Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 228
Filter
1.
Mediators Inflamm ; 2021: 2689600, 2021.
Article in English | MEDLINE | ID: mdl-34733115

ABSTRACT

Chronic obstructive pulmonary disease (COPD), a progressive respiratory disease, is characterized by the alveolar epithelium injury and persistent airway inflammation. It is documented that oscillation and dysregulated expression of circadian clock genes, like Bmal1, Per1, and Per2, involved in COPD pathogenies, including chronic inflammation and imbalanced autophagy level, and targeting the associations of circadian rhythm and autophagy is promising strategies in the management and treatment of COPD. Herein, we reviewed the mechanisms of the circadian clock and the unbalance of the autophagic level in COPD, as well as the link between the two, so as to provide further theoretical bases for the study on the pathogenesis of COPD.


Subject(s)
Autophagy/physiology , Circadian Clocks/physiology , Pulmonary Disease, Chronic Obstructive/etiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , CCAAT-Enhancer-Binding Protein-beta/physiology , Circadian Clocks/genetics , Humans , Mechanistic Target of Rapamycin Complex 1/physiology , Melatonin/physiology
2.
Proc Natl Acad Sci U S A ; 117(42): 26328-26339, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33020261

ABSTRACT

Dendritic cell (DC) maturation is a prerequisite for the induction of adaptive immune responses against pathogens and cancer. Transcription factor (TF) networks control differential aspects of early DC progenitor versus late-stage DC cell fate decisions. Here, we identified the TF C/EBPß as a key regulator for DC maturation and immunogenic functionality under homeostatic and lymphoma-transformed conditions. Upon cell-specific deletion of C/EBPß in CD11c+MHCIIhi DCs, gene expression profiles of splenic C/EBPß-/- DCs showed a down-regulation of E2F cell cycle target genes and associated proliferation signaling pathways, whereas maturation signatures were enriched. Total splenic DC cell numbers were modestly increased but differentiation into cDC1 and cDC2 subsets were unaltered. The splenic CD11c+MHCIIhiCD64+ DC compartment was also increased, suggesting that C/EBPß deficiency favors the expansion of monocytic-derived DCs. Expression of C/EBPß could be mimicked in LAP/LAP* isoform knockin DCs, whereas the short isoform LIP supported a differentiation program similar to deletion of the full-length TF. In accordance with E2F1 being a negative regulator of DC maturation, C/EBPß-/- bone marrow-derived DCs matured much faster enabling them to activate and polarize T cells stronger. In contrast to a homeostatic condition, lymphoma-exposed DCs exhibited an up-regulation of the E2F transcriptional pathways and an impaired maturation. Pharmacological blockade of C/EBPß/mTOR signaling in human DCs abrogated their protumorigenic function in primary B cell lymphoma cocultures. Thus, C/EBPß plays a unique role in DC maturation and immunostimulatory functionality and emerges as a key factor of the tumor microenvironment that promotes lymphomagenesis.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Dendritic Cells/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/physiology , Cell Differentiation , Cell Line , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/metabolism , Protein Isoforms/genetics , Signal Transduction , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Tumor Microenvironment/physiology
3.
Genomics ; 112(3): 2400-2409, 2020 05.
Article in English | MEDLINE | ID: mdl-31981700

ABSTRACT

Perilipin 1 (PLIN1) protein, also known as lipid droplet-associated protein, is encoded by the PLIN1 gene and is able to anchor itself to the membranes of lipid droplets. The phosphorylation of PLIN1 is critical for the mobilization of fat in adipose tissue and plays an important role in regulating lipolysis and lipid storage in adipocytes. However, research on the synthesis and lipid metabolism of lipid droplets by PLIN1 in bovine adipocytes is limited. In the present study, we found that bovine PLIN1 was highly expressed in subcutaneous adipose tissue. The highest level of PLIN1 mRNA expression in bovine adipocytes was observed on day 6 of differentiation. Moreover, the cytoplasmic subcellular localization of PLIN1 was observed in bovine preadipocytes. To elucidate the molecular mechanism of bovine PLIN1 transcriptional regulation, we cloned eight fragments containing the 5' regulatory region of the PLIN1 gene. The results showed that the -209/-17 bp region of the bovine PLIN1 gene was the core promoter region. Based on the transcriptional activities of the promoter vector fragments, the luciferase activity of the mutated fragment, the siRNA interference, and the results of the electrophoretic mobility shift assay (EMSA), we identified the binding sites of E2F transcription factor 1 (E2F1), pleiomorphic adenoma gene 1 (PLAG1), CCAAT enhancer binding protein beta (C/EBPß), and SMAD family member 3 (SMAD3) as the transcriptional activators or repressors of the core promoter region. Further experiments confirmed that the knockdown of the PLIN1 gene affected the ability of these transcription factors to regulate the lipid metabolism in bovine adipocytes. In conclusion, our results reveal a potential mechanism for the transcriptional regulation of PLIN1 in bovine adipocytes.


