Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Transl Med ; 22(1): 436, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720350

ABSTRACT

BACKGROUND: Subarachnoid hemorrhage (SAH) represents a form of cerebrovascular event characterized by a notable mortality and morbidity rate. Fibroblast growth factor 21 (FGF21), a versatile hormone predominantly synthesized by the hepatic tissue, has emerged as a promising neuroprotective agent. Nevertheless, the precise impacts and underlying mechanisms of FGF21 in the context of SAH remain enigmatic. METHODS: To elucidate the role of FGF21 in inhibiting the microglial cGAS-STING pathway and providing protection against SAH-induced cerebral injury, a series of cellular and molecular techniques, including western blot analysis, real-time polymerase chain reaction, immunohistochemistry, RNA sequencing, and behavioral assays, were employed. RESULTS: Administration of recombinant fibroblast growth factor 21 (rFGF21) effectively mitigated neural apoptosis, improved cerebral edema, and attenuated neurological impairments post-SAH. Transcriptomic analysis revealed that SAH triggered the upregulation of numerous genes linked to innate immunity, particularly those involved in the type I interferon (IFN-I) pathway and microglial function, which were notably suppressed upon adjunctive rFGF21 treatment. Mechanistically, rFGF21 intervention facilitated mitophagy in an AMP-activated protein kinase (AMPK)-dependent manner, thereby preventing mitochondrial DNA (mtDNA) release into the cytoplasm and dampening the activation of the DNA-sensing cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway. Conditional knockout of STING in microglia markedly ameliorated the inflammatory response and mitigated secondary brain injuries post-SAH. CONCLUSION: Our results present the initial evidence that FGF21 confers a protective effect against neuroinflammation-associated brain damage subsequent to SAH. Mechanistically, we have elucidated a novel pathway by which FGF21 exerts this neuroprotection through inhibition of the cGAS-STING signaling cascade.


Subject(s)
Fibroblast Growth Factors , Membrane Proteins , Mice, Inbred C57BL , Mitophagy , Neuroinflammatory Diseases , Nucleotidyltransferases , Signal Transduction , Subarachnoid Hemorrhage , Animals , Membrane Proteins/metabolism , Fibroblast Growth Factors/metabolism , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/pathology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/etiology , Mitophagy/drug effects , Signal Transduction/drug effects , Nucleotidyltransferases/metabolism , Male , Mice , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Microglia/metabolism , Microglia/pathology , Microglia/drug effects , Apoptosis/drug effects
2.
J Parkinsons Dis ; 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38640168

ABSTRACT

Background: Multiple system atrophy (MSA) is a disease with diverse symptoms and the commonly used classifications, MSA-P and MSA-C, do not cover all the different symptoms seen in MSA patients. Additionally, these classifications do not provide information about how the disease progresses over time or the expected outcome for patients. Objective: To explore clinical subtypes of MSA with a natural disease course through a data-driven approach to assist in the diagnosis and treatment of MSA. Methods: We followed 122 cases of MSA collected from 3 hospitals for 3 years. Demographic characteristics, age of onset, clinical signs, scale assessment scores, and auxiliary examination were collected. Age at onset; time from onset to assisted ambulation; and UMSARS I, II, and IV, COMPASS-31, ICARS, and UPDRS III scores were selected as clustering elements. K-means, partitioning around medoids, and self-organizing maps were used to analyze the clusters. Results: The results of all three clustering methods supported the classification of three MSA subtypes: The aggressive progression subtype (MSA-AP), characterized by mid-to-late onset, rapid progression and severe clinical symptoms; the typical subtype (MSA-T), characterized by mid-to-late onset, moderate progression and moderate severity of clinical symptoms; and the early-onset slow progression subtype (MSA-ESP), characterized by early-to-mid onset, slow progression and mild clinical symptoms. Conclusions: We divided MSA into three subtypes and summarized the characteristics of each subtype. According to the clustering results, MSA patients were divided into three completely different types according to the severity of symptoms, the speed of disease progression, and the age of onset.

3.
Oncol Rep ; 46(6)2021 12.
Article in English | MEDLINE | ID: mdl-34664678

ABSTRACT

Emerging evidence has shown that microRNA (miR)­497 serves pivotal roles in tumorigenesis, cancer progression, metastasis and chemotherapy resistance in several types of cancer. In the present study, the expression and biological functions of miR­497 host gene (MIR497HG) were investigated in glioma tissue. The expression levels of miR­497 and MIR497HG were measured in glioma, adjacent non­cancerous and normal brain tissue and their association with the prognosis of patients with glioma were analyzed. The biological roles of miR­497 and MIR497HG were investigated in glioma cell lines. In addition, bioinformatics analysis, luciferase reporter assay and functional experiments were performed to identify and validate the downstream targets of miR­497 or MIR497HG. The expression levels of miR­497 and MIR497HG were downregulated in glioma tissue and cell lines compared with those in adjacent non­cancerous and normal brain tissue and normal human cortical neuron cell line. Patients with low miR­497 or MIR497HG expression levels exhibited a poor prognostic outcome. In addition, forced overexpression of miR­497 or MIR497HG significantly inhibited the proliferation and cell cycle progression of glioma cell lines. Furthermore, the results indicated that miR­497 and MIR497HG exerted their biological functions by direct targeting of cyclin E1 and miR­588/tumor suppressor candidate 1. In summary, the data indicated that miR­497 and MIR497HG served as tumor suppressors and may be used as potential therapeutic targets and prognostic biomarkers in glioma.


