Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Biochem Biophys Res Commun ; 703: 149659, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38382358

ABSTRACT

Utilizing RNA sequence (RNA-Seq) splice junction data from a cohort of 1841 B-cell acute lymphoblastic leukemia (B-ALL) patients we define transcriptionally distinct isoforms of ARID5B, a risk-associated gene identified in genome wide association studies (GWAS), which associate with disease survival. Short (S) and long (L) ARID5B transcripts, which differ in an encoded BAH-like chromatin interaction domain, show remarkable correlation to the isoform splicing pattern. Testing of the ARID5B proximal promoter of the S & L isoforms indicated that both are functionally independent in luciferase reporter assays. Increased short isoform expression is associated with decreased event-free and overall survival. The abundance of short and long transcripts strongly correlates to B-ALL prognostic stratification, where B-ALL subtypes with poor outcomes express a higher proportion of the S-isoform. These data demonstrate that the analysis of independent promoters and alternative splicing events are essential for improved risk stratification and a more complete understanding of disease pathology.


Subject(s)
Alternative Splicing , Genome-Wide Association Study , Humans , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splicing , Base Sequence , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Prostate ; 82(1): 97-106, 2022 01.
Article in English | MEDLINE | ID: mdl-34633095

ABSTRACT

BACKGROUND: Interleukin-6 (IL-6) is a pleiotropic cytokine that confers androgen-independence and aggressiveness in prostate cancer (PCa); however, the molecular mechanisms regulating IL-6 expression remain unclear. The expression of ARID5A, an AT-rich interaction domain (ARID) DNA-binding motif-containing transcription factor is positively correlated with IL-6 expression in human PCa. We, therefore, hypothesized that ARID5A could regulate IL-6 expression in PCa. METHODS: The relationship between ARID5A and IL-6 in PCa patients was analyzed using statistical analyses of multiple clinical microarray data sets. To investigate whether ARID5A regulates IL-6 expression, CRISPR-driven ARID5A knockout clones were established in DU145 and PC-3 cells. RESULTS: Analysis of three microarray data sets showed a positive correlation between ARID5A and IL-6 expression. The expression of IL-6 in ARID5A knockout clones was significantly reduced compared with control clones in both PCa cell lines. Knockout of ARID5A did not result in any loss of IL-6 mRNA stability. Instead, we observed a significant decrease in the occupancy of both active RNA Polymerase II and the active histone mark, H3K4me3 at the IL-6 transcriptional start site in ARID5A knockout PCa cells, suggesting a role for transcriptional regulation. CONCLUSIONS: Our study demonstrated that loss of ARID5A downregulates the expression of IL-6 at the transcriptional level.


Subject(s)
DNA-Binding Proteins , Interleukin-6 , Prostatic Neoplasms , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation , Gene Expression Regulation , Gene Knockout Techniques/methods , Histone Code , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Male , PC-3 Cells , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Array Analysis/methods , RNA Stability , Transcription Factors
3.
Biol Res ; 53(1): 45, 2020 Oct 06.
Article in English | MEDLINE | ID: mdl-33023658

ABSTRACT

BACKGROUND: Skeletal muscle has an important role in regulating whole-body energy homeostasis, and energy production depends on the efficient function of mitochondria. We demonstrated previously that AT-rich interactive domain 5b (Arid5b) knockout (Arid5b-/-) mice were lean and resistant to high-fat diet (HFD)-induced obesity. While a potential role of Arid5b in energy metabolism has been suggested in adipocytes and hepatocytes, the role of Arid5b in skeletal muscle metabolism has not been studied. Therefore, we investigated whether energy metabolism is altered in Arid5b-/- skeletal muscle. RESULTS: Arid5b-/- skeletal muscles showed increased basal glucose uptake, glycogen content, glucose oxidation and ATP content. Additionally, glucose clearance and oxygen consumption were upregulated in Arid5b-/- mice. The expression of glucose transporter 1 (GLUT1) and 4 (GLUT4) in the gastrocnemius (GC) muscle remained unchanged. Intriguingly, the expression of TBC domain family member 1 (TBC1D1), which negatively regulates GLUT4 translocation to the plasma membrane, was suppressed in Arid5b-/- skeletal muscle. Coimmunofluorescence staining of the GC muscle sections for GLUT4 and dystrophin revealed increased GLUT4 localization at the plasma membrane in Arid5b-/- muscle. CONCLUSIONS: The current study showed that the knockout of Arid5b enhanced glucose metabolism through the downregulation of TBC1D1 and increased GLUT4 membrane translocation in skeletal muscle.