Subject(s)
Adipocytes/metabolism , Cattle/genetics , Perilipin-1/genetics , Promoter Regions, Genetic , Transcription Factors/metabolism , Adipocytes/enzymology , Adipogenesis/genetics , Animals , Binding Sites , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-beta/physiology , Cattle/metabolism , Cells, Cultured , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/physiology , Gene Expression Regulation , Lipid Metabolism/genetics , Perilipin-1/classification , Perilipin-1/metabolism , Phylogeny , Sequence Alignment , Sequence Analysis, DNA , Sequence Analysis, Protein , Smad3 Protein/metabolism , Smad3 Protein/physiology
4.
Exp Gerontol ; 128: 110752, 2019 12.
Article in English | MEDLINE | ID: mdl-31648009

ABSTRACT

Oncogene-induced senescence (OIS) serves as an important barrier to tumor progression in cells that have acquired activating mutations in RAS and other oncogenes. Senescent cells also produce a secretome known as the senescence-associated secretory phenotype (SASP) that includes pro-inflammatory cytokines and chemokines. SASP factors reinforce and propagate the senescence program and identify senescent cells to the immune system for clearance. The OIS program is executed by several transcriptional effectors that include p53, RB, NF-κB and C/EBPß. In this review, we summarize the critical role of C/EBPß in regulating OIS and the SASP. Post-translational modifications induced by oncogenic RAS signaling control C/EBPß activity and dimerization, and these alterations switch C/EBPß to a pro-senescence form during OIS. In addition, C/EBPß is regulated by a unique 3'UTR-mediated mechanism that restrains its activity in tumor cells to facilitate senescence bypass and suppression of the SASP.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/physiology , Cellular Senescence/physiology , Oncogenes/physiology , Animals , CCAAT-Enhancer-Binding Protein-beta/chemistry , Genes, ras/physiology , Humans , MAP Kinase Signaling System/physiology , NF-kappa B/physiology , Phosphorylation , Protein Multimerization , Signal Transduction/physiology
5.
Front Immunol ; 10: 1734, 2019.
Article in English | MEDLINE | ID: mdl-31404149

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous cell population with potent immunosuppressive functions. They play major roles in cancer and many of the pathologic conditions associated with inflammation. Long non-coding RNAs (lncRNAs) are untranslated functional RNA molecules. The lncRNAs are involved in the control of a wide variety of cellular processes and are dysregulated in different diseases. They can participate in the modulation of immune function and activity of inflammatory cells, including MDSCs. This mini review focuses on the emerging role of lncRNAs in MDSC activity. We summarize how lncRNAs modulate the generation, recruitment, and immunosuppressive functions of MDSCs and the underlying mechanisms.


Subject(s)
Inflammation/immunology , Myeloid-Derived Suppressor Cells/immunology , Neoplasms/immunology , RNA, Long Noncoding/physiology , Animals , CCAAT-Enhancer-Binding Protein-beta/physiology , Cell Lineage , Epigenesis, Genetic , Forecasting , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunotherapy , Mice , Myeloid-Derived Suppressor Cells/classification , Neoplasms/genetics , Neoplasms/therapy , Nitric Oxide/metabolism , Pseudogenes , RNA, Long Noncoding/genetics , RNA, Long Noncoding/immunology , RNA, Neoplasm/immunology , RNA, Neoplasm/physiology , Reactive Oxygen Species/metabolism , Tumor Escape , Tumor Microenvironment
6.
Genet Test Mol Biomarkers ; 23(5): 304-309, 2019 May.
Article in English | MEDLINE | ID: mdl-30969151