Subject(s)
Cell Proliferation/genetics , Cyclin E/genetics , Glioma/genetics , MicroRNAs/genetics , Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Adult , Animals , Brain Neoplasms/genetics , Carcinogenesis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Disease Models, Animal , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Nude , Middle Aged
4.
Medicine (Baltimore) ; 100(5): e24451, 2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33592894

ABSTRACT

RATIONALE: The Miller Fisher syndrome (MFS) is an acute polyradiculoneuritis regarded as an uncommon clinical variant of the Guillain-Barre syndrome (GBS). It is characterized by the clinical triad of ophthalmoplegia, ataxia, and areflexia. The diagnosis of MFS is based on clinical presentation, presence of albuminocytologic dissociation in the cerebrospinal fluid (CSF), and normal brain imaging results. The presence of anti-ganglioside antibodies (GQlb) in the serum is helpful for the diagnosis. A history of upper respiratory tract infection or diarrhea 3 days to 6 weeks before the onset of MFS is common. However, there are some patients with atypical manifestations who are difficult to diagnose. Here, we present an incomplete form of MFS where antibodies against GQ1b were detected in the serum following an Epstein Barr virus (EBV) infection. PATIENT CONCERNS: A 77-year-old Chinese woman was admitted to the hospital with acute diplopia and right blepharoptosis. She had a history of mild upper respiratory tract infection 2 weeks ago. In 1 week, the symptoms rapidly progressed into bilateral ophthalmoplegia and hyporeflexia of the limbs without ataxia. CSF analysis on the third day after onset was normal, without albuminocytologic dissociation. EBV immunoglobulin G (IgG) antibodies were detected in the CSF. GQ1b and GD1b IgG antibodies were positive in the serum and negative in the CSF. No responsible lesion was found on brain imaging examination. DIAGNOSES: In accordance with the progressive bilateral ophthalmoplegia and hyporeflexia, the history of upper respiratory tract infection, the detection of EBV and GQ1b antibodies, and the negative brain imaging examination, the diagnosis of MFS was confirmed. INTERVENTIONS: The patient was administered intravenous immunoglobulin for 5 days. OUTCOMES: She had a favorable outcome after treatment. At the 6-week follow-up, bilateral ocular movement limitation and tendon reflexes had recovered. LESSONS: The diagnosis of MFS can be challenging, especially when encountered with incomplete symptoms and normal CSF results. Attention should be paid to the presence of anti-GQ1b IgG antibodies when the clinical manifestations are incomplete. Furthermore, EBV primary infection could be associated with MFS and considered a potential causative agent.


Subject(s)
Epstein-Barr Virus Infections/complications , Gangliosides/immunology , Miller Fisher Syndrome/etiology , Miller Fisher Syndrome/immunology , Aged , Female , Humans , Immunoglobulins, Intravenous/therapeutic use , Miller Fisher Syndrome/diagnosis , Miller Fisher Syndrome/drug therapy
5.
Sci Rep ; 10(1): 4155, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32139705

ABSTRACT

Malignant gliomas are the most common tumor in central nervous system with poor prognosis. Due to the limitation of histological classification in earlier diagnosis and individualized medicine, it is necessary to combine the molecular signatures and the pathological characteristics of gliomas. Lots of microRNAs presented abnormal expression in gliomas and modulated gliomas development. Exploration the miRNAs profile is helpful for the diagnosis, therapy and prognosis of gliomas. It has been demonstrated that miR-144 plays important roles in solid tumors. However, the detail mechanisms remained unrevealed. In this study, we have demonstrated the level of miR-144 decreased in glioma tissues from patients, especially in gliomas with higher grades. MiR-144 was also validated have lower expression in glioma cell lines compared with cortical neuron cell by using qRT-PCR. The in vitro functional experiment indicated miR-144 improved gliomas progression through repressing proliferation, sensitizing to chemotherapeutics and inhibiting metastasis. We further identified fibroblast growth factor 7 (FGF7) and Caveolin 2 (CAV2) were target genes of miR-144 by luciferase reporter assay and western blotting. The mechanisms study suggested forced FGF7 expression elevated Akt activation and decreased reactive oxygen species (ROS) generation. The MTT and cell cycle assay indicated miR-144 suppressed glioma cells proliferation through modulating FGF mediated Akt signaling pathway. Meanwhile, miR-144 promoted Temozolomide (TMZ) induced apoptosis in glioma cells via increasing ROS production by using FACS. On the other hand, CAV2, as another target of miR-144, accelerated glioma cells migration and invasion via promoting glioma cells EMT progress. Retrieved expression of FGF7 or CAV2 rescued the proliferation and migration function mediated by miR-144. Furthermore, the in vivo experiments in PDX models displayed the anti-tumor function of miR-144, which could be retrieved by overexpression of FGF7 and CAV2. Taken together, these findings indicated miR-144 acted as a potential target against gliomas progression and uncovered a novel regulatory mechanism, which may provide a new therapeutic strategy and prognostic indicator for gliomas.


Subject(s)
Caveolin 2/metabolism , Fibroblast Growth Factor 7/metabolism , Glioma/metabolism , Glioma/pathology , MicroRNAs/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Caveolin 2/genetics , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Fibroblast Growth Factor 7/genetics , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Reactive Oxygen Species/metabolism
6.
J Mol Neurosci ; 70(3): 422-432, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31732924

ABSTRACT

Delayed neurologic sequelae (DNS) are among the most serious complications of carbon monoxide (CO) poisoning caused partly by elevated neuroinflammation. WIN 55,212-2, a non-selective agonist of cannabinoid receptors, has been demonstrated to have anti-inflammatory properties in various brain disorders. The anti-inflammatory action of WIN 55,212-2 is potentially associated with driving microglial M2 polarization. ST2 signaling is important in regulating inflammatory responses and microglial polarization. Therefore, we aimed to investigate the neuroprotective effect of WIN 55,212-2 on DNS after CO poisoning and elucidate its relationship with ST2-mediated microglial M2 polarization. The behavioral tests showed that treatment with WIN 55,212-2 significantly ameliorates the cognitive impairment induced by CO poisoning. This behavioral improvement was accompanied by reduced neuron loss, decreased production of pro-inflammatory cytokines, and a limited number of microglia in the hippocampus. Moreover, WIN 55,212-2 elevated the protein expression of IL-33 (the ligand of ST2) and ST2, increased the ratio of CD206-positive (M2 phenotype) and ST2-positive microglia, and augmented production of M2 microglia-associated cytokines in the hippocampus of CO-exposed rats. Furthermore, we observed that the WIN 55,212-2-mediated increases in ST2 protein expression, CD206-positive and ST2-positive microglia, and microglia-associated cytokines were blocked by the cannabinoid receptor 2 (CB2R) antagonist AM630 but not by the cannabinoid receptor 1 (CB1R) antagonist AM251. In contrast, the WIN 55,212-2-induced upregulation of the IL-33 protein expression was inhibited by AM251 but not by AM630. Altogether, these findings reveal cannabinoid receptors as promising therapeutic agents for CO poisoning and identify ST2 signaling-related microglial M2 polarization as a new mechanism of cannabinoid-induced neuroprotection.


Subject(s)
Benzoxazines/pharmacology , Cannabinoids/pharmacology , Carbon Monoxide Poisoning/drug therapy , Interleukin-1 Receptor-Like 1 Protein/metabolism , Microglia/drug effects , Morpholines/pharmacology , Naphthalenes/pharmacology , Neuroprotective Agents/pharmacology , Signal Transduction , Animals , Benzoxazines/therapeutic use , Cannabinoids/therapeutic use , Cognition , Interleukin-33/genetics , Interleukin-33/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Male , Mannose Receptor , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Microglia/metabolism , Morpholines/therapeutic use , Naphthalenes/therapeutic use , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Cannabinoid/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
7.
Aging (Albany NY) ; 11(22): 10266-10283, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31770107

ABSTRACT

OBJECTIVE: This study was conducted to elucidate the long non-coding RNA FOXD2-AS1 (lncRNA FOXD2-AS1) expression in glioma and its mechanism on the biological features of glioma cells and the drug resistance of temozolomide (TMZ). RESULTS: Highly expressed FOXD2-AS1 was found in glioma. There was more powerful chemotherapeutic resistance of TMZ resistant cell lines than that of the parent cell lines. Silence of FOXD2-AS1 suppressed proliferation and drug resistance and promoted apoptosis of drug-resistant glioma cells. Overexpressed FOXD2-AS1 presented an opposite trend. FOXD2-AS1 could be used as a competing endogenous RNA to adsorb miR-98-5p, thereby up-regulating CPEB4. CONCLUSION: Our study suggests that down-regulated FOXD2-AS1 repressed invasion, proliferation, migration and drug resistance of drug-resistant glioma cells while stimulating their apoptosis via increasing miR-98-5p and inhibiting CPEB4 expression. METHODS: FOXD2-AS1, microRNA-98-5p (miR-98-5p) and cytoplasmic polyadenylation element binding (CPEB4) expression in glioma tissues were tested. Expression of E-cadherin, N-cadherin and Vimentin in glioma cells were explored. A series of assays were conducted to detect the function of FOXD2-AS1 in migration, proliferation, apoptosis, and invasion of glioma cells. Changes in drug-resistance of cells under TMZ treatment were examined, and tumor formation in nude mice was performed to test the changes of drug resistance in vivo.


Subject(s)
Drug Resistance, Neoplasm/genetics , Glioma/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA-Binding Proteins/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Mice , Mice, Nude , Neoplasm Invasiveness/genetics
8.
Theranostics ; 9(20): 5956-5975, 2019.
Article in English | MEDLINE | ID: mdl-31534531

ABSTRACT

Mesenchymal stem cell-derived exosomes (MSC-Exo) have robust anti-inflammatory effects in the treatment of neurological diseases such as epilepsy, stroke, or traumatic brain injury. While astrocytes are thought to be mediators of these effects, their precise role remains poorly understood. To address this issue, we investigated the putative therapeutic effects and mechanism of MSC-Exo on inflammation-induced alterations in astrocytes. Methods: Lipopolysaccharide (LPS)-stimulated hippocampal astrocytes in primary culture were treated with MSC-Exo, which were also administered in pilocarpine-induced status epilepticus (SE) mice. Exosomal integration, reactive astrogliosis, inflammatory responses, calcium signaling, and mitochondrial membrane potentials (MMP) were monitored. To experimentally probe the molecular mechanism of MSC-Exo actions on the inflammation-induced astrocytic activation, we inhibited the nuclear factor erythroid-derived 2, like 2 (Nrf2, a key mediator in neuroinflammation and oxidative stress) by sgRNA (in vitro) or ML385 (Nrf2 inhibitor) in vivo. Results: MSC-Exo were incorporated into hippocampal astrocytes as well as attenuated reactive astrogliosis and inflammatory responses in vitro and in vivo. Also, MSC-Exo ameliorated LPS-induced aberrant calcium signaling and mitochondrial dysfunction in culture, and SE-induced learning and memory impairments in mice. Furthermore, the putative therapeutic effects of MSC-Exo on inflammation-induced astrocytic activation (e.g., reduced reactive astrogliosis, NF-κB deactivation) were weakened by Nrf2 inhibition. Conclusions: Our results show that MSC-Exo ameliorate inflammation-induced astrocyte alterations and that the Nrf2-NF-κB signaling pathway is involved in regulating astrocyte activation in mice. These data suggest the promising potential of MSC-Exo as a nanotherapeutic agent for the treatment of neurological diseases with hippocampal astrocyte alterations.


Subject(s)
Astrocytes/immunology , Exosomes/metabolism , Inflammation/immunology , Mesenchymal Stem Cells/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunochemistry , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Real-Time Polymerase Chain Reaction
9.
Front Neurol ; 10: 383, 2019.
Article in English | MEDLINE | ID: mdl-31040819

ABSTRACT

Objective: Ischemic stroke leads to cellular death and tissue damage by depriving the areas of glucose and oxygen supplies. The effective treatment of stroke remains a challenge for modern medicine. This study used an oxygen-glucose deprivation (OGD) model of human umbilical vein endothelial cells (HUVECs) to mimic ischemic injuries and explored the role and mechanism of intelectin-1. Methods: Intelectin-1 was transduced into the HUVECs using a lentiviral vector. The PI3K/Akt signaling was examined in intelectin-induced eNOS phosphorylation. The PI3K inhibitor LY294002 was dealed in HUVECs. Results: Our results demonstrated an increase in capillary density, decrease in apoptotic cells, and increase in HIF-1α protein expression following intelectin-1 treatment. Real-time PCR and Western blotting revealed the increased intelectin-1 expression alongside eNOS and Akt phosphorylation with enhanced bcl-2 expression under OGD. Capillary density decreased significantly after LY294002 treatment. Conclusion: These results suggest intelectin-1 promotes angiogenesis, inhibits oxidative stress and reduces apoptosis by stimulating the Akt-eNOS signaling pathway in response to ischemia in vitro.

10.
Neurol Sci ; 39(11): 1981-1984, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30109465

ABSTRACT

Anti-γ-aminobutyric acid-B (GABAB) receptor encephalitis is an autoimmune encephalitis associated with antibodies against neuronal cell surface antigens, which are primarily observed with small-cell lung cancer, melanoma, and thymoma. Here, we first report a case on the association between a relatively frequent cancer, gastric adenocarcinoma (GAC), and a rare GABAB receptor antibody limbic encephalitis. The patient was treated with immunotherapy and combined-drug chemotherapy, which were partially effective in terms of stabilizing the tumor and relieving neurological symptoms. This report indicates that, when patients present with GABAB receptor antibody limbic encephalitis, regular and broad screening for tumors including gastric adenocarcinoma should also be considered.


Subject(s)
Adenocarcinoma/complications , Encephalitis/complications , Encephalitis/immunology , Receptors, GABA-B/immunology , Stomach Neoplasms/complications , Adenocarcinoma/diagnostic imaging , Aged , Autoantibodies/blood , Brain/diagnostic imaging , Brain/pathology , Encephalitis/diagnostic imaging , Humans , Magnetic Resonance Imaging , Male , Receptors, GABA-B/genetics , Receptors, GABA-B/metabolism , Stomach Neoplasms/diagnostic imaging , Tomography Scanners, X-Ray Computed
11.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(1): 117-122, 2018 Jan 30.
Article in Chinese | MEDLINE | ID: mdl-33177029

ABSTRACT

OBJECTIVE: To explore the imaging biomarker for early diagnosis and disease course monitoring of Parkinson disease (PD) in arterial spin labeling (ASL) technique. METHODS: Between July, 2014 and May, 2017, 23 patients with PD underwent magnetic resonance imaging (MRI) and ASL examinations in our hospital, including 13 in the early stage and 10 in advanced stages. Voxel-based analysis (VBA) was used to observe the regional cerebral blood flow (rCBF) characteristics in PD patients in different stages and three-dimensional continuous arterial spin labeling (3D CASL) was used to analyze the mean cerebral blood flow (mCBF). RESULTS: No significant difference was found in mCBF among PD patients in the early stage, patients in advanced stages and normal control subjects (P=0.30). Compared with the normal control group, the patients with early-stage PD had decreased rCBF in resting state mainly in the right superior occipital gyrus and the right superior frontal gyrus as revealed by VBA (P < 0.001); the patients with advanced PD showed decreased rCBF mainly in the left precentral gyrus and the postcentral gyrus (P < 0.001). The patients with advanced PD exhibited lowered rCBF in the right substantia nigra and the bilateral corpus callosum as compared with the early-stage patients (P < 0.001). CONCLUSIONS: VBA of ASL reveals rCBF alterations in association with the disease progression in PD patients, suggesting that this technique might provide assistance in identification of potential markers for early PD diagnosis and for monitoring the disease course.

12.
RSC Adv ; 8(61): 35031-35041, 2018 Oct 10.
Article in English | MEDLINE | ID: mdl-35547044

ABSTRACT

OBJECTIVE: To investigate the role of FOXO1 and miR-183-96-182 clusters in ox-LDL induced endothelial cell apoptosis. METHODS: FOXO1 overexpression (OE) and knockdown (KD) as well as AKT1 OE in human umbilical vein endothelial cells (HUVECs) and human aortic endothelial cells (HAECs) were achieved by lentiviral transduction. Upregulation of miR-183-5p, miR-182-5p or miR-96-5p was mimicked by agomir treatment. FOXO1 gene transcription was monitored by FOXO1 promotor reporter assay. Cell apoptosis in culture was monitored by TiterTACS in situ detection. Regulation of FOXO1 gene expression by an miRNA targeting mechanism was monitored by AGO2-RNA immunoprecipitation assay. RESULTS: FOXO1 mRNA and protein expression levels in ox-LDL treated HUVECs or HAECs were significantly upregulated due to transcriptional and miRNA targeting mechanisms. MiR-183-5p, miR-182-5p and miR-96-5p expression levels in HUVECs or HAECs were significantly reduced by ox-LDL treatment, the overexpression of which by agomir treatment partially reduced the FOXO1 mRNA/protein expression levels and cell apoptosis which was upregulated by ox-LDL treatment. FOXO1 overexpression antagonized the effect of the agomir treatment indicated above. MiR-183-5p, miR-182-5p and miR-96-5p agomir treatment partially rescued the FOXO1 pSer256/total FOXO1 protein ratio and the AKT1 pSer473 level that were reduced by ox-LDL treatment in the HUVECs or HAECs. AKT1 overexpression significantly reduced FOXO1 protein expression, increased miR-182-5p and miR-183-5p expression, and partially alleviated ox-LDL induced HUVEC or HAEC apoptosis in an miR-183-5p and miR-182-5p-dependent manner. CONCLUSION: miR-183-96-182 clusters could partially alleviate ox-LDL-induced apoptosis in HUVECs or HAECs by targeting FOXO1.

13.
Oncol Rep ; 38(2): 1251-1258, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28677777

ABSTRACT

microRNA-33a (miR-33a) belongs to the miR-33 family that is implicated in the progression of various types of cancers. Aberrant expression of miR-33a has been detected in several human cancers, and has been shown to regulate the migration and invasion as well as proliferation and apoptosis of tumor cells. However, the clinical significance and precise mechanisms underlying the dysfunction of miR-33a in glioma have not been well investigated in previous studies. In this study, overexpression of miR-33a was observed in clinical glioma specimens and cell lines. Clinicopathological detection revealed that miR-33a highly expressing patients showed large tumor sized and advanced World Health Organization (WHO) grade as well as reduced overall survival. Furthermore, the results of in vitro experiments confirmed that loss of miR-33a resulted in reduced proliferation and enhanced apoptosis in U251 cells, while miR-33a restoration showed opposite effects in U87 cells. Further studies indicated that miR-33a knockdown restrained tumor growth of glioma in vivo. miR-33a negatively regulated the expression of sirtuin 6 (SIRT6) at both mRNA and protein levels via targeting the 3'UTR of SIRT6 mRNA. SIRT6 was underexpressed and inversely correlated with miR-33a expression in the glioma tissues. Mechanistically, SIRT6 overexpression increased the levels of lactate dehydrogenase (LDH) and reactive oxygen species (ROS) while it reduced cell survival under H2O2 treatment. In addition, SIRT6 restoration led to apoptosis with alterative expression of Bax, Bcl-2, cleaved caspase-8, and inhibition of Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathway in glioma. Thus, our studies demonstrated that the deregulation of miR-33a may promote tumor development in human glioma by regulating the expression of its target gene, SIRT6.


Subject(s)
Apoptosis/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Glioma/pathology , MicroRNAs/genetics , Oxidative Stress/genetics , Sirtuins/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Female , Follow-Up Studies , Glioma/genetics , Glioma/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Sirtuins/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Brain Res Bull ; 130: 27-35, 2017 04.
Article in English | MEDLINE | ID: mdl-28017783

ABSTRACT

Ischemic stroke is an acute life-threatening disease, which causes neurological dysfunction. The formation of new blood vessels around the infarct is vital to the restoration of perfusion and healing of brain tissue. Studies have shown that intelectin-1 (omentin) promotes endothelial cell function and angiogenesis in response to ischemia and inhibits apoptosis in rats with unilateral hind limb surgery. In the present study, we investigated the neuroprotective role of intelectin-1 following focal cerebral ischemia. We specifically assessed the effect of increased expression of intelectin-1 in promoting angiogenesis and reducing apoptosis. The treatment was administered using a lentiviral vector, 7 days prior to surgery. The surgery was performed using the established middle cerebral artery occlusion (MCAO) model in rats, and the outcome was evaluated 7 days after injury. Our results demonstrated a significant reduction in brain infarction volume following LV-intelectin-1 treatment. Additionally, CD34 and capillary density were increased in the cerebral ischemic penumbra. Real-time PCR and Western blot revealed an increased expression of intelectin-1, and phosphorylation of eNOS and AKT with enhanced expression of bcl-2 in brain tissues. These data suggest that the successful delivery of LV-intelectin-1 ameliorated ischemic brain injury. It promoted endothelial cell function and revasc ularization, and inhibited apoptosis in response to ischemia by stimulating the Akt-eNOS signaling pathway.


Subject(s)
Apoptosis , Brain Ischemia/physiopathology , Cytokines/metabolism , Lectins/metabolism , Neovascularization, Physiologic , Stroke/physiopathology , Animals , Brain Ischemia/complications , Brain Ischemia/pathology , Cerebral Cortex/metabolism , Male , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction , Stroke/complications , Stroke/pathology , Up-Regulation
15.
Inflammation ; 40(2): 473-485, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28032248

ABSTRACT

Advanced glycation end products (AGEs) have been confirmed to induce dysfunction in endothelial progenitor cells (EPCs) and play key roles in pathogenesis of diabetes-related vascular complications. The major function of sirtuin 3 (SIRT3) is to orchestrate oxidative metabolism and control reactive oxygen species (ROS) homeostasis, which are more closely related to EPCs' dysfunction. Our study therefore was designed to explore the role of SIRT3 on AGEs-induced EPCs dysfunction of. EPCs isolated from healthy adults were stimulated with AGEs and the expression of SIRT3 was assessed. Then, EPCs transfected with ad-SIRT3 or siRNA-SIRT3 were cultured with or without AGEs. EPCs function, including proliferation, migration; expression of manganese superoxide dismutase (MnSOD), ROS production, and interleukin-8 (IL-8); and vascular endothelial growth factor (VEGF) production were measured. In some experiments, EPCs were pre-cultured with anti-receptor for advanced glycation end products (RAGE) antibody or anti-neutralizing antibody, and then proliferation, migration, expression of MnSOD, ROS production, and IL-8 and VEGF production were measured. Our results showed that SIRT3 expressed in EPCs and AGEs decreased SIRT3 expression. SIRT3 knockdown with siRNA-SIRT3 promoted dysfunction in EPCs whereas SIRT3 activation with ad-SIRT3 strengthened anti-oxidant capacity and protected AGE-impaired dysfunction. Moreover, RAGE may involve in AGEs-decreased SIRT3 expression in EPCs. These data suggested an important role of SIRT3 in regulating EPCs bioactivity.


Subject(s)
Antioxidants/pharmacology , Endothelial Progenitor Cells/metabolism , Sirtuin 3/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelial Progenitor Cells/drug effects , Glycation End Products, Advanced/pharmacology , Humans , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products , Sirtuin 3/drug effects
16.
Physiol Behav ; 159: 72-9, 2016 May 15.
Article in English | MEDLINE | ID: mdl-26992957

ABSTRACT

Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) share specific molecular mechanisms, and agents with proven efficacy in one may be useful against the other. The glucagon-like peptide-1 (GLP-1) receptor agonist exendin-4 has similar properties to GLP-1 and is currently in clinical use for T2DM treatment. Thus, this study was designed to characterize the effects of exendin-4 on the impairment of learning and memory induced by amyloid protein (Aß) and its probable molecular underlying mechanisms. The results showed that (1) intracerebroventricular (i.c.v.) injection of Aß1-42 resulted in a significant decline of spatial learning and memory of rats in water maze tests; (2) pretreatment with exendin-4 effectively and dose-dependently protected against the Aß1-42-induced impairment of spatial learning and memory; (3) exendin-4 treatment significantly decreased the expression of Bax and cleaved caspase-3 and increased the expression of Bcl2 in Aß1-42-induced Alzheimer's rats. The vision and swimming speed of the rats among all groups in the visible platform tests did not show any difference. These findings indicate that systemic pretreatment with exendin-4 can effectively prevent the behavioral impairment induced by neurotoxic Aß1-42, and the underlying protective mechanism of exendin-4 may be involved in the Bcl2, Bax and caspase-3 pathways. Thus, the application of exendin-4 or the activation of its signaling pathways may be a promising strategy to ameliorate the degenerative processes observed in AD.


Subject(s)
Amyloid beta-Peptides/adverse effects , Glucagon-Like Peptide 1/agonists , Maze Learning/drug effects , Memory/drug effects , Peptides/pharmacology , Spatial Learning/drug effects , Venoms/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Exenatide , Male , Memory/physiology , Rats , Rats, Sprague-Dawley , Spatial Learning/physiology
17.
Cell Physiol Biochem ; 35(4): 1633-42, 2015.
Article in English | MEDLINE | ID: mdl-25824461

ABSTRACT

BACKGROUND: Homer is a family of post synaptic density proteins functionally and physically attached to target proteins at proline-rich sequences. Reducing Homer1b/c expression has been shown in previous studies to be protective against excitotoxic insults, implicating Homer1b/c in the physiological regulation of aberrant neuronal excitability. METHODS: To test the efficacy of a Homer1b/c reducing therapy for disorders with a detrimental hyperexcitability profile in mice, we used small interfere RNA (siRNA) to decrease endogenous Homer1b/c expression in mouse hippocampus. The baseline motor and cognitive behavior was measured by sensorimotor tests, Morris water maze and elevated plus maze tasks. The anti-epileptic effects of Homer1b/c knockdown were determined in two chemically induced seizure models induced by Picrotoxin (PTX) or pentylenetetrazole (PTZ) administration. RESULTS: The results of sensorimotor tests, Morris water maze and elevated plus maze tasks showed that Homer1b/c reduction had no effect on baseline motor or cognitive behavior. In two chemically induced seizure models, mice with reduced Homerb/c protein had less severe seizures than control mice. Total Homer1b/c protein levels and seizure severity were highly correlated, such that those mice with the most severe seizures also had the highest levels of Homer1b/c. In addition, the phosphorylation of mammalian target of rapamycin (mTOR) and its target protein S6 was significantly inhibited in Homer1b/c down-regulated mice. Homer1b/c knockdown-induced inhibition of mTOR pathway was partially ablated by the metabotropic glutamate receptor 5 (mGluR5) agonist CHPG. CONCLUSION: Our results demonstrate that endogenous Homer1b/c is integral for regulating neuronal hyperexcitability in adult animals and suggest that reduction of Homer1b/c could protect against chemically induced seizures through inhibition mTOR pathway.


Subject(s)
Carrier Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Behavior, Animal , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Disease Models, Animal , Down-Regulation/drug effects , Glycine/analogs & derivatives , Glycine/pharmacology , Hippocampus/metabolism , Homer Scaffolding Proteins , Maze Learning , Mice , PC12 Cells , Pentylenetetrazole/toxicity , Phenylacetates/pharmacology , Phosphorylation , Picrotoxin/toxicity , RNA Interference , Rats , Receptor, Metabotropic Glutamate 5/agonists , Receptor, Metabotropic Glutamate 5/metabolism , Ribosomal Protein S6 Kinases/metabolism , Seizures/chemically induced , Seizures/metabolism , Seizures/pathology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors
18.
Jpn J Clin Oncol ; 45(2): 169-75, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25416813

ABSTRACT

OBJECTIVE: Esophageal squamous cell carcinoma is increasingly treated with trimodality therapy. The objective of this Phase I/II clinical study is to assess the efficacy and safety of neoadjuvant radiochemotherapy with docetaxel and cisplatin and radiotherapy in patients with esophagectomy for locally advanced squamous cell carcinoma of the esophagus with neoadjuvant chemoradiotherapy. METHODS: Patients with esophageal squamous cell carcinoma received radiochemotherapy (50 Gy/25 fractions during Weeks 1-5) using a three-dimensional conformal radiation therapy or intensity-modulated radiation therapy technique together with weekly docetaxel (20 mg/m(2) at dose levels 1 and 2, 25 mg/m(2) at dose level 3 on Weeks 1-5) and cisplatin (30 mg/m(2) at dose level 1, 40 mg/m(2) at dose levels 2 and 3 on Weeks 1-5) from January 2009 to December 2011. The dose-limiting toxicities and maximum tolerated dose were the primary endpoints and overall response rate and progression-free survival were the secondary endpoints. RESULTS: Over this timeframe, a total of 49 patients completed trimodality therapy. Thirteen patients were treated at dose level 1, 21 patients at dose level 2 and 15 patients at dose level 3.The maximum tolerated dose for docetaxel was 20 mg/m(2) and cisplatin 40 mg/m(2). The complete response or partial response was observed in 26.5% (13/49) of patients. Thirty-four patients (69.4%) were treated with neoadjuvant radiochemotherapy followed by surgical resection. The median progression-free survival and median overall survival for all patients (n = 49) were 8 and 17.2 months, respectively. The median overall survival was 27.5 months for patients treated at dose level 2. CONCLUSION: Neoadjuvant radiochemotherapy with docetaxel 20 mg/m(2) and cisplatin 40 mg/m(2) was effective and tolerable induction regimen in patients with esophageal tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/therapy , Esophageal Neoplasms/therapy , Esophagectomy , Neoadjuvant Therapy/methods , Radiotherapy, Conformal , Adult , Aged , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/radiotherapy , Carcinoma, Squamous Cell/surgery , Chemoradiotherapy , Cisplatin/administration & dosage , Disease-Free Survival , Docetaxel , Dose-Response Relationship, Drug , Drug Administration Schedule , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Esophageal Neoplasms/radiotherapy , Esophageal Neoplasms/surgery , Female , Humans , Kaplan-Meier Estimate , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Prospective Studies , Radiotherapy, Intensity-Modulated , Taxoids/administration & dosage , Treatment Outcome
19.
J Stroke Cerebrovasc Dis ; 23(10): 2598-2606, 2014.
Article in English | MEDLINE | ID: mdl-25280822

ABSTRACT

BACKGROUND: Excessive inflammation and apoptosis contribute to the pathogenesis of ischemic brain damage. Nuclear factor-kappa B (NF-κB) is considered to be a key protein complex involved in this cascade of events. The aim of the present study was to clarify the protection mechanism of the mesenchymal stem cells (MSCs). METHODS: Lewis rats (N = 90) were randomly assigned to three groups: (1) the sham-operated group; (2) the saline group, in which the animals underwent rat transient middle cerebral artery occlusion (tMCAO, for 2 hours) and were treated with saline through the tail vein; and (3) the MSCs group, in which the animals underwent tMCAO (for 2 hours) and were infused with cultured human MSCs (4 × 10(6)/0.4 ml PBS) through the tail vein. At days 1 and 3 post-MSCs infusion, real-time PCR, and Western blot, immunohistochemical analyses were applied for tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and P-IKKß, p53, and B-cell lymphoma 2 (Bcl-2) expression levels. RESULTS: TNF-α, IL-1ß messenger RNA (mRNA) and P-IκB-α, P-IKKß, p53 protein expression levels were significantly increased in the saline group compared with the sham group. However, IκB-α and Bcl-2 protein expression levels were markedly decreased in the saline group. After injection of BrdU(+) MSCs, the expression levels of TNF-α, IL-1ß mRNA and P-IκB-α, P-IKKß, p53 protein were significantly decreased. Contrary to these findings, IκB-α, Bcl-2 protein expression levels were markedly increased. In addition, we found that infarct area was significantly reduced in MSCs group. CONCLUSIONS: These results suggest that MSCs' neuroprotection is attributable to its anti-inflammatory and antiapoptotic effect through inhibition of NF-κB.


Subject(s)
Apoptosis , Brain Ischemia/therapy , Brain/metabolism , Inflammation/metabolism , Mesenchymal Stem Cell Transplantation/methods , Animals , Brain/pathology , Brain Ischemia/metabolism , Disease Models, Animal , Humans , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , Infarction, Middle Cerebral Artery/complications , Interleukin-1beta/metabolism , Male , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Rats , Rats, Inbred Lew , Treatment Outcome , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
20.
Int J Mol Sci ; 15(2): 3172-85, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24566142

ABSTRACT

Intracellular calcium homeostasis disorder and mitochondrial dysfunction are involved in many acute and chronic brain diseases, including ischemic brain injury. An imbalance in mitochondrial fission and fusion is one of the most important structural abnormalities found in a large number of mitochondrial dysfunction related diseases. Here, we investigated the effects of mitochondrial division inhibitor A (mdivi A) and mdivi B, two small molecule inhibitors of mitochondrial fission protein dunamin-related protein 1 (Drp-1), in neuronal injury induced by oxygen-glucose deprivation (OGD) in PC12 cells. We found that mdivi A and mdivi B inhibited OGD-induced neuronal injury through attenuating apoptotic cell death. These two inhibitors also preserved mitochondrial function, as evidenced by reduced reactive oxygen species (ROS) generation and cytochrome c release, as well as prevented loss of mitochondrial membrane potential (MMP). Moreover, mdivi A and mdivi B significantly suppressed mitochondrial Ca(2+) uptake, but had no effect on cytoplasmic Ca(2+) after OGD injury. The results of calcium imaging and immunofluorescence staining showed that Drp-1 inhibitors attenuated endoplasmic reticulum (ER) Ca(2+) release and prevented ER morphological changes induced by OGD. These results demonstrate that Drp-1 inhibitors protect against ischemic neuronal injury through inhibiting mitochondrial Ca(2+) uptake from the ER store and attenuating mitochondrial dysfunction.


Subject(s)
Calcium/metabolism , Dynamins/antagonists & inhibitors , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/pharmacology , Mitochondria/drug effects , Animals , Cell Hypoxia , Cell Survival/drug effects , Cytochromes c/metabolism , Dynamins/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , PC12 Cells , Rats , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...