Subject(s)
DNA-Binding Proteins/genetics , GTPase-Activating Proteins/genetics , Glucose , Muscle, Skeletal , Transcription Factors/genetics , Animals , Biological Transport , Down-Regulation , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 4/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/metabolism
4.
FASEB J ; 32(4): 1868-1879, 2018 04.
Article in English | MEDLINE | ID: mdl-29196500

ABSTRACT

The AT-rich interaction domain (ARID) family of proteins regulates gene expression, development, and differentiation. Although Arid5b has important functions in adipogenesis and chondrogenesis, the role of Arid5b in skeletal muscle myogenesis has not been investigated. Therefore, we isolated primary skeletal muscle cells from Arid5b+/+ and Arid5b-/- mice and characterized differentiation in these cells. We found that Arid5b-/- primary skeletal muscle cells showed differentiation defects and impaired sarcomeric assembly. Microarray analysis revealed down-regulation of the prostanoid biosynthesis pathway in Arid5b-/- myoblasts, including the genes encoding cyclooxygenase (COX)-1 ( Ptgs1) and prostaglandin (PG)I synthase ( Ptgis). Down-regulation of COX-1 and PGI synthase was confirmed by real-time PCR and Western blot analyses. Correspondingly, the production of PGI2, as measured by ELISA, was reduced in Arid5b-/- cells relative to Arid5b+/+ cells. Boyden chamber assays showed that migration was increased but chemotaxis was impaired in Arid5b-/- cells. Myoblast fusion was also inhibited in Arid5b-/- cells compared with Arid5b+/+ cells. Treatment with the PGI2 analog iloprost rescued the defects in myotube formation, migration, and fusion. These results demonstrate that Arid5b has a novel and essential role in skeletal muscle differentiation by regulating PGI2 production.-Murray, J., Whitson, R. H., Itakura, K. Reduced prostaglandin I2 signaling in Arid5b-/- primary skeletal muscle cells attenuates myogenesis.


Subject(s)
DNA-Binding Proteins/genetics , Epoprostenol/pharmacology , Muscle Development , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism , Transcription Factors/genetics , Animals , Cells, Cultured , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Myoblasts/cytology , Myoblasts/drug effects , Signal Transduction
5.
Prostate ; 78(16): 1238-1247, 2018 12.
Article in English | MEDLINE | ID: mdl-30027545

ABSTRACT

BACKGROUND: The androgen receptor (AR) is one of the most important and dynamically regulated factors in prostate cancer (PCa) progression. Despite the importance of AR expression regulation, the precise mechanisms are not fully understood. ARID5B, an AT-rich interaction domain DNA-binding motif-containing transcription factor, is expressed higher in primary PCa than normal prostate, and correlated with AR expression. We therefore hypothesized that ARID5B could regulate AR expression. METHODS: Correlation between AR and ARID5B expression was analyzed using publicly and commercially available microarray data. To examine the role of ARID5B in AR expression, ARID5B was knocked down in VCaP and LNCaP cells, then mRNA and protein levels of AR were measured and an in vitro cell proliferation assay was performed. Chromatin immunoprecipitation was performed to further examine molecular mechanisms. RESULTS: Knockdown of ARID5B suppressed the AR mRNA and protein expression in VCaP and LNCaP cells and decreased in vitro cell proliferation. Suppression of ARID5B decreased the occupancy of active RNA polymerase II in the AR promoter, indicating that ARID5B regulates AR transcription. The active histone mark, H3K4me3, occupancy was decreased with ARID5B knockdown. CONCLUSION: Our study revealed that AR transcription is positively regulated by ARID5B through H3K4me3 recruitment in the AR promoter. Our findings reveal novel mechanisms of AR transcription, which is dynamically regulated in prostate tumor progression.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation , Disease Progression , Humans , Male , Promoter Regions, Genetic , Prostatic Neoplasms/pathology , Transcription, Genetic
7.
Cancer Cell Int ; 14: 45, 2014.
Article in English | MEDLINE | ID: mdl-24932134