ABSTRACT

Aims: Metastasis is a significant obstacle to curing esophageal squamous cell carcinoma (ESCC). The CCAAT/enhancer binding protein ß (C/EBPß) and matrix metalloproteinase 3 (MMP3) are thought to play key roles in cancer invasion and metastasis. In this study, we aimed to detect whether C/EBPß-mediated tumor invasion was dependent on MMP3. In addition, we determined whether C/EBPß upregulation was associated with MMP3 levels and metastatic status in patients with ESCC. Materials and Methods: A total of 126 patients with ESCC were recruited for this study. The mRNA and protein levels of C/EBPß and MMP3 in ESCC cell lines and specimens from ESCC patient were determined by reverse transcription-polymerase chain reaction and western blot, respectively. Tumor cell invasion was analyzed using an in vitro Matrigel Invasion Assay. The correlation between C/EBPß and MMP3 expression was determined by Pearson's correlation analysis. Results: Both mRNA and protein levels of MMP3 were upregulated by C/EBPß overexpression and downregulated by C/EBPß siRNA in KYSE150 cell cultures. The promotion of ESCC cell invasion through C/EBPß was inhibited by MMP3 siRNA. The level of C/EBPß was correlated with MMP3 and metastatic status in patients with ESCC. Conclusions: C/EBPß upregulation promoted tumor cell invasion in an MMP3-dependent manner in vitro and was associated with metastatic status in ESCC.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/physiology , Esophageal Squamous Cell Carcinoma/genetics , Matrix Metalloproteinase 3/physiology , Aged , CCAAT-Enhancer-Binding Protein-beta/genetics , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , China , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/physiopathology , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Up-Regulation
7.
Sci Rep ; 8(1): 16613, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30413755

ABSTRACT

Myogenesis is regulated by the coordinated expression of muscle regulatory factors, a family of transcription factors that includes MYOD, MYF5, myogenin and MRF4. Muscle regulatory factors are basic helix-loop-helix transcription factors that heterodimerize with E proteins to bind the regulatory regions of target genes. Their activity can be inhibited by members of the Inhibitor of DNA binding and differentiation (ID) family, which bind E-proteins with high affinity, thereby preventing muscle regulatory factor-dependent transcriptional responses. CCAAT/Enhancer Binding protein beta (C/EBPß) is a transcription factor expressed in myogenic precursor cells that acts to inhibit myogenic differentiation, though the mechanism remains poorly understood. We identify Id3 as a novel C/EBPß target gene that inhibits myogenic differentiation. Overexpression of C/EBPß stimulates Id3 mRNA and protein expression, and is required for C/EBPß-mediated inhibition of myogenic differentiation. Misexpression of C/EBPß in myogenic precursors, such as in models of cancer cachexia, prevents the differentiation of myogenic precursors and we show that loss of Id3 rescues differentiation under these conditions, suggesting that the stimulation of Id3 expression by C/EBPß is an important mechanism by which C/EBPß inhibits myogenic differentiation.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/physiology , Cachexia/pathology , Cell Differentiation , Inhibitor of Differentiation Proteins/metabolism , Muscle Development , Muscle, Skeletal/cytology , Myoblasts/cytology , Animals , Cachexia/genetics , Cachexia/metabolism , Culture Media, Conditioned/pharmacology , Gene Expression Regulation, Developmental , Humans , Inhibitor of Differentiation Proteins/genetics , Male , Mice, Knockout , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Prostatic Neoplasms/physiopathology , Tumor Cells, Cultured
8.
Rinsho Ketsueki ; 59(6): 798-804, 2018.
Article in Japanese | MEDLINE | ID: mdl-29973462

ABSTRACT

Under stress conditions such as infection, inflammation, and hematopoietic recovery following chemotherapy or transplantation, the hematopoietic system is required to meet the increasing demands, especially from myeloid cells. Therefore, an understanding of the molecular mechanism underlying stress hematopoiesis is clinically imperative. We previously showed that C/EBPß, which is a transcription factor required for emergency granulopoiesis, plays a pivotal role at the level of hematopoietic stem/progenitor cells under stress conditions. Upon exposure to stress, the C/EBPß protein is upregulated in the hematopoietic stem cells. A close examination of C/EBPß knockout mice revealed that C/EBPß regulates the proliferation and differentiation of hematopoietic stem cells at the cost of the self-renewing activity. Further elucidation of the functions and regulation of C/EBPß in hematopoietic stem cells will facilitate an understanding of stress hematopoiesis.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/physiology , Cell Differentiation , Cell Proliferation , Granulocytes , Hematopoietic Stem Cells/cytology , Animals , Hematopoiesis , Mice , Mice, Knockout
9.
Rinsho Ketsueki ; 59(6): 805-811, 2018.
Article in Japanese | MEDLINE | ID: mdl-29973463

ABSTRACT

Recent research has revealed that macrophages and monocytes comprise various subtypes. Previously, we demonstrated that JMJD3 is vital for macrophage differentiation in response to allergic stimuli. Moreover, we substantiated that Trib1 controls the differentiation of tissue-resident macrophages in peripheral organs, such as adipose tissue. This study aims to elucidate that Ceacam1+Msr1+Ly6C-F4/80-Mac1+ monocytes are essential for the development of fibrosis. Remarkably, these cell types harbor bilobed-like nucleus and some granules in the cytoplasm. Thus, we named these as cell segregated-nucleus-containing atypical monocytes (SatM). The results revealed that NFIL6 is critical for the differentiation of SatM, and the lack of this protein causes a complete deficiency of SatM. Furthermore, the development of fibrosis was prevented in NFIL6-/- chimeric mice and the adoptive transfer of SatM into NFIL6-/- chimeric mice resulted in fibrosis. Thus, macrophage and monocytes comprised multiple subtypes with functional diversity.


Subject(s)
Cell Differentiation , Macrophages/cytology , Monocytes/cytology , Animals , CCAAT-Enhancer-Binding Protein-beta/physiology , Fibrosis , Macrophages/classification , Mice , Mice, Knockout , Monocytes/classification
10.
Blood ; 130(16): 1809-1818, 2017 10 19.
Article in English | MEDLINE | ID: mdl-28807982

ABSTRACT

The transcription factor CCAAT/enhancer-binding protein ß (C/EBPß) is highly expressed in monocytes/macrophages. However, its roles in monopoiesis are largely unknown. Here, we investigated the roles of C/EBPß in monopoiesis. Further subdivision of monocytes revealed that Cebpb messenger RNA was highly upregulated in Ly6C- monocytes in bone marrow. Accordingly, the number of Ly6C- monocytes was significantly reduced in Cebpb-/- mice. Bone marrow chimera experiments and Mx1-Cre-mediated deletion of Cebpb revealed a cell-intrinsic and monocyte-specific requirement for C/EBPß in monopoiesis. In Cebpb-/- mice, turnover of Ly6C- monocytes was highly accelerated and apoptosis of Ly6C- monocytes was increased. Expression of Csf1r, which encodes a receptor for macrophage colony-stimulating factor, was significantly reduced in Ly6C- monocytes of Cebpb-/- mice. C/EBPß bound to positive regulatory elements of Csf1r and promoted its transcription. Collectively, these results indicate that C/EBPß is a critical factor for Ly6C- monocyte survival, at least in part through upregulation of Csf1r.


Subject(s)
Apoptosis/genetics , CCAAT-Enhancer-Binding Protein-beta/physiology , Monocytes/physiology , Animals , Antigens, Ly/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , COS Cells , Cell Differentiation/genetics , Cell Survival/genetics , Cells, Cultured , Chlorocebus aethiops , Gene Expression Regulation , Mice , Mice, Knockout , Monocytes/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology
11.
Anim Sci J ; 88(9): 1291-1297, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28139071

