Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Neuro Oncol ; 25(5): 841-853, 2023 05 04.
Article in English | MEDLINE | ID: mdl-36239338

ABSTRACT

BACKGROUND: ALKBH5 is aberrantly activated and exerts critical roles in facilitating the development of glioblastoma. However, the underlying activation mechanism by which ALKBH5 protein is increased in glioblastoma is not completely understood. Our study aimed to elucidate the signaling pathways involved in mediating ALKBH5 protein stability. METHODS: The contribution of deubiquitinating enzymes (DUB) to the fluctuation of ALKBH5 protein expression was globally profiled with western blot analysis. Mass spectrometry and immunoprecipitation were performed to identify the USP36 and ALKBH5 interaction. The effects of USP36 on the stability of ALKBH5 were detected with in vivo and in vitro ubiquitination assays. Cell proliferation assays, neurosphere formation, limited dilution assay, and intracranial tumor growth assays were implemented to assess the collaborative capacities of USP36 and ALKBH5 in tumorigenesis. RESULTS: Ubiquitin-specific peptidase 36 (USP36), as a potential ALKBH5-activating DUB, played an essential role in stabilization of ALKBH5 and regulation of ALKBH5-mediated gene expression in glioblastoma. The depletion of USP36 drastically impaired cell proliferation deteriorated the self-renewal of GSCs and sensitized GSCs to temozolomide (TMZ) treatment. Furthermore, the deletion of USP36 substantially decreased the in vivo tumor growth when monitored by bioluminescence imaging. Our findings indicate that USP36 regulates the protein degradation and expression of ALKBH5, and the USP36-ALKBH5 axis orchestrates glioma tumorigenesis. CONCLUSION: Our findings identify USP36 as a DUB of ALKBH5 and its role in glioblastoma progression, which may serve as a potential therapeutic target for glioblastoma treatment.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/drug therapy , Ubiquitin Thiolesterase/genetics , Ubiquitination , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Proliferation , Carcinogenesis , Cell Transformation, Neoplastic , Cell Line, Tumor , AlkB Homolog 5, RNA Demethylase/genetics , AlkB Homolog 5, RNA Demethylase/metabolism
2.
Oxid Med Cell Longev ; 2022: 9306614, 2022.
Article in English | MEDLINE | ID: mdl-35915613

ABSTRACT

As a previously discovered target of DNA damage, Na+/H+ exchanger 1 (NHE1) plays a role in regulation of intracellular pH (pHi) through the extrusion of intracellular proton (H+) in exchange for extracellular sodium (Na+). Its abnormal expression and dysfunction have been reported in solid tumor and hematopoietic malignancies. Here, we reported that suppression of NHE1 in BCR-ABL+ hematopoietic malignancies' K562 cells treated with Etoposide was manipulated by miR-19 and c-MYC. Inhibition of miR-19 or c-MYC enhanced the expression of NHE1 and sensitized K562 cells to Etoposide in vitro. The in vivo nude mouse transplantation model was also performed to confirm the enhanced sensitivity of K562 cells to Etoposide by inhibiting the miR-19 or c-MYC pathway. TCGA analysis conferred a negative correlation between miR-19 level and leukemia patients' survival. Thus, our results provided a potential management by which the c-MYC-miRNA 19 pathway might have a crucial impact on sensitizing K562 cells to Etoposide in the therapeutic approaches.


Subject(s)
Hematologic Neoplasms , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , MicroRNAs , Sodium-Hydrogen Exchanger 1/metabolism , Animals , Etoposide/pharmacology , Etoposide/therapeutic use , Gene Expression Regulation, Leukemic , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , MicroRNAs/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Up-Regulation
3.
Cancer Cell ; 38(6): 857-871.e7, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33125861

ABSTRACT

Brain metastasis is a major cause of cancer mortality, but its molecular mechanisms are severely understudied. In addition, little is known regarding the role of m6A reader YTHDF3 in human diseases. Here, we show that YTHDF3 overexpression clinically correlates with brain metastases in breast cancer patients. YTHDF3 promotes cancer cell interactions with brain endothelial cells and astrocytes, blood-brain barrier extravasation, angiogenesis, and outgrow. Mechanistically, YTHDF3 enhances the translation of m6A-enriched transcripts for ST6GALNAC5, GJA1, and EGFR, all associated with brain metastasis. Furthermore, overexpression of YTHDF3 in brain metastases is attributed to increased gene copy number and the autoregulation of YTHDF3 cap-independent translation by binding to m6A residues within its own 5' UTR. Our work uncovers an essential role of YTHDF3 in controlling the interaction between cancer cells and brain microenvironment, thereby inducing brain metastatic competence.


Subject(s)
Adenosine/analogs & derivatives , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Breast Neoplasms/pathology , RNA-Binding Proteins/metabolism , Up-Regulation , 5' Untranslated Regions , Adenosine/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Mice , Neoplasm Transplantation , Survival Analysis
4.
Cancer Res ; 80(5): 1049-1063, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31888886

ABSTRACT

Wnt/ß-catenin signaling activates the transcription of target genes to regulate stem cells and cancer development. However, the contribution of epigenetic regulation to this process is unknown. Here, we report that Wnt activation stabilizes the epigenetic regulator KDM4C that promotes tumorigenesis and survival of human glioblastoma cells by epigenetically activating the transcription of Wnt target genes. KDM4C protein expression was upregulated in human glioblastomas, and its expression directly correlated with Wnt activity and Wnt target gene expression. KDM4C was essential for Wnt-induced gene expression and tumorigenesis of glioblastoma cells. In the absence of Wnt3a, protein kinase R phosphorylated KDM4C at Ser918, inducing KDM4C ubiquitination and degradation. Wnt3a stabilized KDM4C through inhibition of GSK3-dependent protein kinase R activity. Stabilized KDM4C accumulated in the nucleus and bound to and demethylated TCF4-associated histone H3K9 by interacting with ß-catenin, promoting HP1γ removal and transcriptional activation. These findings reveal that Wnt-KDM4C-ß-catenin signaling represents a novel mechanism for the transcription of Wnt target genes and regulation of tumorigenesis, with important clinical implications. SIGNIFICANCE: These findings identify the Wnt-KDM4C-ß-catenin signaling axis as a critical mechanism for glioma tumorigenesis that may serve as a new therapeutic target in glioblastoma.


Subject(s)
Brain Neoplasms/genetics , Carcinogenesis/genetics , Epigenesis, Genetic , Glioblastoma/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Wnt Proteins/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , DNA Demethylation , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Histones/genetics , Histones/metabolism , Humans , Protein Stability , Transcription Factor 4/genetics , Transcription, Genetic , Ubiquitination/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
5.
Int J Mol Med ; 43(3): 1395-1405, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30628649

ABSTRACT

Neuromyelitis optica (NMO) is a refractory autoimmune inflammatory disease of the central nervous system without an effective cure. Autologous bone marrow­derived mesenchymal stem cells (BM­MSCs) are considered to be promising therapeutic agents for this disease due to their potential regenerative, immune regulatory and neurotrophic effects. However, little is known about the cytological features of BM­MSCs from patients with NMO, which may influence any therapeutic effects. The present study aimed to compare the proliferation, differentiation and senescence of BM­MSCs from patients with NMO with that of age­ and sex­matched healthy subjects. It was revealed that there were no significant differences in terms of cell morphology or differentiation capacities in the BM­MSCs from the patients with NMO. However, in comparison with healthy controls, BM­MSCs derived from the Patients with NMO exhibited a decreased proliferation rate, in addition to a decreased expression of several cell cycle­promoting and proliferation­associated genes. Furthermore, the cell death rate increased in BM­MSCs from patients under normal culture conditions and an assessment of the gene expression profile further confirmed that the BM­MSCs from patients with NMO were more vulnerable to senescence. Platelet­derived growth factor (PDGF), as a major mitotic stimulatory factor for MSCs and a potent therapeutic cytokine in demyelinating disease, was able to overcome the decreased proliferation rate and increased senescence defects in BM­MSCs from the patients with NMO. Taken together, the results from the present study have enabled the proposition of the possibility of combining the application of autologous BM­MSCs and PDGF for refractory and severe patients with NMO in order to elicit improved therapeutic effects, or, at the least, to include PDGF as a necessary and standard growth factor in the current in vitro formula for the culture of NMO patient­derived BM­MSCs.


Subject(s)
Mesenchymal Stem Cells/metabolism , Neuromyelitis Optica/metabolism , Apoptosis , Biomarkers , Case-Control Studies , Cell Cycle , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cellular Senescence , Gene Expression , Humans , Immunophenotyping , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/pathology , Neuromyelitis Optica/genetics , Neuromyelitis Optica/pathology
6.
Cytokine ; 118: 35-41, 2019 06.
Article in English | MEDLINE | ID: mdl-30017390

ABSTRACT

Among myriads of distinct chemical modification in RNAs, the dynamic, reversible and fine-tuned methylation of N6-methyladenosine (m6A) is the most prevalent modification in eukaryotic mRNAs. This RNA mark is generated by proteins that act as m6A writers and can be reversed by proteins that act as m6A erasers. The RNA m6A modification is also mediated by another group of proteins capable of recognizing m6A that act as m6A readers. The m6A modification exerts direct control over the RNA metabolism including mRNA processing, mRNA exporting, translation initiation, mRNA stability and the biogenesis of long-non-coding RNA (LncRNA), thereby can influence various aspects of cell function. Evidently, m6A is intimately associated with cancer development and progression such as self-renewal capacity of cancer stem cells, proliferation, apoptosis and therapeutic resistance, and immune response. In this review, we will discuss the regulation and function of m6A, the various functions ascribed to these proteins and the emerging concepts that impact our knowledge of these proteins and their roles in the epitranscriptome. Conceivably, m6A may play pivotal roles in cytokine and immune response and carcinogenesis.


Subject(s)
Adenosine/analogs & derivatives , Carcinogenesis/metabolism , Cytokines/metabolism , Neoplasms/metabolism , RNA/metabolism , Adenosine/metabolism , Animals , Carcinogenesis/pathology , Humans , Methylation , Neoplasms/pathology
7.
Cancer Res ; 79(1): 72-85, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30425057

ABSTRACT

Aberrant activation of ß-catenin signaling is a critical driver for tumorigenesis, but the mechanism underlying this activation is not completely understood. In this study, we demonstrate a critical role of ß-catenin signaling in stabilization of enhancer of zeste homolog 2 (EZH2) and control of EZH2-mediated gene repression in oncogenesis. ß-Catenin/TCF4 activated the transcription of the deubiquitinase USP1, which then interacted with and deubiquitinated EZH2 directly. USP1-mediated stabilization of EZH2 promoted its recruitment to the promoters of CDKN1B, RUNX3, and HOXA5, resulting in enhanced enrichment of histone H3K27me3 and repression of target gene expression. In human glioma specimens, expression levels of nuclear ß-catenin, USP1, and EZH2 correlated with one another. Depletion of ß-catenin/USP1/EZH2 repressed glioma cell proliferation in vitro and tumor formation in vivo. Our findings indicate that a ß-catenin-USP1-EZH2 axis orchestrates the interplay between dysregulated ß-catenin signaling and EZH2-mediated gene epigenetic silencing during glioma tumorigenesis. SIGNIFICANCE: These findings identify the ß-catenin-USP1-EZH2 signaling axis as a critical mechanism for glioma tumorigenesis that may serve as a new therapeutic target in glioblastoma.


Subject(s)
Carcinogenesis/pathology , Enhancer of Zeste Homolog 2 Protein/chemistry , Gene Expression Regulation, Neoplastic , Glioma/pathology , Ubiquitin-Specific Proteases/metabolism , beta Catenin/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Glioma/genetics , Glioma/metabolism , Humans , Mice , Mice, Nude , Prognosis , Protein Stability , Survival Rate , Tumor Cells, Cultured , Ubiquitin-Specific Proteases/genetics , Xenograft Model Antitumor Assays , beta Catenin/genetics
8.
J Immunol ; 197(7): 2610-7, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27543617