ABSTRACT

BACKGROUND: We have reported that the glucosamine suppressed the proliferation of the human prostate carcinoma cell line DU145 through inhibition of STAT3 signaling. DU145 cells autonomously express IL-6 and the IL-6/STAT3 signaling is activated. IL-6 receptor subunits are subject to N-glycosylation, a posttranslational modification which is important for protein stability and function. We speculated that the inhibition of STAT3 phosphorylation by glucosamine might be a functional consequence of the reduced N-glycosylation of gp130. METHODS: The human prostate cancer cell lines DU145 and PC-3 and human melanoma cell line A2058 were used in this study. Glucosamine effects on N-glycosylation of glycoproteins were determined by Western blot analysis. IL-6 binding to DU145 cells was analyzed by flow cytometry. The cell proliferation suppression was investigated by colorimetric Janus green staining method. RESULTS: In DU145 cells glucosamine reduced the N-glycosylation of gp130, decreased IL-6 binding to cells and impaired the phosphorylation of JAK2, SHP2 and STAT3. Glucosamine acts in a very similar manner to tunicamycin, an inhibitor of protein N-glycosylation. Glucosamine-mediated inhibition of N-glycosylation was neither protein- nor cell-specific. Sensitivity of DU145, A2058 and PC-3 cells to glucosamine-induced inhibition of N-glycosylation were well correlated to glucosamine cytotoxicity in these cells. CONCLUSION: Our results suggested that the glucosamine-induced global inhibition of protein N-glycosylation might be the basic mechanism underlying its multiple biochemical and cellular effects.

8.
Oncotarget ; 14: 723-737, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37477523

ABSTRACT

Patients with cancer cachexia have a poor prognosis and impaired quality of life. Numerous studies using preclinical models have shown that inflammatory cytokines play an important role in the development of cancer cachexia; however, no clinical trial targeting cytokines has been successful. Therefore, it is essential to identify molecular mechanisms to develop anti-cachexia therapies. Here we identified the uncharacterized transcript KIAA0930 as a candidate cachexic factor based on analyses of microarray datasets and an in vitro muscle atrophy assay. While conditioned media from pancreatic, colorectal, gastric, and tongue cancer cells caused muscle atrophy in vitro, conditioned medium from KIAA0930 knockdown cells did not. The PANC-1 orthotopic xenograft study showed that the tibialis anterior muscle weight and cross-sectional area were increased in mice bearing KIAA0930 knockdown cells compared to control mice. Interestingly, KIAA0930 knockdown did not cause consistent changes in the secretion of inflammatory cytokines/chemokines from a variety of cancer cell lines. An initial characterization experiment showed that KIAA0930 is localized in the cytosol and not secreted from cells. These data suggest that the action of KIAA0930 is independent of the expression of cytokines/chemokines and that KIAA0930 could be a novel therapeutic target for cachexia.


Subject(s)
Neoplasms , Quality of Life , Humans , Animals , Mice , Muscle, Skeletal/metabolism , Neoplasms/metabolism , Muscular Atrophy/metabolism , Cachexia/metabolism , Cytokines/metabolism , Phenotype , Chemokines/metabolism
9.
Front Immunol ; 14: 1170475, 2023.
Article in English | MEDLINE | ID: mdl-37483604

ABSTRACT

During B cell development in bone marrow, large precursor B cells (large Pre-B cells) proliferate rapidly, exit the cell cycle, and differentiate into non-proliferative (quiescent) small Pre-B cells. Dysregulation of this process may result in the failure to produce functional B cells and pose a risk of leukemic transformation. Here, we report that AT rich interacting domain 5B (ARID5B), a B cell acute lymphoblastic leukemia (B-ALL) risk gene, regulates B cell development at the Pre-B stage. In both mice and humans, we observed a significant upregulation of ARID5B expression that initiates at the Pre-B stage and is maintained throughout later stages of B cell development. In mice, deletion of Arid5b in vivo and ex vivo exhibited a significant reduction in the proportion of immature B cells but an increase in large and small Pre-B cells. Arid5b inhibition ex vivo also led to an increase in proliferation of both Pre-B cell populations. Metabolic studies in mouse and human bone marrow revealed that fatty acid uptake peaked in proliferative B cells then decreased during non-proliferative stages. We showed that Arid5b ablation enhanced fatty acid uptake and oxidation in Pre-B cells. Furthermore, decreased ARID5B expression was observed in tumor cells from B-ALL patients when compared to B cells from non-leukemic individuals. In B-ALL patients, ARID5B expression below the median was associated with decreased survival particularly in subtypes originating from Pre-B cells. Collectively, our data indicated that Arid5b regulates fatty acid metabolism and proliferation of Pre-B cells in mice, and reduced expression of ARID5B in humans is a risk factor for B cell leukemia.


