiol ; 54(6): 944-59, 2013 Jun.
Article En | MEDLINE | ID: mdl-23543751

Late embryogenesis abundant (LEA) proteins accumulate to high levels during the late stage of seed maturation and in response to water deficit, and are involved in protecting higher plants from damage caused by environmental stresses, especially drought. In the present study, a novel maize (Zea mays L.) group 3 LEA gene, ZmLEA3, was identified and later characterized using transgenic tobacco plants to investigate its functions in abiotic and biotic stresses. Transcript accumulation demonstrated that ZmLEA3 was induced in leaves by high salinity, low temperature, osmotic and oxidative stress as well as by signaling molecules such as ABA, salicylic acid (SA) and methyl jasmonate (MeJA). The transcript of ZmLEA3 could also be induced by pathogens [Pseudomonas syringae pv. tomato DC3000 (pst dc3000)]. ZmLEA3 is located in the cytosol and the nucles. Further study indicated that the ZmLEA3 protein could bind Mn(2+), Fe(3+), Cu(2+) and Zn(2+). Overexpression of ZmLEA3 in transgenic tobacco (Nicotiana tabacum) and yeast (GS115) conferred tolerance to osmotic and oxidative stresses. Interestingly, we also found that overexpression of ZmLEA3 in transgenic tobacco increased the hypersensitive cell death triggered by pst dc3000 and enhanced the expression of PR1a, PR2 and PR4 when compared with the wild type. Thus, we proposed that the ZmLEA3 protein plays a role in protecting plants from damage by protecting protein structure and binding metals under osmotic and oxidative stresses. In addition, ZmLEA3 may also enhance transgenic plant tolerance to biotic stress.


Plant Proteins/metabolism , Stress, Physiological , Zea mays/physiology , Adaptation, Physiological/genetics , Amino Acid Sequence , Chromatography, Affinity , Computational Biology , Gene Expression Regulation, Plant , Green Fluorescent Proteins/metabolism , L-Lactate Dehydrogenase/metabolism , Metals/metabolism , Molecular Sequence Data , Osmotic Pressure , Oxidation-Reduction , Oxidative Stress/genetics , Plant Leaves/genetics , Plant Leaves/physiology , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/isolation & purification , Plants, Genetically Modified , Protein Binding/genetics , Protein Transport , Pseudomonas syringae/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Stress, Physiological/genetics , Subcellular Fractions/metabolism , Nicotiana/genetics , Nicotiana/immunology , Nicotiana/microbiology , Zea mays/genetics
19.
Neurochem Res ; 38(5): 1022-33, 2013 May.
Article En | MEDLINE | ID: mdl-23475428

Although human amnion derived mesenchymal stem cells (AMSC) are a promising source of stem cells, their therapeutic potential for traumatic brain injury (TBI) has not been widely investigated. In this study, we evaluated the therapeutic potential of AMSC using a rat TBI model. AMSC were isolated from human amniotic membrane and characterized by flow cytometry. After induction, AMSC differentiated in vitro into neural stem-like cells (AM-NSC) that expressed higher levels of the neural stem cell markers, nestin, sox2 and musashi, in comparison to undifferentiated AMSC. Interestingly, the neurotrophic factors, brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin 3 (NT-3), glial cell derived neurotrophic factor (GDNF) and ciliary neurotrophic factor (CNTF) were markedly upregulated after neural stem cell induction. Following transplantation in a rat TBI model, significant improvements in neurological function, brain tissue morphology, and higher levels of BDNF, NGF, NT-3, GDNF and CNTF, were observed in the AM-NSC group compared with the AMSC and Matrigel groups. However, few grafted cells survived with minimal differentiation into neural-like cells. Together, our results suggest that transplantation of AM-NSC promotes functional rehabilitation of rats with TBI, with enhanced expression of neurotrophic factors a likely mechanistic pathway.


Amnion/cytology , Brain Injuries/therapy , Neural Stem Cells/cytology , Animals , Base Sequence , Cell Differentiation , DNA Primers , Female , Humans , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
20.
Neurosci Lett ; 525(2): 129-34, 2012 Sep 13.
Article En | MEDLINE | ID: mdl-22902990

Tenascin-R (TN-R) is a neural specific protein and an important molecule involved in inhibition of axonal regeneration after spinal cord injury (SCI). Here we report on rabbit-derived TN-R polyclonal antibody, which acts as a TN-R antagonist with high titer and high specificity, promoted neurite outgrowth and sprouting of rat cortical neurons cultured on the inhibitory TN-R substrate in vitro. When locally administered into the lesion sites of rats received spinal cord dorsal hemisection, these TN-R antibodies could significantly decrease RhoA activation and improve functional recovery from corticospinal tract (CST) transection. Thus, passive immunotherapy with specific TN-R antagonist may represent a promising repair strategy following acute SCI.


Antibodies/pharmacology , Axons/drug effects , Spinal Cord Injuries/therapy , Tenascin/antagonists & inhibitors , Animals , Animals, Newborn , Antibodies/therapeutic use , Axons/physiology , Cells, Cultured , Female , Hindlimb/physiopathology , Immunization, Passive , Motor Activity , Nerve Regeneration , Neurites/drug effects , Neurites/physiology , Rabbits , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/immunology , Spinal Cord Injuries/physiopathology , Tenascin/immunology , rhoA GTP-Binding Protein/metabolism
...