ABSTRACT

Myasthenia gravis (MG) is a chronic humoral immunity-mediated autoimmune disorder of the neuromuscular junction characterized by muscle weakness. Follicular helper T (Tfh) cells may be the key Th cell subset that promotes MG development, as their major function is helping B cell activation and Ab production. Aberrance of thymus-derived Tfh cells might be implicated in autoimmune diseases including MG; just how circulating Tfh cells, especially those from patients with a normal thymus, contribute to MG pathogenesis remains to be uncovered. In this article, we characterize a population of circulating CD4(+)CXCR5(+)PD-1(+) Tfh cells in ocular and generalized MG patients without thymic abnormalities and demonstrate that the circulating Tfh cells are significantly enriched in generalized MG patients but not in ocular MG patients compared with healthy subjects, whereas a proportion of follicular regulatory T cells decreased in MG patients. In addition, the frequency of plasma cells and B cells was higher and the serum levels of IL-6/IL-21 were also elevated in these MG patients. The activated Tfh1 and Tfh17 in Tfh cells are the major source for IL-21 production in MG patients. A strong correlation between Tfh cells and the plasma cell frequency and anti-acetylcholine receptor Ab titers was evident in generalized MG patients. In particular, we found that Tfh cells derived from MG patients promoted B cells to produce Abs in an IL-21 signaling-dependent manner. Collectively, our results suggest that circulating Tfh cells may act on autoreactive B cells and thus contribute to the development of MG in patients without thymic abnormalities.


Subject(s)
B-Lymphocytes/immunology , Myasthenia Gravis/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Myasthenia Gravis/pathology , Prospective Studies
9.
Sci Rep ; 6: 21890, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26902096

ABSTRACT

Soluble receptor for advanced glycation end products (sRAGE) is an anti-inflammatory factor that mitigates the proinflammatory effects of high mobility group box 1 (HMGB1). The aim of this study was to investigate whether Guillain-Barré syndrome (GBS)-related inflammation are mediated by sRAGE and HMGB1. We measured serum sRAGE, HMGB1, IL-6, and TNF-α levels in 86 patients with GBS and analysed associations between sRAGE or HMGB1 and clinical variables in these subjects. In addition, we determined cerebrospinal fluid sRAGE and HMGB1 levels in a cross-sectional study of 50 patients with GBS who had matched serum samples. We found serum sRAGE levels in patients with the acute motor axonal neuropathy (AMAN) subtype of GBS, but not other subtypes, were significantly lower than those in healthy controls, and were significantly correlated with GBS disability score and Erasmus GBS outcome score, while serum HMGB1, IL-6, and TNF-α levels in all subtypes of GBS were significantly higher than those in healthy controls. Moreover, increased sRAGE levels and decreased HMGB1 levels after treatment were observed. Our results showed that serum sRAGE may be a useful biomarker for inflammation in the AMAN GBS subtype, while HMGB1 may be related to the inflammatory process across all types of GBS.

10.
Circulation ; 132(12): 1104-1112, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26202811

ABSTRACT

BACKGROUND: Inflammatory and immune responses triggered by brain ischemia worsen clinical outcomes of stroke and contribute to hemorrhagic transformation, massive edema, and reperfusion injury associated with intravenous alteplase. We assessed whether a combination of the immune-modulator fingolimod and alteplase is safe and effective in attenuating reperfusion injury in patients with acute ischemic stroke treated within the first 4.5 hours of symptom onset. METHODS AND RESULTS: In this multicenter trial, we randomly assigned 25 eligible patients with hemispheric ischemic stroke stemming from anterior or middle cerebral arterial occlusion to receive alteplase alone and 22 patients to receive alteplase plus oral fingolimod 0.5 mg daily for 3 consecutive days within 4.5 hours of the onset of ischemic stroke. Compared with patients who received alteplase alone, patients who received the combination of fingolimod with alteplase exhibited lower circulating lymphocytes, smaller lesion volumes (10.1 versus 34.3 mL; P=0.04), less hemorrhage (1.2 versus 4.4 mL; P=0.01), and attenuated neurological deficits in National Institute of Health Stroke Scales (4 versus 2; P=0.02) at day 1. Furthermore, restrained lesion growth from day 1 to 7 (-2.3 versus 12.1 mL; P<0.01) with a better recovery at day 90 (modified Rankin Scale score 0-1, 73% versus 32%; P<0.01) was evident in patients given fingolimod and alteplase. No serious adverse events were recorded in all patients. CONCLUSIONS: In this pilot study, combination therapy of fingolimod and alteplase was well tolerated, attenuated reperfusion injury, and improved clinical outcomes in patients with acute ischemic stroke. These findings need to be tested in further clinical trials. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT02002390.


Subject(s)
Edema, Cardiac/prevention & control , Fibrinolytic Agents/therapeutic use , Fingolimod Hydrochloride/therapeutic use , Immunologic Factors/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Stroke/drug therapy , Tissue Plasminogen Activator/therapeutic use , B-Lymphocytes/drug effects , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Fibrinolytic Agents/pharmacology , Fingolimod Hydrochloride/pharmacology , Humans , Immunologic Factors/pharmacology , Magnetic Resonance Imaging , Male , Middle Aged , Pilot Projects , Recovery of Function/drug effects , Stroke/pathology , T-Lymphocytes/drug effects , Tissue Plasminogen Activator/pharmacology , Treatment Outcome
11.
Neurosci Bull ; 31(6): 755-62, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25958190

ABSTRACT

Intracerebral hemorrhage (ICH) leads to high rates of death and disability. The pronounced inflammatory reactions that rapidly follow ICH contribute to disease progression. Our recent clinical trial demonstrated that oral administration of an immune modulator fingolimod restrained secondary injury derived from initial hematoma, but the mechanisms remain unknown. In this study, we aim to investigate the effects of fingolimod on inflammatory mediators and vascular permeability in the clinical trial of oral fingolimod for intracerebral hemorrhage (ICH). The results showed that fingolimod decreased the numbers of circulating CD4(+) T, CD8(+) T, CD19(+) B, NK, and NKT cells and they recovered quickly after the drug was stopped. The plasma ICAM level was decreased and IL-10 was increased by fingolimod. Interestingly, fingolimod protected vascular permeability as indicated by a decreased plasma level of MMP9 and the reduced rT1%. In conclusion, modulation of systemic inflammation by fingolimod demonstrates that it is an effective therapeutic agent for ICH. Fingolimod may prevent perihematomal edema enlargement by protecting vascular permeability.


Subject(s)
Capillary Permeability/drug effects , Cerebral Hemorrhage/drug therapy , Fingolimod Hydrochloride/therapeutic use , Immunosuppressive Agents/therapeutic use , Inflammation Mediators/blood , Cell Separation , Cytokines/biosynthesis , Cytokines/drug effects , Humans , Lymphocytes/drug effects , Single-Blind Method
12.
J Neuroimmunol ; 278: 239-46, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25468778

ABSTRACT

While follicular helper T (Tfh) cells have been shown to be involved in many autoimmune diseases, the association of Tfh cells with the disease activity of neuromyelitis optica spectrum disorders (NMOSDs) remains unclear. In this study, the CD4(+)CXCR5(+)PD-1(+) Tfh cell population in peripheral blood mononuclear cells (PBMCs) obtained from NMOSD patients, age- and gender-matched healthy controls, and multiple sclerosis patients was compared by flow cytometry. The serum levels of IL-21, IL-6, IL-17, TNF-α and IL-10 were analyzed by ELISA assays. We found that in NMOSD, the Tfh cell frequency is higher than that of healthy subjects and multiple sclerosis (MS) patients. There are more Tfh cells in the relapsing stage than the remitting stage of NMOSD, thus demonstrating the close association of the Tfh cell population with disease activity. Methylprednisolone, which is used to control disease relapses, significantly decreased the proportion of Tfh cells in NMOSD patients.


Subject(s)
Cytokines/metabolism , Multiple Sclerosis/pathology , Neuromyelitis Optica/immunology , Neuromyelitis Optica/pathology , T-Lymphocytes, Helper-Inducer/immunology , Adult , Aged , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin G/blood , Leukocytes, Mononuclear , Male , Methylprednisolone/pharmacology , Methylprednisolone/therapeutic use , Middle Aged , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Neuromyelitis Optica/drug therapy , Severity of Illness Index , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/pathology , Young Adult
13.
Mol Med Rep ; 11(1): 489-93, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25351212

ABSTRACT

Rheumatoid arthritis (RA) is a systemic autoimmune disease and collagen-induced arthritis (CIA) is an animal model for RA. Micheliolide (MCL) is a novel compound with strong anti-inflammatory effects. The present study was conducted to evaluate the therapeutic effects of MCL on RA. Mice were randomly divided into four groups and the CIA model mice were treated with methotrexate (MTX), MCL and dimethyl sulfoxide. A score associated with the severity of arthritis was assigned on alternate days from the 22nd day for 60 days. Histopathological changes and the serum levels of cytokines were measured on day 85. The results demonstrated that the MCL treatment group had arthritis scores lower than the CIA group and higher than the MTX group; compared with the CIA group, MCL and MTX significantly reduced the swelling of the paws and suppressed the degeneration of articular cartilage. Expression levels of macrophage colony-stimulating factor (M-CSF), tissue inhibitors of metalloproteinases-1 (TIMP-1) and complement component 5a (C5/C5a) were lower in the mice with arthritis compared with normal mice, however, following treatment with MCL and MTX, all the mice exhibited significant recovery to differing degrees. Unlike the MTX group, the MCL group failed to recover the level of soluble intercellular adhesion molecule-1. In addition, the cytokine of B-lymphocyte chemoattractant (BLC) solely presented in the MCL group. These results suggest that MCL may be considered for use as a novel therapeutic treatment against RA and that changes in the expression of cytokines C5/C5a, TIMP-1, M-CSF and BLC may underlie the mechanism by which MCL effects changes in this disease.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Experimental , Arthritis, Rheumatoid/drug therapy , Sesquiterpenes, Guaiane/pharmacology , Animals , Antirheumatic Agents/administration & dosage , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Female , Immunosuppressive Agents/pharmacology , Male , Methotrexate/pharmacology , Mice , Sesquiterpenes, Guaiane/administration & dosage
14.
Int J Biochem Cell Biol ; 54: 111-21, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25043687

ABSTRACT

In our previous study, we have found that the tumor multidrug resistance mediated by P-glycoprotein could be reversed by sustained intracellular acidification through down-regulating the multidrug resistance gene 1 mRNA and P-glycoprotein expression. However, the molecular events linking the intracellular acidification and the regulation of P-glycoprotein remain unclear. In the present study, the molecular pathways involved in the regulation of P-glycoprotein expression by the intracellular acidification were investigated. We found that the P-glycoprotein expression was down-regulated by the intracellular acidification through inhibition of p38 mitogen-activated protein kinase (MAPK) and the activation of c-Jun N-terminal kinase (JNK) in the resisitant K562/DOX cells. In the sensitive K562 and HL60 cell lines, the changes of the p38 MAPK expression after the acidification are not as obvious as that of K562/DOX cells, but the activation of extracellular signal-regulated kinase (ERK) is also observed, which indicates that the down-regulation of p38 MAPK by the intracellular acidification might be the resistant cell line specific. Blockade of ERK and JNK signaling by the inhibitors or RNA interference increased p38MAPK activities suggesting that cross-talk within MAPKs is also important for this response. Our study provides the first direct evidence that the reversal of P-glycoprotein-mediated multidrug resistance by intracellular acidification is mediated by the crosstalk of MAPK signaling pathways.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Acids/metabolism , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Antibiotics, Antineoplastic/pharmacology , Blotting, Western , Cell Physiological Phenomena , Cell Proliferation/drug effects , Down-Regulation , Doxorubicin/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , HL-60 Cells , Humans , Hydrogen-Ion Concentration , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , K562 Cells , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
15.
Cell Physiol Biochem ; 33(1): 185-94, 2014.
Article in English | MEDLINE | ID: mdl-24481225

ABSTRACT

BACKGROUND/AIMS: Na+/H+ exchanger 1 (NHE1) is an important regulator of intracellular pH (pHi). High pHi is required for cell proliferation and differentiation. Our previous study has proven that the pHi of mesenchymal stem cells is higher than that of normal differentiated cells and similar to tumor cells. NHE1 is highly expressed in both mesenchymal stem cells and tumor cells. Targeted inhibition of NHE1 could induce differentiation of K562 leukemia cells. In the present paper we explored whether inhibition of NHE1 could induce differentiation of mesenchymal stem cells. METHODS: MSCs were obtained from human umbilical cord and both the surface phenotype and functional characteristics were analyzed. Selective NHE1 inhibitor cariporide was used to treat human umbilical cord-derived mesenchymal stem cells (hUC-MSCs). The pHi and the differentiation of hUC-MSCs were compared upon cariporide treatment. The putative signaling pathway involved was also explored. RESULTS: The pHi of hUC-MSCs was decreased upon cariporide treatment. Cariporide up-regulated the osteogenic differentiation of hUC-MSCs while the adipogenic differentiation was not affected. For osteogenic differentiation, ß-catenin expression was up-regulated upon cariporide treatment. CONCLUSION: Decreased pHi induced by cariporide differentially contributes to hUC-MSCs differentiation.


Subject(s)
Cell Differentiation/drug effects , Guanidines/pharmacology , Intracellular Space/metabolism , Mesenchymal Stem Cells/cytology , Sulfones/pharmacology , Umbilical Cord/cytology , Adipogenesis/drug effects , Cation Transport Proteins/metabolism , Cell Death/drug effects , Gene Knockdown Techniques , Humans , Hydrogen-Ion Concentration , Intracellular Space/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Phenotype , RNA, Small Interfering/metabolism , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/metabolism , Up-Regulation/drug effects , beta Catenin/metabolism
16.
Leuk Lymphoma ; 55(9): 2135-40, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24188478

ABSTRACT

CCAAT/enhancer binding protein homologous protein (CHOP) expression increases when Na(+)-H(+) exchanger 1 (NHE1) is inhibited. Endoplasmic reticulum (ER) stress has been shown to trigger tumor cell death through CHOP. We therefore hypothesized that NHE1 activity correlates with ER stress and confers pharmaceutical potential to NHE1 inhibitor as an anti-tumor agent. The present study showed that treatment with the NHE1 inhibitor cariporide led to ER stress-induced up-regulation of the death receptor 5 (DR5) which is mediated by CHOP at the transcriptional level. We also determined that ER stress-induced Janus kinase (JNK) activation was responsible for the modulation of CHOP. Combining cariporide with tumor necrosis factor related apoptosis-inducing ligand (TRAIL) led to a significantly enhanced level of apoptosis that was abrogated by siRNA silencing of CHOP. This study provides a potential mechanistic rationale for the use of NHE1 inhibitor in combination with DR5 agonists to induce apoptosis in leukemia.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Endoplasmic Reticulum Stress , Guanidines/pharmacology , Leukemia/genetics , Leukemia/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Sulfones/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Humans , Transcription Factor CHOP/genetics , Transcriptional Activation
17.
Cancer Cell Int ; 13(1): 117, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24257075

ABSTRACT

BACKGROUND: Chronic myeloid leukemia is a clonal myeloproliferative disorder disease in which BCR/ABL plays an important role as an oncoprotein and molecular target. Despite the success of targeted therapy using tyrosine kinase inhibitors, CML remains largely incurable, most likely due to the treatment resistance after firstly chemical therapy. So know well the unique molecular pathway of CML is very important. METHODS: The expressions of CD44 in different leukemia patients and cell lines were detected by real-time PCR and western blotting. The effects of CD44 on proliferation of K562 cells were determined using the MTT and colony formation assays, and even in a nude mouse transplantation model. Then, the cell cycle changes were detected by flow cytometric analysis and the early apoptosis of cells was detected by the annexin V/propidium iodide double-staining assay. The expressions of the cycles and apoptosis-related proteins p21, Cyclin D1 and Bcl-2 were analyzed by western blot and real-time PCR assay. Finally, the decreased nuclear accumulation of ß-catenin was detected by western blotting and immunefluorescence. RESULTS: Firstly, we showed that CD44 expression was increased in several kinds of leukemia patients and K562 cells. By contrast, the down-regulation of CD44 resulted in decreased proliferation with a G0/G1 arrest of cell cycle in K562 cells according to the MTT assay and the flow cytometric analysis. And no significant induction of both the early and late phases of apoptosis was shown by the annexin V-FITC and PI staining. During this process, p21 and cyclin D1 are the major causes for cell cycle arrest. In addition, we found CD44 down-regulation decreased the expression of ß-catenin and increased the expression of phosphorylated ß-catenin. The instability of Wnt/ß-catenin pathway induced by increased expression of p-ß-catenin resulted in a decreased nuclear accumulation in CD44 silenced K562 cells. In the nude mouse transplantation model, we also found the same results. CONCLUSIONS: These results show that K562 cells depend to a greater extent on CD44 for proliferation, and CD44 down-regulation may induce a cell cycle arrest through Wnt/ß-catenin pathway. CD44 blockade may be beneficial in therapy of CML.

18.
Leuk Res ; 37(11): 1583-91, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24125838

ABSTRACT

CUEDC2, a newly reported protein, has been found to be ubiquitously expressed in human tissues and repress NF-κB activity. To study the role of CUEDC2 in chronic myeloid leukemia (CML), we explored the function of CUEDC2 in CML cells through using the CML cell line K562 and its imatinib resistant cells K562/G01. K562 cells expressed a relatively higher level of CUEDC2 compared to K562/G01 cells. Knockdown of CUEDC2 in K562 cells resulted in decreased cell apoptosis after imatinib treatment; when CUEDC2 was overexpressed in K562/G01 cells, imatinib induced more cell apoptosis. By analyzing the activity of NF-κB, the results indicated a negative association between the expression of CUEDC2 and NF-κB signaling pathway in these CML cells. Our data suggested that the expression level of CUEDC2 has an inverse correlation with imatinib resistance and activity of NF-κB signaling pathway in CML cells, CUEDC2 could regulate imatinib sensitivity in CML cells at least partially through NF-κB signaling pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Benzamides/pharmacology , Carrier Proteins/metabolism , Drug Resistance, Neoplasm , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Membrane Proteins/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Adaptor Proteins, Signal Transducing , Blotting, Western , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Proliferation , Flow Cytometry , Fluorescent Antibody Technique , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , NF-kappa B/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Tumor Cells, Cultured
19.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 21(4): 861-5, 2013 Aug.
Article in Chinese | MEDLINE | ID: mdl-23998575

ABSTRACT

This study was aimed to investigate the influence of short hairpin RNA (shRNA) on proliferation of human leukemia cell line THP-1. The shRNA targeting the site 732-752 of DOT1L mRNA was designed and chemically synthesized, then a single-vector lentiviral, tet-inducible shRNA-DOT1L system (Plko-Tet-On) was generated. Thereafter, the THP-1 cells with lentivirus were infected to create stable cell line with regulatable shRNA expression. The expression of DOT1L in the THP-1 cell line was assayed by RT-PCR. Effect of shRNA-DOT1L on the proliferation of THP-1 cells was detected with MTT method,and the change of colony forming potential of THP-1 cells was analyzed by colony forming unit test. Cell cycle distribution was tested by flow cytometry. The results indicated that the expression of DOT1L was statistically lower than that in the control groups. The proliferation and colony forming capacity of THP-1 cells were significantly inhibited. The percentage of cells at G0/G1 phase increased in THP-1/shRNA cells treated with Dox while the percentage of cells at S phase significantly decreased as compared with that in the control group. It is concluded that the shRNA targeting DOT1L can effectively inhibit the proliferation of acute monocytic leukemia cell line THP-1.


Subject(s)
Cell Proliferation , Leukemia, Monocytic, Acute/genetics , Methyltransferases/genetics , RNA, Small Interfering , Cell Line, Tumor , Genetic Vectors , Humans , Lentivirus/genetics
20.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 21(1): 45-8, 2013 Feb.
Article in Chinese | MEDLINE | ID: mdl-23484689

ABSTRACT

This study was purposed to explore the changes of possible angiogenetic factors other than VEGF after inhibition of NHE1 and their related mechanisms. The K562 cells were treated by NHE1 specific inhibitor cariporide, the angiogenesis factors after inhibition of NHE1 were screened by using protein chip, the IL-8 expression level after cariporide treatment was detected by real-time quantitative PCR; the K562 cells with stable interference of NHE1 were constructed, the IL-8 expression level after interference of NHE1 was detected by real-time quantitative PCR; the p38 phosphorylation level in K562 cells treated with cariporide was detected by Western blot. After treatment of K562 cells with p38 inhibitor SB203580, the IL-8 expression level was decreased by real-time quantitative PCR. The results of protein chip showed that IL-8 expression decreased after cariporide treatment. Real-time quantitative PCR confirmed this inhibitory effect. The p38 phosphorylation level increased after cariporide treatment. The down-regulation of IL-8 expression induced by cariporide treatment was partially restored after K562 cells were treated with p38 inhibitor SB203580. It is concluded that the inhibition of NHE1 can inhibit IL-8 expression through up-regulation of p38 phosphorylation.


Subject(s)
Cation Transport Proteins/antagonists & inhibitors , Interleukin-8/metabolism , Sodium-Hydrogen Exchangers/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism , Down-Regulation , Guanidines/pharmacology , Humans , Imidazoles/pharmacology , K562 Cells , Phosphorylation/drug effects , Pyridines/pharmacology , Sodium-Hydrogen Exchanger 1 , Sulfones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...