Subject(s)
Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Transcription Factors , Animals , Humans , Mice , Cell Proliferation , DNA-Binding Proteins/metabolism , Fatty Acids , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cells, B-Lymphoid/metabolism , Transcription Factors/metabolism
10.
Front Endocrinol (Lausanne) ; 13: 1083311, 2022.
Article in English | MEDLINE | ID: mdl-36743919

ABSTRACT

Emerging evidence suggests that AT-Rich Interaction Domain 5b (Arid5b) may play a role in energy metabolism in various tissues. To study the metabolic function of Arid5b in skeletal muscle, we generated skeletal muscle-specific Arid5b knockout (Arid5b MKO) mice. We found that Arid5b MKO skeletal muscles preferentially utilized fatty acids for energy generation with a corresponding increase in FABP4 expression. Interestingly, in Arid5b MKO mice, the adipose tissue weight decreased significantly. One possible mechanism for the decrease in adipose tissue weight could be the increase in phospho-HSL and HSL expression in white adipose tissue. While glucose uptake increased in an insulin-independent manner in Arid5b MKO skeletal muscle, glucose oxidation was reduced in conjunction with downregulation of the mitochondrial pyruvate carrier (MPC). We found that glucose was diverted into the pentose phosphate pathway as well as converted into lactate through glycolysis for export to the bloodstream, fueling the Cori cycle. Our data show that muscle-specific deletion of Arid5b leads to changes in fuel utilization in skeletal muscle that influences metabolism in other tissues. These results suggest that Arid5b regulates systemic metabolism by modulating fuel selection.


Subject(s)
DNA-Binding Proteins , Gene Deletion , Glucose , Muscle, Skeletal , Transcription Factors , Animals , Mice , Adipose Tissue/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Glucose/metabolism , Insulin/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Mol Metab ; 66: 101612, 2022 12.
Article in English | MEDLINE | ID: mdl-36243318

ABSTRACT

OBJECTIVE: Adipose tissue is the largest endocrine organ. When activated by cancer cells, adipocytes secrete adipocytokines and release fatty acids, which are then transferred to cancer cells and used for structural and biochemical support. How this metabolic symbiosis between cancer cells and adipocytes affects skeletal muscle and thermogenesis during cancer cachexia is unknown. Cancer cachexia is a multiorgan syndrome and how the communication between tissues is established has yet to be determined. We investigated adipose tissue secretory factors and explored their role in crosstalk of adipocytes, muscle, and tumor during pancreatic cancer cachexia. METHODS: We used a pancreatic cancer cachexia mouse model generated by syngenic implantation of pancreatic ductal adenocarcinoma (PDAC) cells (KPC) intraperitoneally into C57BL/6 mice and Lcn2-knockout mice. For in vitro studies, adipocytes (3T3-L1 and primary adipocytes), cachectic cancer cells (Panc0203), non-cachectic cancer cells (Du145 cells), and skeletal muscle cells (C2C12 myoblasts) were used. RESULTS: To identify molecules involved in the crosstalk of adipose tissue with muscle and tumors, we treated 3T3-L1 adipocytes with conditioned medium (CM) from cancer cells. Upon screening the secretomes from PDAC-induced adipocytes, several adipocytokines were identified, including lipocalin 2 (Lcn2). We investigated Lcn2 as a potential mediator of cachexia induced by adipocytes in response to PDAC. During tumor progression, mice exhibited a decline in body weight gain, which was accompanied by loss of adipose and muscle tissues. Tumor-harboring mice developed drastic hypothermia because of a dramatic loss of fat in brown adipose tissue (BAT) and suppression of the thermogenesis pathway. We inhibited Lcn2 with an anti-Lcn2 antibody neutralization or genomic ablation in mice. Lcn2 deficiency significantly improved body temperature in tumor-bearing mice, which was supported by the increased expression of Ucp1 and ß3-adrenergic receptor in BAT. In addition, Lcn2 inhibition abrogated the loss of fat and muscle in tumor-bearing mice. In contrast to tumor-bearing WT mice, the corresponding Lcn2-knockout mice showed reduced ATGL expression in iWAT and decreased the expression of muscle atrophy molecular markers MuRF-1 and Fbx32. CONCLUSIONS: This study showed that Lcn2 is causally involved in the dysregulation of adipose tissue-muscle-tumor crosstalk during pancreatic cancer cachexia. Therapeutic targets that suppress Lcn2 may minimize the progression of cachexia.