ABSTRACT

Endometrial remodeling is important for successful embryo development and implantation in pigs. Therefore, this study investigated change of proteins regulating endometrial remodeling on follicular and luteal phase in porcine endometrial tissues. The endometrial tissue samples were collected from porcine uterus during follicular and luteal phase, vascular endothelial growth factor (VEGF), myoglobin and cysteine-rich protein 2 (CRP2) proteins were expressed by immnofluorescence, immunoblotting, and determined by 2-DE and MALDI-TOF/MS. We found that VEGF, myoglobin and CRP2 were strongly localized in endometrial tissues during luteal phase, but not follicular phase. The protein levels of VEGF, myoglobin and CRP2 in endometrial tissues were higher than luteal phase (P < 0.05). These results may provide understanding of intrauterine environment during estrous cycle in pigs, and will be used in animal reproduction for developing specific biomarkers in the future.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Endometrium/metabolism , Follicular Phase/genetics , Follicular Phase/metabolism , Luteal Phase/genetics , Luteal Phase/metabolism , Myoglobin/metabolism , Swine/metabolism , Swine/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/physiology , Embryo Implantation/genetics , Embryo Implantation/physiology , Embryonic Development/genetics , Embryonic Development/physiology , Estrous Cycle/genetics , Estrous Cycle/physiology , Female , Myoglobin/physiology , Vascular Endothelial Growth Factor A/physiology
12.
Nat Rev Cancer ; 17(2): 116-130, 2017 02.
Article in English | MEDLINE | ID: mdl-27977008

ABSTRACT

Recent studies across multiple tumour types are starting to reveal a recurrent regulatory architecture in which genomic alterations cluster upstream of functional master regulator (MR) proteins, the aberrant activity of which is both necessary and sufficient to maintain tumour cell state. These proteins form small, hyperconnected and autoregulated modules (termed tumour checkpoints) that are increasingly emerging as optimal biomarkers and therapeutic targets. Crucially, as their activity is mostly dysregulated in a post-translational manner, rather than by mutations in their corresponding genes or by differential expression, the identification of MR proteins by conventional methods is challenging. In this Opinion article, we discuss novel methods for the systematic analysis of MR proteins and of the modular regulatory architecture they implement, including their use as a valuable reductionist framework to study the genetic heterogeneity of human disease and to drive key translational applications.


Subject(s)
Neoplasms/etiology , Animals , CCAAT-Enhancer-Binding Protein-beta/physiology , Cell Transformation, Neoplastic , Disease Progression , Drug Resistance, Neoplasm , Glucocorticoids/pharmacology , Homeostasis , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Oncogenes , Systems Biology
13.
J Immunol ; 198(3): 1297-1307, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28011933

ABSTRACT

During macrophage activation, expression of IL-1R-associated kinase (IRAK)-M is induced to suppress TLR-mediated responses and is a hallmark of endotoxin tolerance. Endotoxin tolerance requires tight regulation of genes occurring at the transcriptional and epigenetic levels. To identify novel regulators of IRAK-M, we used RAW 264.7 macrophages and performed a targeted RNA interference screen of genes encoding chromatin-modifying enzymes, signaling molecules, and transcription factors involved in macrophage activation. Among these, the transcription factor CCAAT/enhancer binding protein (C/EBP)ß, known to be involved in macrophage inactivation, was necessary for the induction of IRAK-M expression. Chromatin immunoprecipitation showed that C/EBPß was recruited to the IRAK-M promoter following LPS stimulation and was indispensable for IRAK-M transcriptional activation. Among histone 3-modifying enzymes, our screen showed that knockdown of the histone 3 lysine 27 (H3K27) methyltransferase and part of the polycomb recessive complex 2, enhancer of Zeste 2, resulted in IRAK-M overexpression. In contrast, knockdown of the H3K27 demethylase ubiquitously transcribed tetratricopeptide repeat X chromosome suppressed the induction of IRAK-M in response to LPS stimulation. Accordingly, we demonstrated that H3K27 on the IRAK-M promoter is trimethylated in unstimulated cells and that this silencing epigenetic mark is removed upon LPS stimulation. Our data propose a mechanism for IRAK-M transcriptional regulation according to which, in the naive state, polycomb recessive complex 2 repressed the IRAK-M promoter, allowing low levels of expression; following LPS stimulation, the IRAK-M promoter is derepressed, and transcription is induced to allow its expression.


Subject(s)
Epigenesis, Genetic , Interleukin-1 Receptor-Associated Kinases/genetics , Macrophages/metabolism , Transcription, Genetic , Animals , CCAAT-Enhancer-Binding Protein-beta/physiology , Cells, Cultured , Dealkylation , Lipopolysaccharides/pharmacology , Mice , NF-kappa B/physiology , Promoter Regions, Genetic
14.
Cancer Res ; 76(20): 6030-6042, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27550451

ABSTRACT

The ability of tumor cells to escape immune destruction and their acquired resistance to chemotherapy are major obstacles to effective cancer therapy. Although immune checkpoint therapies such as anti-PD-1 address these issues in part, clinical responses remain limited to a subpopulation of patients. In this report, we identified IL34 produced by cancer cells as a driver of chemoresistance. In particular, we found that IL34 modulated the functions of tumor-associated macrophages to enhance local immunosuppression and to promote the survival of chemoresistant cancer cells by activating AKT signaling. Targeting IL34 in chemoresistant tumors resulted in a remarkable inhibition of tumor growth when accompanied with chemotherapy. Our results define a pathogenic role for IL34 in mediating immunosuppression and chemoresistance and identify it as a tractable target for anticancer therapy. Cancer Res; 76(20); 6030-42. ©2016 AACR.


Subject(s)
Immune Tolerance , Interleukins/physiology , Lung Neoplasms/drug therapy , Macrophages/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/physiology , Cell Differentiation , Cell Line, Tumor , Cell Polarity , Drug Resistance, Neoplasm , Humans , Interleukins/analysis , Interleukins/antagonists & inhibitors , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice , Monocytes/cytology , NF-kappa B/physiology , Proto-Oncogene Proteins c-akt/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology
15.
Chin Med J (Engl) ; 129(9): 1108-12, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27098798

ABSTRACT

BACKGROUND: Adipocytes behave like a rich source of pro-inflammatory cytokines including monocyte chemoattractant protein-1 (MCP-1). Oxidized low-density lipoprotein (oxLDL) participates in the local chronic inflammatory response, and high-density lipoprotein could counterbalance the proinflammatory function of oxLDL, but the underlying mechanism is not completely understood. This study aimed to evaluate the effect of apolipoprotein A-I mimetic peptide L-4F on the secretion and expression of MCP-1 in fully differentiated 3T3-L1 adipocytes induced by oxLDL and to elucidate the possible mechanisms. METHODS: Fully differentiated 3T3-L1 adipocytes were incubated in the medium containing various concentration of L-4F (0-50 µg/ml) with oxLDL (50 µg/ml) stimulated, with/without protein kinase A (PKA) inhibitor H-89 (10 µmol/L) preincubated. The concentrations of MCP-1 in the supernatant, the mRNA expression of MCP-1, the levels of CCAAT/enhancer binding protein α (C/EBPα), and CCAAT/enhancer binding protein ß (C/EBPß) were evaluated. The monocyte chemotaxis assay was performed by micropore filter method using a modified Boyden chamber. RESULTS: OxLDL stimulation induced a significant increase of MCP-1 expression and secretion in 3T3-L1 adipocytes, which were inhibited by L-4F preincubation in a dose-dependent manner. PKA inhibitor H-89 markedly reduced the oxLDL-induced MCP-1 expression, but no further decrease was observed when H-89 was used in combination with L-4F (50 µg/ml) (P > 0.05). OxLDL stimulation showed no significant effect on C/EBPα protein level but increased C/EBPß protein level in a time-dependent manner. H-89 and L-4F both attenuated C/EBPß protein level in oxLDL-induced 3T3-L1 adipocytes. CONCLUSIONS: OxLDL induces C/EBPß protein synthesis in a time-dependent manner and enhances MCP-1 secretion and expression in 3T3-L1 adipocytes. L-4F dose-dependently counterbalances the pro-inflammatory effect of oxLDL, and cyclic AMP/PKA-C/EBPß signaling pathway may participate in it.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/physiology , Chemokine CCL2/metabolism , Cyclic AMP-Dependent Protein Kinases/physiology , Cyclic AMP/physiology , Lipoproteins, LDL/antagonists & inhibitors , Peptides/pharmacology , Signal Transduction/physiology , 3T3-L1 Cells , Animals , CCAAT-Enhancer-Binding Protein-beta/analysis , Chemokine CCL2/genetics , Humans , Lipoproteins, LDL/pharmacology , Mice
16.
Cancer Lett ; 375(1): 152-161, 2016 May 28.
Article in English | MEDLINE | ID: mdl-26968249

ABSTRACT

The purpose of this study was to ascertain the mechanisms by which advanced prostate cancer cells resist bortezomib therapy. Several independent studies have shown that cells are protected from proteasome inhibition by increased autophagic activity. We investigated whether C/EBPß, a transcription factor involved in the control of autophagic gene expression, regulates resistance to proteasome inhibition. In PC3 cells over-expressing C/EBPß, turnover of autophagic substrates and expression of core autophagy genes were increased. Conversely, C/EBPß knockdown suppressed autophagosome-lysosome fusion. We also found that C/EBPß knockdown suppressed REDD1 expression to delay early autophagy, an effect rescued by exogenous REDD1. Cells with suppressed C/EBPß levels showed delayed autophagy activation upon bortezomib treatment. Knockdown of C/EBPß sensitized PC3 cells to bortezomib, and blockade of autophagy by chloroquine did not further increase cell death in cells expressing shRNA targeting C/EBPß. Lastly, we observed a decreased growth of PC3 cells and xenografts with C/EBPß knockdown and such xenografts were sensitized to bortezomib treatment. Our results demonstrate that C/EBPß is a critical effector of autophagy via regulation of autolysosome formation and promotes resistance to proteasome inhibitor treatment by increasing autophagy.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , CCAAT-Enhancer-Binding Protein-beta/physiology , Prostatic Neoplasms/metabolism , Transcription Factors/genetics , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cell Survival , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Lysosomes/metabolism , Male , Membrane Fusion , Mice, Inbred NOD , Mice, SCID , Phagosomes/metabolism , Prostatic Neoplasms/drug therapy , Protein Binding , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
17.
J Mol Endocrinol ; 56(2): 91-100, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26576644

ABSTRACT

G protein-coupled receptor 120 (GPR120), an adipogenic receptor critical for the differentiation and maturation of adipocytes, plays an important role in controlling obesity in both humans and rodents and, thus, is an attractive target of obesity treatment studies. However, the mechanisms that regulate the expression of porcine GPR120 remain unclear. In this study, electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) techniques were used to analyze and identify the binding of C/EBPß (transcription factor CCAAT/enhancer binding protein beta) to the GPR120 promoter. C/EBPß overexpression and RNA interference studies showed that C/EBPß regulated GPR120 promoter activity and endogenous GPR120 expression. The binding site of C/EBPß in the GPR120 promoter region from -101 to -87 was identified by promoter deletion analysis and site-directed mutagenesis. Overexpression of C/EBPß increased endogenous GPR120 expression in pig kidney cells (PK). Furthermore, when endogenous C/EBPß was knocked down, GPR120 mRNA and protein levels were decreased. The stimulatory effect of C/EBPß on GPR120 transcription and its ability to bind the transcription factor-binding site were confirmed by luciferase, ChIP, and EMSA. Moreover, the mRNA and protein expression levels of C/EBPß were induced by high fat diet feeding. Taken together, it can be concluded that C/EBPß plays a vital role in regulating GPR120 transcription and suggests HFD-feeding induces GPR120 transcription by influencing C/EBPß expression.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/physiology , Receptors, G-Protein-Coupled/genetics , Transcription, Genetic , Transcriptional Activation , 3T3-L1 Cells , Animals , Base Sequence , Binding Sites , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Sequence Analysis, DNA , Sus scrofa , Up-Regulation
18.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 31(11): 1483-7, 2015 Nov.
Article in Chinese | MEDLINE | ID: mdl-26522356

ABSTRACT

OBJECTIVE: To explore the regulatory role of CCAAT/enhancer-binding protein beta (C/EBPß) in invasion and migration of 786-O human renal cancer cells. METHODS: C/EBPß adenoviral overexpression vector (pAd-C/EBPß) and C/EBPß siRNA were constructed and respectively transfered into 786-O cells. Wound healing and Transwell(TM) invasion methods were used to detect the invasion and migration of the cells. Laser scanning confocal microscopy was applied to determine the translocation of nuclear factor κB (NF-κB), the central regulator of epithelial-mesenchymal transition (EMT); Western blotting was performed to examine the levels of EMT key markers E-cadherin, vimentin and tenascin. RESULTS: When C/EBPß was overexpressed, invasion and migration of 786-O cells were enhanced, translocation of NF-κB increased, E-cadherin level was reduced, and vimentin and tenascin levels were elevated. When C/EBPß was knocked down, the results were completely opposite to those of C/EBPß overexpression: invasion and migration of 786-O cells were inhibited, translocation of NF-κB decreased, E-cadherin level was elevated, and vimentin and tenascin expressions were suppressed. Moreover, the above changes were not found in the cells that co-treated with pAd-C/EBPß and C/EBPß siRNA. CONCLUSION: C/EBPß could promote the invasion and migration of human renal carcinoma 786-O cells, and had an inducing effect on nuclear translocation of NF-κB in 786-O cells.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/physiology , Cell Movement , Kidney Neoplasms/pathology , NF-kappa B/physiology , Active Transport, Cell Nucleus , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Humans , Neoplasm Invasiveness
19.
PLoS Genet ; 11(9): e1005505, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26372225

ABSTRACT

Schmid metaphyseal chondrodysplasia (MCDS) involves dwarfism and growth plate cartilage hypertrophic zone expansion resulting from dominant mutations in the hypertrophic zone collagen, Col10a1. Mouse models phenocopying MCDS through the expression of an exogenous misfolding protein in the endoplasmic reticulum (ER) in hypertrophic chondrocytes have demonstrated the central importance of ER stress in the pathology of MCDS. The resultant unfolded protein response (UPR) in affected chondrocytes involved activation of canonical ER stress sensors, IRE1, ATF6, and PERK with the downstream effect of disrupted chondrocyte differentiation. Here, we investigated the role of the highly conserved IRE1/XBP1 pathway in the pathology of MCDS. Mice with a MCDS collagen X p.N617K knock-in mutation (ColXN617K) were crossed with mice in which Xbp1 was inactivated specifically in cartilage (Xbp1CartΔEx2), generating the compound mutant, C/X. The severity of dwarfism and hypertrophic zone expansion in C/X did not differ significantly from ColXN617K, revealing surprising redundancy for the IRE1/XBP1 UPR pathway in the pathology of MCDS. Transcriptomic analyses of hypertrophic zone cartilage identified differentially expressed gene cohorts in MCDS that are pathologically relevant (XBP1-independent) or pathologically redundant (XBP1-dependent). XBP1-independent gene expression changes included large-scale transcriptional attenuation of genes encoding secreted proteins and disrupted differentiation from proliferative to hypertrophic chondrocytes. Moreover, these changes were consistent with disruption of C/EBP-ß, a master regulator of chondrocyte differentiation, by CHOP, a transcription factor downstream of PERK that inhibits C/EBP proteins, and down-regulation of C/EBP-ß transcriptional co-factors, GADD45-ß and RUNX2. Thus we propose that the pathology of MCDS is underpinned by XBP1 independent UPR-induced dysregulation of C/EBP-ß-mediated chondrocyte differentiation. Our data suggest that modulation of C/EBP-ß activity in MCDS chondrocytes may offer therapeutic opportunities.


Subject(s)
Bone Diseases/pathology , CCAAT-Enhancer-Binding Protein-beta/antagonists & inhibitors , Cell Differentiation/physiology , Chondrocytes/pathology , DNA-Binding Proteins/physiology , Endoplasmic Reticulum Stress/physiology , Transcription Factors/physiology , Unfolded Protein Response/physiology , Animals , CCAAT-Enhancer-Binding Protein-beta/physiology , DNA-Binding Proteins/genetics , Gene Expression Profiling , Mice , Mice, Transgenic , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , X-Box Binding Protein 1
20.
Biochem Biophys Res Commun ; 464(2): 654-8, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26168729

ABSTRACT

The CCAAT/enhancer-binding protein ß (C/EBPß) transcription factor is required for granulopoiesis under stress conditions. However, little is known about its roles in steady state hematopoiesis. Here, we analyzed the peripheral blood and bone marrow of Cebpb(-/-) mice at steady state by flow cytometry and unexpectedly found that the number of peripheral blood monocytes was severely reduced, while the number of bone marrow monocytes was maintained. The ability of Cebpb(-/-) bone marrow cells to give rise to macrophages/monocytes in vitro was comparable to that of wild-type bone marrow cells. Apoptosis of monocytes was enhanced in the peripheral blood, but not in the bone marrow of Cebpb(-/-) mice. These results indicate that C/EBPß is required for the survival of monocytes in peripheral blood.


Subject(s)
Apoptosis/physiology , CCAAT-Enhancer-Binding Protein-beta/physiology , Monocytes/cytology , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Lymphocyte Count , Macrophages/cytology , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL