Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Environ Toxicol ; 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38409934

ABSTRACT

BACKGROUND: N6 -methyladenosine (m6 A) mediates RNA modification in various biological processes. It plays a key role in hepatocellular carcinoma (HCC) through regulating methyltransferase. The present study aims to analyze the correlation between the m6 A and the immune status of HCC, and to construct an m6 A-related prognostic signature for HCC. METHODS: HCC subtypes with different m6 A modification activities were identified based on the m6 A-related genes. Lasso Cox regression was applied to construct an m6 A-related prognostic model for HCC. Then, the prognostic potential of the constructed signature was evaluated and validated in the external validation dataset. Small interfering RNAs were designed to knockdown FBXO5. CCK-8 assay, Edu staining, wound healing assay, and Transwell cell invasion assay were used to detect cell proliferation, migration, and invasion ability. RESULTS: Two m6 A-related HCC subtypes were identified. The m6 A modification active group showed an immune suppressive microenvironment compared to the m6 A modification inactive group. The differentially expressed genes (DEGs) between the HCC subtypes were screened. Enrichment analysis was performed using the DEGs. Subsequently, an m6 A-related prognostic model was established. The prognostic model performed well in both training and validation datasets. Moreover, knockdown of FBXO5, one of the genes in the prognostic model, inhibited the proliferation, migration, and invasion of HepG2 cells. CONCLUSIONS: The heterogeneity of m6 A RNA methylation is associated with immune status in HCC. The constructed m6 A-related gene-based signature can predict the prognosis of HCC patients. The genes in the prognostic model also have therapeutic potential for HCC.

2.
Mol Neurobiol ; 60(7): 3741-3757, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36940077

ABSTRACT

Excessive activation of aldose reductase (AR) in the brain is a risk factor for aggravating cerebral ischemia injury. Epalrestat is the only AR inhibitor with proven safety and efficacy, which is used in the clinical treatment of diabetic neuropathy. However, the molecular mechanisms underlying the neuroprotection of epalrestat remain unknown in the ischemic brain. Recent studies have found that blood-brain barrier (BBB) damage was mainly caused by increased apoptosis and autophagy of brain microvascular endothelial cells (BMVECs) and decreased expression of tight junction proteins. Thus, we hypothesized that the protective effect of epalrestat is mainly related to regulating the survival of BMVECs and tight junction protein levels after cerebral ischemia. To test this hypothesis, a mouse model of cerebral ischemia was established by permanent middle cerebral artery ligation (pMCAL), and the mice were treated with epalrestat or saline as a control. Epalrestat reduced the ischemic volume, enhanced BBB function, and improved the neurobehavior after cerebral ischemia. In vitro studies revealed that epalrestat increased the expression of tight junction proteins, and reduced the levels of cleaved-caspase3 and LC3 proteins in mouse BMVECs (bEnd.3 cells) exposed to oxygen-glucose deprivation (OGD). In addition, bicalutamide (an AKT inhibitor) and rapamycin (an mTOR inhibitor) increased the epalrestat-induced reduction in apoptosis and autophagy related protein levels in bEnd.3 cells with OGD treatment. Our findings suggest that epalrestat improves BBB function, which may be accomplished by reducing AR activation, promoting tight junction proteins expression, and upregulating AKT/mTOR signaling pathway to inhibit apoptosis and autophagy in BMVECs.


Subject(s)
Brain Injuries , Brain Ischemia , Mice , Animals , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Aldehyde Reductase/metabolism , Aldehyde Reductase/pharmacology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Cerebral Infarction/metabolism , Brain Injuries/metabolism , Glucose/metabolism , Tight Junction Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism
3.
Allergy Asthma Clin Immunol ; 18(1): 66, 2022 Aug 04.
Article in English | MEDLINE | ID: mdl-35927714

ABSTRACT

BACKGROUND: Several miRNAs are now known to have clear connections to the pathogenesis of asthma. The present study focused on the potential role of miR-3934 during asthma development. METHODS: miR-3934 was detected as a down-regulated miRNA in basophils by sequencing analysis. Next, the expression levels of miR-3934 in peripheral blood mononuclear cells of 50 asthma patients and 50 healthy volunteers were examined by RT-qPCR methods. The basophils were then treated with AGEs and transfected with miR-3934 mimics. The apoptosis levels were examined by flow cytometry assay; and the expression levels of cytokines were detected using the ELISA kits. Finally, the Western blot was performed to examined the expression of key molecules in the TGF-ß/Smad signaling pathway. RESULTS: miR-3934 was down-regulated in the basophils of asthmatic patients. The expression of the pro-inflammatory cytokines IL-6, IL-8 and IL-33 was enhanced in basophils from asthmatic patients, and this effect was partially reversed by transfection of miR-3934 mimics. Furthermore, receiver operating characteristics analysis showed that miR-3934 levels can be used to distinguish asthma patients from healthy individuals. miR-3934 partially inhibited advanced glycation end products-induced increases in basophil apoptosis by suppressing expression of RAGE. CONCLUSION: Our results indicate that miR-3934 acts to mitigate the pathogenesis of asthma by targeting RAGE and suppressing TGF-ß/Smad signaling.

4.
Int Immunopharmacol ; 97: 107811, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34091117

ABSTRACT

Multiple sclerosis (MS) is a neurodegenerative and demyelinating autoimmune disease mediated by autoreactive T cells that affects the central nervous system (CNS). Electroacupuncture (EA) has emerged as an alternative or supplemental treatment for MS, but the mechanism by which EA may alleviate MS symptoms is unresolved. Here, we examined the effects of EA at the Zusanli (ST36) acupoint on mice with experimental autoimmune encephalomyelitis (EAE), the predominant animal model of MS. The effects of EA on EAE emergence, inflammatory cell levels, proinflammatory cytokines, and spinal cord pathology were examined. EA treatment attenuated the EAE clinical score and associated spinal cord demyelination, while reducing the presence of proinflammatory cytokines in mononuclear cells (MNCs), downregulating microRNA (miR)-155, and upregulating the opioid peptide precursor proopiomelanocortin (POMC) in the CNS. Experiments in which cultured neurons were transfected with a miR-155 mimic or a miR-155 inhibitor further showed that the direct modulation of miR-155 levels could regulate POMC levels in neurons. In conclusion, the alleviation of EAE by EA is characterized by reduced proportions of Th1/Th17 cells and increased proportions of Th2 cells, POMC upregulation, and miR-155 downregulation, while miR-155 itself can suppress POMC expression. These results, support the hypothesis that the effects of EA on EAE may involve the downregulation of miR-155.


Subject(s)
Electroacupuncture , Encephalomyelitis, Autoimmune, Experimental/therapy , MicroRNAs/metabolism , Multiple Sclerosis/therapy , Animals , Down-Regulation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Mice , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , Multiple Sclerosis/immunology , Pro-Opiomelanocortin/genetics , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Up-Regulation/immunology
5.
Immunol Lett ; 229: 18-26, 2021 01.
Article in English | MEDLINE | ID: mdl-33238163

ABSTRACT

Bone marrow mesenchymal stem cells (BMSCs) have the immuno-modulatory capacity to ameliorate autoimmune diseases, such as multiple schlerosis (MS), systemic lupus erythematosus and rheumatoid arthritis. However, BMSC-mediated immunosuppression can be challenging to achieve. The efficacy of BMSC transplantation may be augmented by an adjuvant therapy. Here, we demonstrated that treatment of mice with experimental autoimmune encephalomyelitis (EAE), a model of MS, with BMSCs over-expressing microRNA (miR)-23b provided better synergistic and longer-term therapeutic effects than treatment with traditional BMSCs. Over-expression of miR-23b enhanced the ability of BMSCs to inhibit differentiation of Th17 cells and reduced IL-17 secretion. Compared to traditional BMSCs, the miR-23b over-expressing BMSCs (miR23b-BMSCs) exhibited enhanced secretion of tumor growth factor beta 1 (TGF-ß1), a cytokine that promotes the differentiation of regulatory T (Treg) cells. Pathologically, miR23b-BMSC transplantation delayed EAE progression, apparently by reducing the Th17/Treg cell ratio and inhibiting inflammatory cell infiltration across the blood-brain barrier, and thus slowing spinal cord demyelination. These results may lead to better utility of BMSCs as a treatment for autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Animals , Biomarkers , Cell Line , Cytokines/metabolism , Disease Management , Disease Models, Animal , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Gene Expression , Genetic Vectors/genetics , Humans , Immunophenotyping , Inflammation Mediators/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mice , Signal Transduction , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transduction, Genetic , Treatment Outcome
6.
Neurochem Int ; 141: 104860, 2020 12.
Article in English | MEDLINE | ID: mdl-33022312

ABSTRACT

Cerebral ischemia-reperfusion injury induces a secondary immune inflammatory reaction that exacerbates brain injury and clinical prognosis. Dendritic cells (DCs) and microglia are both important regulators of neuroinflammation. Studies have confirmed that a large number of cells express the DC surface marker CD11c in the ischemic area, and some of these cells also express microglial markers. However, the specific mechanism of transformation between microglia and DCs and their roles in the process of cerebral ischemia-reperfusion injury are still not clear. In this study, we established a mouse model and flow cytometry was used to detect the expression of mature DC surface molecules in activated microglia. IFN-γ knockout mice were used to determine the regulatory effect of IFN-γ on microglial transformation. We found that CD11c+ cells were derived from microglia after ischemia-reperfusion injury, and this group of cells highly expressed MHC-II molecules and other costimulatory molecules, such as CD80 and CD86, which were regulated by IFN-γ and its downstream signaling molecules ERK/c-myc. In summary, our results showed in cerebral ischemia-reperfusion injury, IFN-γ regulates the transformation of microglia to DC-like cells. Microglial-derived DC-like cells possess the ability to present antigens and activate naïve T cells which is regulated by the ERK/c-myc signaling pathway.


Subject(s)
Dendrites/drug effects , Interferon-gamma/genetics , MAP Kinase Signaling System/drug effects , Microglia/drug effects , Proto-Oncogene Proteins c-myc/drug effects , Reperfusion Injury/pathology , Animals , CD11 Antigens/metabolism , Dendrites/pathology , Genes, MHC Class II , Interferon-gamma/physiology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Primary Cell Culture , Receptors, Interferon/biosynthesis , T-Lymphocytes
7.
FASEB J ; 34(10): 13762-13775, 2020 10.
Article in English | MEDLINE | ID: mdl-32808351

ABSTRACT

Cerebral ischemia causes damage to the structure and function of the blood-brain barrier (BBB) and alleviating BBB destruction will be of great significance for the treatment and prognosis of ischemic stroke. Recently, microRNAs have been shown to play a critical role in BBB integrity. However, the potential mechanism by which microRNA-182 (miR-182) affects the BBB in ischemic stroke remains unclear. We demonstrated for the first time that cerebral ischemia leads to a significant progressive increase in miR-182 after pMCAO, and bEnd.3 cells are the primary target cells of miR-182. In miR-182 KD transgenic mice, infarct volume, and BBB permeability were attenuated, and tight junction (TJ) proteins increased. Inhibition of miR-182 with an antagomir reduced OGD-induced apoptosis of bEnd.3 cells and the loss of ZO-1 and Occludin. To further explore the mechanism by which miR-182 regulates BBB integrity, we detected the apoptotic proteins Bcl-2/Bax and demonstrated that mTOR and FOXO1 were the targets of miR-182. Inhibition of mTOR/FOXO1 by rapamycin/AS1842856 decreased the ratio of Bcl-2/Bax and exacerbated TJ protein loss. Taken together, inhibition of miR-182 protects BBB integrity by reducing endothelial cell apoptosis through the mTOR/FOXO1 pathway. Thus, miR-182 may be a potential target for the treatment of BBB disruption during cerebral ischemia.


Subject(s)
Blood-Brain Barrier/metabolism , Infarction, Middle Cerebral Artery/metabolism , MicroRNAs/metabolism , Signal Transduction , Animals , Apoptosis , Cell Line , Cells, Cultured , Down-Regulation , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Infarction, Middle Cerebral Artery/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
8.
Front Cell Dev Biol ; 8: 680, 2020.
Article in English | MEDLINE | ID: mdl-32850807

ABSTRACT

Long non-coding RNAs (lncRNAs) have been reported to interact with BRCA1/2 to regulate homologous recombination (HR) by diverse mechanisms in ovarian cancers (OvCa). However, genome-wide screening of BRCA1/2-related lncRNAs and their clinical significance is still unexplored. In this study, we constructed a global BRCA1/2-directed lncRNA-associated ceRNA network by integrating paired lncRNA expression profiles, miRNA expression profiles, and BRCA1/2 expression profiles in BRCA1/2 wild-type patients and identified 111 BRCA1/2-related lncRNAs. Using the stepwise regression and Cox regression analysis, we developed a BRCA1/2-directed lncRNA signature (BRCALncSig), composing of three lncRNAs (LINC01619, DLX6-AS1, and AC004943.2) from the list of 111 BRCA1/2-related lncRNAs, which was an independent prognostic factor and was able to classify the patients into high- and low-risk groups with significantly different survival in the training dataset (HR = 2.73, 95 CI 1.65-4.51, p < 0.001). The prognostic performance of the BRCALncSig was further validated in the testing dataset (HR = 1.9, 95 CI 1.21-2.99, p = 0.005) and entire TCGA dataset (HR = 2.17, 95 CI 1.56-3.01, p < 0.001). Furthermore, the BRCALncSig is associated with chemo-response and was also capable of discriminating nonequivalent outcomes for patients achieving complete response (CR) (log-rank p = 0.003). Functional analyses suggested that mRNAs co-expressed with the BRCALncSig were enriched in cancer-related or cell proliferation-related biological processes and pathways. In summary, our study highlighted the clinical implication of BRCA1/2-directed lncRNAs in the prognosis and treatment response of BRCA1/2 wild-type patients.

9.
Biomed Res Int ; 2020: 8916729, 2020.
Article in English | MEDLINE | ID: mdl-32851091

ABSTRACT

AURKA, a cell cycle-regulated kinase, is associated with malignant transformation and progression in many cancer types. We analyzed the expression change of AURKA in pan-cancer and its effect on the prognosis of cancer patients using the TCGA dataset. We revealed that AURKA was extensively elevated and predicted a poor prognosis in most of the detected cancer types, with an exception in colon cancer. AURKA was elevated in colon cancer, but the upregulation of AURKA indicated better outcomes of colon cancer patients. Then we revealed that undermethylation of the AURKA gene and several transcription factors contributed to the upregulation of AURKA in colon cancer. Moreover, we demonstrated that AURKA overexpression promoted the death of colon cancer cells induced by Oxaliplatin, whereas knockdown of AURKA significantly weakened the chemosensitivity of colon cancer cells to Oxaliplatin. Mechanistically, AURKA inhibited DNA damage response by suppressing the expression of various DNA damage repair genes in a TP53-dependent manner, which can partly explain that ARUKA is associated with a beneficial outcome of colon cancer. This study provided a possibility to use AURKA as a biomarker to predict the chemosensitivity of colon cancer to platinum in the clinic.


Subject(s)
Aurora Kinase A/genetics , Colonic Neoplasms/drug therapy , Oxaliplatin/pharmacology , Tumor Suppressor Protein p53/genetics , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA Damage/drug effects , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Mice , Oxaliplatin/adverse effects , Xenograft Model Antitumor Assays
10.
Int J Biol Macromol ; 161: 692-703, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32535204

ABSTRACT

Mutation in TMEM240 is suggested to cause SCA21, but the specific mechanism has not been clarified. The subcellular localization, specific biological function, and corresponding mechanism of action of TMEM240 have also not been delineated. In this study, the mRNA and protein expression of TMEM240 were assessed using qPCR and western blotting, respectively. Live cell imaging was used to establish the sub-cellular location of TMEM240, and electron microscopy was used to determine the morphology and distribution of TMEM240 in the cell. TMEM240 was specifically expressed in the neurons. Exogenous TMEM240 formed a multilayered cell structure, which we refer to as TMEM240-Body (T240-Body). T240-Body was separated and purified by centrifugation and filtration. An anchor protein His-tagged-GFP-BP on Ni-NTA agarose was used to pull down T240-GFP binding proteins. Both the N-terminal and the C-terminal of TMEM240 were confirmed to be inside the T240-Body. Co-localization experiments suggested that peroxisomes might contribute to T240-Body formation, and the two transmembrane regions of TMEM240 appear to be essential for formation of the T240-Body. Emerin protein contributed to formation of T240-Body when combined with TMEM240. Overall, this study provides new insights into TMEM240, which inform future research to further our understanding of its biological function.


Subject(s)
Brain , Membrane Proteins/metabolism , Mutation , Neurons , Peroxisomes , Spinocerebellar Degenerations , Animals , Brain/metabolism , Brain/ultrastructure , Hep G2 Cells , Humans , Membrane Proteins/genetics , Mice , Neurons/metabolism , Neurons/ultrastructure , Peroxisomes/genetics , Peroxisomes/metabolism , Peroxisomes/ultrastructure , Spinocerebellar Degenerations/genetics , Spinocerebellar Degenerations/metabolism , Spinocerebellar Degenerations/pathology
11.
Int Immunopharmacol ; 84: 106515, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32311672

ABSTRACT

Immunity-related GTPase family M1 protein (lRGM1) plays an important role in host resistance to infection, immune inflammation, and tumors, and it is expressed in various tissues and cells, including the central nervous system, cardiovascular system, bone marrow-derived cells, glioma, and melanoma. However, the effect of IRGM1 in the muscles has not been reported to date. In this study, Irgm1-/- mice were used to evaluate the effect of lrgm1 on regeneration after skeletal muscle injury. The tibialis anterior muscle in Irgm1-/- mice was poorly repaired after BaCl2-induced injury, whereas lrgm1 knockout itself had no significant effect on the differentiation of myoblasts. However, the microenvironment of Irgm1-/- mice with a high interferon-gamma level inhibited the differentiation of myoblasts in vivo. These results suggest that lrgm1 knockout indirectly inhibits skeletal muscle regeneration after injury, providing new insights into the biological function of IRGM1.


Subject(s)
GTP-Binding Proteins/physiology , Muscle, Skeletal/physiology , Animals , Barium Compounds , Cell Differentiation , Cells, Cultured , Chlorides , GTP-Binding Proteins/genetics , Interferon-gamma/physiology , Male , Mice, Knockout , Muscle, Skeletal/injuries , Regeneration , Satellite Cells, Skeletal Muscle/physiology
12.
Int Immunopharmacol ; 82: 106335, 2020 Feb 25.
Article in English | MEDLINE | ID: mdl-32109680

ABSTRACT

Osteopontin (OPN) is a multifunctional extracellular matrix phosphoprotein that has a specific and complicated structure, and contributes to numerous physiological and pathological activities. The mechanism of OPN in many diseases has been confirmed; however, the role of OPN in myasthenia gravis (MG) remains unclear. In this study, we recombined rat OPN protein in vitro, and assessed how OPN affects the development of autoimmunity using an experimental autoimmune myasthenia gravis (EAMG) rat model. The results showed that the concentration of OPN in serum was up-regulated. Both mRNA and protein levels in splenocytes increased in the EAMG model. OPN treatment in vitro strongly promoted the differentiation of Th1 cells, and inhibited the differentiation of Treg cells. Intraperitoneal injection of OPN revealed the early incidence of EAMG, and more serious disease. This effect was accompanied by an increased percentage of Th1 cells. In conclusion, OPN likely exacerbates the pathogenesis of EAMG by promoting the differentiation of Th1 cells and inhibiting the differentiation of Treg cells.

13.
Int Immunopharmacol ; 81: 106176, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32044667

ABSTRACT

A high salt diet (HSD) is among the most important risk factors for many diseases. One mechanism by which HSD aggravates cerebral ischemic injury is independent of blood pressure changes. The direct role of HSD in inflammation after cerebral ischemia is unclear. In this research, after twenty-one days of being fed a high salt diet, permanent focal ischemia was induced in mice via operation. At 12 h and 1, 3 and 5 days postischemia, the effects of HSD on the lesion volume, microglia polarization, aldose reductase (AR) expression, and inflammatory processes were analyzed. We report that in mice, surplus dietary salt promotes inflammation and increases the activation of classical lipopolysaccharide (LPS)-induced microglia/macrophages (M1). This effect depends on the expression of the AR protein in activated microglia after permanent middle cerebral artery ligation (pMCAL) in HSD mice. The administration of either the AR inhibitor Epalrestat or a p38-neutralizing antibody blocked the polarization of microglia and alleviated stroke injury. In conclusion, HSD promotes polarization in pro-inflammatory M1 microglia by upregulating the expression of the AR protein via p38/MAPK, thereby exacerbating the development of ischemia stroke.


Subject(s)
Brain Ischemia/metabolism , Macrophages/physiology , Microglia/physiology , Salts/administration & dosage , Androgen Receptor Antagonists/administration & dosage , Animals , Brain Ischemia/pathology , Cell Differentiation , Cytokines/metabolism , Eating , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , Receptors, Androgen/metabolism , Rhodanine/administration & dosage , Rhodanine/analogs & derivatives , Salts/adverse effects , Signal Transduction , Th1 Cells/immunology , Thiazolidines/administration & dosage , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Biomed Res Int ; 2019: 4068521, 2019.
Article in English | MEDLINE | ID: mdl-31886210

ABSTRACT

PURPOSE: Osteosarcoma is one of the most common primary malignant, aggressive bone neoplasms. However, the mechanisms of osteosarcoma proliferation, migration, and invasion are not well understood. To explore the possible mechanism of osteosarcoma progression, we used a public database for gene analysis to identify the possible factors that are important in osteosarcoma progression. Nuclear factor interleukin 3 (NFIL3) regulated was highly expressed in sarcoma tissues. In this study, we meant to probe the function of NFIL3 in osteosarcoma proliferation, migration, and invasion. METHODS: The expression of NFIL3 in osteosarcoma tissues was analysed via RT-PCR and immunohistochemistry staining. In order to elucidate the function of NFIL3 in osteosarcoma, we performed cell growth assays and colony formation assays to explore the role of NFIL3 in proliferation in osteosarcoma cells. Futhermore, we analysed osteosarcoma cell migration and invasion via wound healing assays and transwell migration and invasion assays. RESULTS: NFIL3 is overexpressed in osteosarcoma tissues; 15 of the 20 osteosarcoma tissues analysed highly expressed NFIL3. Our in vitro experiments confirmed that NFIL3 promoted the proliferation of M6-63 and SaOS2 cells (P < 0.01). In addition, NFIL3 promoted the migration and invasion of osteosarcoma cells (P < 0.05). CONCLUSION: NFIL3 is highly expressed in osteosarcoma tissues and thus promotes the proliferation, migration, and invasion of osteosarcoma cells. NFIL3 is potential to become a new target for development of novel treatment strategies of osteosarcoma.


Subject(s)
Basic-Leucine Zipper Transcription Factors/biosynthesis , Bone Neoplasms/metabolism , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Osteosarcoma/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Female , Humans , Male , Neoplasm Proteins/genetics , Osteosarcoma/genetics , Osteosarcoma/pathology
15.
Eur J Immunol ; 49(12): 2184-2194, 2019 12.
Article in English | MEDLINE | ID: mdl-31411745

ABSTRACT

MicroRNA 182 is important for the clonal expansion of CD4+ T cells (Th) following IL-2 stimulation and is a potential therapeutic target for autoimmune diseases. In the present study, we investigated the role of microRNA 182 in the differentiation of pro-inflammatory CD4+ T helper cell by overexpressing or silencing microRNA 182 expression both in in vivo and in vitro settings. We report that in the studied Chinese cohort, microRNA 182 is upregulated in patients with relapse and remitting multiple sclerosis (RRMS) and this upregulation is associated with increased IFN-γ producing CD4+ Th1 cells in the circulation. In the murine experimental autoimmune encephalomyelitis (EAE) model, global microRNA 182 overexpression exacerbates clinical symptoms and results in augmented CD4+ IFN-γ+ Th1 and CD4+ IL-17+ Th17 differentiation in vivo. Addition of microRNA 182 mimics in vitro represses both the protein expression and transcriptional activity of hypoxia induced factor 1α (HIF-1α) but increases the level of IFN-γ transcripts in sorted murine CD4+ T cells. Together, our results provide evidence that microRNA 182 may be one of the transitional hubs contribution to regulate Th cells expansion in response to self-antigens and differentiation of antigen specific Th cells during the progression of autoimmune inflammations.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , MicroRNAs/immunology , Multiple Sclerosis/immunology , Th1 Cells/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Interferon-gamma/immunology , Interleukin-17/immunology , Mice , Multiple Sclerosis/pathology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology
16.
Int Immunopharmacol ; 75: 105822, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31437793

ABSTRACT

Metformin, the most widely used medicine for type 2 diabetes, displays anti-inflammatory functions via activating AMP-activated protein kinase (AMPK). Circulating autoantibodies and disequilibrium of helper T cells and regulatory T cells are pathological hallmarks of myasthenia gravis (MG). Rectify the imbalance of different T cell populations has become an important therapeutic strategy to treat MG. In this study, we assessed the effect of metformin on the development of autoimmunity using an experimental autoimmune myasthenia gravis (EAMG) rat model. We first provided evidence that oral administration of metformin attenuated the onset of EAMG. This effect was accompanied by a substantial decrease of circulating auto-antibody levels with no effect on blood glucose level. While metformin treatment in vitro showed little effect on inducible Treg, metformin strongly inhibited Th17 cell differentiation through the increase of reactive oxygen species and AMPK. Furthermore, an attenuation of antigen-induced IgG2b antibody production by two different doses of metformin was also observed in the AChR-specific recall response. In conclusion, the above results indicate that metformin may have therapeutic value for the clinical treatment of MG.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , AMP-Activated Protein Kinases/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Antibodies/blood , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Blood Glucose/drug effects , Disease Models, Animal , Female , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Rats, Inbred Lew , Reactive Oxygen Species/immunology , Receptors, Cholinergic/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
17.
J Cell Biochem ; 120(8): 14076-14087, 2019 08.
Article in English | MEDLINE | ID: mdl-30968973

ABSTRACT

Intermittent hypobaric hypoxia can produce a protective effect on both the nervous system and non-nervous system tissues. Intermittent hypobaric hypoxia preconditioning has been shown to protect rats from cardiac ischemia-reperfusion injury by decreasing cardiac iron levels and reactive oxygen species (ROS) production, thereby decreasing oxidative stress to achieve protection. However, the specific mechanism underlying the protective effect of hypobaric hypoxia is unclear. To shed light on this phenomenon, we subjected Sprague-Dawley rats to hypobaric hypoxic preconditioning (8 hours/day). The treatment was continued for 4 weeks. We then measured the iron content in the heart, liver, spleen, and kidney. The iron levels in all of the assessed tissues decreased significantly after hypobaric hypoxia treatment, corroborating previous results that hypobaric hypoxia may produce its protective effect by decreasing ROS production by limiting the levels of catalytic iron in the tissue. We next assessed the expression levels of several proteins involved in iron metabolism (transferrin receptor, L-ferritin, and ferroportin1 [FPN1]). The increased transferrin receptor and decreased L-ferritin levels after hypobaric hypoxia were indicative of a low-iron phenotype, while FPN1 levels remained unchanged. We also examined hepcidin, transmembrane serine proteases 6 (TMPRSS6), erythroferrone (ERFE), and erythropoietin (EPO) levels, all of which play a role in the regulation of systemic iron metabolism. The expression of hepcidin decreased significantly after hypobaric hypoxia treatment, whereas the expression of TMPRSS6 and ERFE and EPO increased sharply. Finally, we measured serum iron and total iron binding capacity in the serum, as well as red blood cell count, mean corpuscular volume, hematocrit, red blood cell distribution width SD, and red blood cell distribution width CV. As expected, all of these values increased after the hypobaric hypoxia treatment. Taken together, our results show that hypobaric hypoxia can stimulate erythropoiesis, which systemically draws iron away from nonhematopoietic tissue through decreased hepcidin levels.


Subject(s)
Hypoxia/metabolism , Iron/metabolism , Animals , Apoferritins/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Erythrocyte Indices , Erythrocytes/metabolism , Erythropoietin/blood , Erythropoietin/metabolism , Hematocrit , Hepcidins/metabolism , Hypoxia/blood , Iron/blood , Male , Membrane Proteins , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptors, Transferrin/metabolism , Serine Endopeptidases
18.
Neurosci Bull ; 35(3): 507-518, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30796753

ABSTRACT

Myasthenia gravis (MG) is a prototypical antibody-mediated neurological autoimmune disease with the involvement of humoral immune responses in its pathogenesis. T follicular helper (Tfh) cells have been implicated in many autoimmune diseases. However, whether and how Tfh cells are involved in MG remain unclear. Here, we established and studied a widely-used and approved animal model of human MG, the rat model with acetylcholine receptor alpha (AChRα) subunit (R-AChR97-116)-induced experimental autoimmune myasthenia gravis (EAMG). This model presented mild body-weight loss 10 days after the first immunization (representing the early stage of disease) and more obvious clinical manifestations and body-weight loss 7 days after the second immunization (representing the late stage of disease). AChR-specific pre-Tfh cells and mature Tfh cells were detected in these two stages, respectively. In co-cultures of Tfh cells and B cells, the number of IgG2b-secreting B cells and the level of anti-AChR antibodies in the supernatant were higher in the cultures containing EAMG-derived Tfh cells. In immunohistochemistry and immunofluorescence assays, a substantial number of CD4+/Bcl-6+ T cells and a greater number of larger germinal centers were observed in lymph node tissues resected from EAMG rats. Based on these results, we hypothesize that an AChR-specific Tfh cell-mediated humoral immune response contributes to the development of EAMG.


Subject(s)
Immunity, Humoral , Myasthenia Gravis, Autoimmune, Experimental/immunology , Receptors, Cholinergic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , B-Lymphocytes/immunology , Disease Models, Animal , Female , Lymph Nodes/immunology , Protein Subunits/immunology , Proto-Oncogene Proteins c-bcl-6/immunology , Rats, Inbred Lew , Receptor Cross-Talk
19.
Genomics ; 110(6): 337-346, 2018 11.
Article in English | MEDLINE | ID: mdl-29366861

ABSTRACT

Long non-coding RNAs have the potential to regulate immune responses. Their impact on multiple sclerosis has remained elusive. For illustrating their roles in experimental autoimmune encephalomyelitis (EAE) pathogenesis, we investigated the differential expression of lncRNAs and mRNAs in CD4+Th cells obtained from myelin oligodendrocytic glycoprotein35-55(MOG35-55)-induced EAE and complete Freund's adjuvant (CFA) controls. We observed differential expression of 1112 lncRNAs and 519 mRNAs in CD4+Th cells. The functional network showed lncRNAs had the capacity to modulate EAE pathogenesis via regulating many known EAE regulators such as Ptpn6. Predicting the function of lncRNAs demonstrated that dysregulated lncRNAs were closely associated with the development of EAE. These dysregulated lncRNAs may have function in EAE and they could be novel biomarkers and therapeutic targets of EAE. However, the precise mechanisms and biological functions of these specific lncRNAs in EAE pathogenesis require further study.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Regulation , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Gene Expression Profiling , Mice , Myelin-Oligodendrocyte Glycoprotein/pharmacology , Myelin-Oligodendrocyte Glycoprotein/toxicity , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , T-Lymphocytes, Helper-Inducer/drug effects
20.
Sci Rep ; 7(1): 14736, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29116111

ABSTRACT

miRNAs, a class of short but stable noncoding RNA molecules, have been revealed to play important roles in the DNA damage response (DDR). However, their functions in cancer genome instability and the consequent clinical effect as the response to chemotherapy have not been fully elucidated. In this study, we utilized multidimensional TCGA data and the known miRNAs involved in DDR to identify a miRNA-regulatory network that responds to DNA damage. Additionally, based on the expression of ten miRNAs in this network, we developed a 10-miRNA-score that predicts defects in the homologous recombination (HR) pathway and genome instability in ovarian cancer. Importantly, consistent with the association between HR defects and improved response to chemotherapeutic agents, the 10-miRNA-score predicts the outcome of ovarian cancer patients treated with platinum agents, with a surprisingly better performance than the indexes of DNA damage. Therefore, our study demonstrates the implication of miRNA expression on cancer genome instability and provides an alternative method to identify DDR defects in patients who show the best effect with platinum drug treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Genomic Instability , MicroRNAs/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Platinum Compounds/therapeutic use , Female , Gene Regulatory Networks , Humans , Prognosis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...