Subject(s)
Cachexia , Hypothermia , Lipocalin-2 , Pancreatic Neoplasms , Animals , Mice , Adipocytes/metabolism , Adipokines/metabolism , Adipose Tissue, Brown/metabolism , Cachexia/etiology , Cachexia/metabolism , Hypothermia/complications , Hypothermia/metabolism , Lipocalin-2/genetics , Lipocalin-2/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms
12.
Mol Cell Endocrinol ; 528: 111246, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33757861

ABSTRACT

Mice with global deletion of Arid5b expression are lean and resistant to diet-induced obesity, and Arid5b is required for adipogenesis in a variety of in vitro models. To determine whether the lean phenotype of Arid5b-/- mice can be explained by its absence in adipose tissues, we generated mice with Fabp4-mediated ablation of Arid5b. Arid5b expression was ablated in adipocytes, from Fabp4-CREpos; Arid5bFLOX/FLOX (FSKO) mice. FSKO mice were not lean when maintained on standard chow, but males were resistant to weight gains when placed on high-fat diets (HFD). This was mainly due to decreased lipid accumulation in subcutaneous (inguinal) white adipose tissue (IWAT), and the liver. Lipid accumulation proceeded normally in gonadal WAT (GWAT) and glucose intolerance developed to the same degree in FSKO and WT controls when subjected to HFD. CD68-positive macrophages were also significantly reduced in both inguinal and gonadal fat depots. RNA-Seq analysis of IWAT adipocytes from FSKO mice on HFD revealed significant decreases in the expression of genes associated with inflammation. Although Arid5b expression was normal in livers of FSKO mice, tissue weight gains and triglyceride accumulation, and expression of genes involved in lipid metabolism were markedly reduced in livers of FSKO mice on HFD. These results suggest that Arid5b plays a critical role in lipid accumulation in specific WAT depots, and in the inflammatory signaling from WAT depots to liver that lead to lipid accumulation and hepatic steatosis.


Subject(s)
DNA-Binding Proteins/genetics , Diet, High-Fat/adverse effects , Fatty Acid-Binding Proteins/genetics , Fatty Liver/prevention & control , Obesity/prevention & control , Transcription Factors/genetics , Animals , Disease Models, Animal , Fatty Liver/genetics , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation/drug effects , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/chemically induced , Obesity/genetics , Obesity/metabolism , Sequence Analysis, RNA , Subcutaneous Fat/metabolism
13.
Biochem Biophys Res Commun ; 391(1): 277-81, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19913508

ABSTRACT

Mice lacking modulator recognition factor-2 (Mrf-2; ARID5B) have less fat in brown and white adipose tissues, partly because of a defect in adipocyte differentiation. We have also shown that knockdown of Mrf-2 decreases the expression of the adipogenic transcription factors C/EBPalpha and PPARgamma, and inhibits adipogenesis in 3T3-L1 preadipocytes. Since these transcription factors may also contribute to the maintenance of adipocyte function, we examined the effects of siRNA targeted to Mrf-2 on triglyceride metabolism in mature 3T3-L1-derived adipocytes. As it did in differentiating adipocytes, knockdown of Mrf-2 decreased the expression of both C/EBPalpha and PPARgamma. Knockdown of Mrf-2 also activated both lipolysis and triglyceride synthesis, and caused a significant increase in the ratio of glycerol release to free fatty acid release. This suggests that knockdown of Mrf-2 increases the rate of fatty acid recycling in 3T3-L1-derived adipocytes. Continual cycling of fatty acids through lipolysis and triglyceride synthesis could lead to dissipation of energy. Therefore, the activation of such a futile cycle via the suppression of Mrf-2 could be an effective treatment for obesity and diabetes.


Subject(s)
Adipocytes/metabolism , DNA-Binding Proteins/metabolism , Lipolysis/genetics , Transcription Factors/metabolism , Triglycerides/metabolism , 3T3-L1 Cells , Animals , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Fatty Acids, Nonesterified/metabolism , Gene Knockdown Techniques , Mice , PPAR gamma/antagonists & inhibitors , PPAR gamma/biosynthesis , Transcription Factors/genetics
14.
Cancer Cell Int ; 9: 25, 2009 Sep 10.
Article in English | MEDLINE | ID: mdl-19744341

ABSTRACT

BACKGROUND: Glucosamine is known as a toxic agent for several malignant cell lines and transplanted tumors with little toxicity to normal host tissues. However, the mechanisms underlying anticancer activity of glucosamine are poorly understood. To study the mechanisms, the human prostate cancer DU145 cells were used for the model. RESULTS: Glucosamine at concentration 2 mM suppressed proliferation and induced death of DU145 cells. Detailed analysis showed that glucosamine decreased DNA synthesis, arrested cell cycle at G1 phase and induced apoptosis. The effects of glucosamine were associated with up-regulation of p21waf1/cip, a CDK inhibitor. Our further studies identified glucosamine as an inhibitor of signal transducer and activator of transcription (STAT) 3 which is constitutively activated in many cancer cells including DU145 cells. Glucosamine inhibited phosphorylation of STAT3 at the Tyr705 residue and as a result, reduced STAT3 DNA binding and transcriptional activities. Indeed, the expression of apoptosis inhibitor survivin, which is well known target of STAT3, was suppressed. Contrary to DU145 cells, glucosamine did not affect proliferation of other human prostate cancer PC-3 and C4-2B cells, in which STAT 3 signal pathway is not constitutively active. CONCLUSION: Our data identifies glucosamine as a suppressor of STAT3 signaling and suggests that anticancer activity of glucosamine may be attributed to the suppression of STAT3 activity. Potential application of glucosamine for the treatment of tumors with constitutively active STAT3 is proposed.

15.
Mol Endocrinol ; 22(2): 441-53, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17962384

ABSTRACT

Previous study showed that mice lacking modulator recognition factor-2 (Mrf-2) were lean, with significant decreases in white adipose tissue. One postulated mechanism for the lean phenotype in Mrf-2 knockout mice is a defect in adipogenesis. In order to investigate this further, we examined the effects of Mrf-2 deficiency on adipogenesis in vitro. In mouse fibroblasts (MEFs) derived from Mrf-2(-/-) embryos, and in 3T3-L1 cells after knockdown of Mrf-2 by small interference RNA (siRNA) there was a potent inhibition of hormone-induced lipid accumulation, and significant decreases in the expression of the adipogenic transcription factors CCAAT/enhancer-binding protein (C/EBP) alpha and peroxisome proliferator-activated receptor-gamma and the mature adipocyte genes they control. Transduction of Mrf-2(-/-) MEFs with a retroviral vector expressing the longer Mrf-2 splice variant (Mrf-2B) stimulated both gene expression and lipid accumulation. Because 3T3-L1 cells are committed to the adipocyte lineage, we used this simpler model system to examine the effects of Mrf-2 deficiency on adipocyte maturation. Analyses of both mRNA and protein revealed that knockdown of Mrf-2 in 3T3-L1 cells prolonged the expression of C/EBP homologous protein-10, a dominant-negative form of C/EBP. Consistent with these findings, suppression of Mrf-2 also inhibited the DNA-binding activity of C/EBPbeta. These data suggest that Mrf-2 facilitates the induction of the two key adipogenic transcription factors C/EBPalpha and peroxisome proliferator-activated receptor-gamma indirectly by permitting hormone-mediated repression of the adipogenic repressor C/EBP homologous protein-10.


Subject(s)
Adipogenesis/genetics , DNA-Binding Proteins/genetics , Fibroblasts/metabolism , Transcription Factors/genetics , 3T3-L1 Cells , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/genetics , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Gene Deletion , Gene Expression Regulation, Developmental , Immunoblotting , Mice , Models, Genetic , Mutation , PPAR gamma/genetics , PPAR gamma/metabolism , Protein Binding , RNA, Small Interfering/genetics , Transcription Factors/metabolism
16.
J Vis Exp ; (147)2019 05 10.
Article in English | MEDLINE | ID: mdl-31132037

ABSTRACT

To express cellular phenotypes in organisms, living cells execute gene expression accordingly, and transcriptional programs play a central role in gene expression. The cellular transcriptional machinery and its chromatin modification proteins coordinate to regulate transcription. To analyze transcriptional regulation at the molecular level, several experimental methods such as electrophoretic mobility shift, transient reporter and chromatin immunoprecipitation (ChIP) assays are available. We describe a modified ChIP assay in detail in this article because of its advantages in directly showing histone modifications and the interactions between proteins and DNA in cells. One of the key steps in a successful ChIP assay is chromatin shearing. Although sonication is commonly used for shearing chromatin, it is difficult to identify reproducible conditions. Instead of shearing chromatin by sonication, we utilized enzymatic digestion with micrococcal nuclease (MNase) to obtain more reproducible results. In this article, we provide a straightforward ChIP assay protocol using MNase.


Subject(s)
Chromatin Immunoprecipitation/methods , Mammals/metabolism , Micrococcal Nuclease/metabolism , Androgens/pharmacology , Animals , Cell Line, Tumor , Chromatin/metabolism , DNA/genetics , Enhancer Elements, Genetic/genetics , Genome, Human , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , Sonication
17.
Medicines (Basel) ; 6(4)2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31618900

ABSTRACT

Background: Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) selectively kills tumor cells in cancer patients. However, patients often develop TRAIL resistance; thus, agents that can sensitize cells to TRAIL therapy would be beneficial clinically. Methods: Immunoblotting, flow cytometry, confocal microscopy, qPCR and caspase 8 activity assays were used to investigate whether glucosamine (GlcN) can sensitize cancer cells to TRAIL thereby enhancing apoptosis and potentially improving clinical response. Results: GlcN sensitized DU145 cells to TRAIL-induced apoptosis but did not increase death receptor 5 (DR5) cell surface expression. Once treated, these cells responded to TRAIL-induced apoptosis through both extrinsic and intrinsic apoptotic pathways as evidenced by the cleavage of both caspases 8 and 9. The combination of GlcN and TRAIL suppressed the expression of key anti-apoptotic factors cFLIP, BCL-XL, MCL-1 and XIAP and translocated BAK to the mitochondrial outer membrane thereby facilitating cytochrome C and SMAC release. In addition to the activation of apoptotic pathways, TRAIL-mediated inflammatory responses were attenuated by GlcN pretreatment reducing nuclear NF-kB levels and the expression of downstream target genes IL-6 and IL-8. Conclusions: GlcN/TRAIL combination could be a promising strategy for treating cancers by overcoming TRAIL resistance and abrogating TRAIL-induced inflammation.

18.
Nat Biotechnol ; 40(9): 1317-1318, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36085502
19.
Biosci Trends ; 9(6): 377-85, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26781795

ABSTRACT

This study demonstrated that ARID5B mRNA is present in mouse cardiomyocyte HL-1 cells, and that ARID5B siRNA constantly knocked down ARID5B gene expression to the 40% level of control. AMPKα2 protein was elevated in such ARID5B knockdown HL-1 cells, and this was accompanied by an increase in the level of phosphorylated AMPKα. Since AMPKα2 mRNA levels did not change in ARID5B knockdown cells, the stability of AMPKα2 protein was investigated using inhibitors for protein synthesis and proteasomal degradation. Treatment of HL-1 cells with either cycloheximide or MG132 caused an appreciable increase in the amount of AMPKα2 protein in ARID5B knockdown cells, which suggests that knockdown of ARID5B mRNA extends the half-life of AMPKα2 protein in HL-1 cells via yet unidentified mechanisms. As for the expected downstream consequences of AMPKα2 activation, we found thus far that glucose uptake, fatty acid uptake, or fatty acid oxidation remained unchanged in HL-1 cells after knockdown of ARID5B. Further studies are required to understand the mechanisms for ARID5B knockdown and resulting AMPKα2 activation, and also to identify which metabolic pathways are affected by AMPKα2 activation in these cells. In summary, this study provided the foundation for an in vitro cell culture system to study possible roles of ARID5B in cardiomyocytes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , DNA-Binding Proteins/physiology , Myocytes, Cardiac/metabolism , Transcription Factors/physiology , Animals , Cell Line , Cycloheximide/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fatty Acids/metabolism , Gene Knockdown Techniques , Glucose/metabolism , Leupeptins/pharmacology , Male , Mice , Myocardium/metabolism , Oxidation-Reduction , Phosphorylation , RNA, Messenger/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL