Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Cell Biochem ; 123(3): 644-656, 2022 03.
Article in English | MEDLINE | ID: mdl-34989006

ABSTRACT

The factor binding inducer of short transcripts-1 (FBI-1) is a POZ-domain Kruppel-like (POK) family of transcription factors and is known as a proto-oncogene or tumor suppressor in various carcinomas. However, the role of FBI-1 on epithelial-to-mesenchymal transition (EMT) and invasiveness in lung cancer remains unknown. Preliminarily, clinical data such as tissue microarray, Kaplan-Meier, and Oncomine were analyzed to confirm the correlation between lung cancer metastasis and FBI-1. To investigate the function of FBI-1 in EMT in lung cancer, EMT was measured in FBI-1-deficient or FBI-1-overexpressing cells. FBI-1 showed decreased expression in tumors metastasized to lymph nodes than in the primary tumor. In addition, it was also associated with improved survival rates of lung cancer patients. FBI-1 knockdown improved E-to-N-cadherin switching, migration, and invasion in A549 cells, similar to the initiation of EMT stimulated by transforming growth factor- ß1 (TGF-ß1). In contrast, overexpression of FBI-1 inhibited the transcription and activation of Smad2, thereby interfering with EMT, despite stimulation by TGF-ß1. These results suggest that FBI-1 plays a negative role in EMT in lung cancer via the TGF-ß1 signaling pathway, implying its use as a new potential therapeutic target and diagnostic indicator for early stage of lung cancer metastasis.


Subject(s)
Adenocarcinoma of Lung , DNA-Binding Proteins , Epithelial-Mesenchymal Transition , Lung Neoplasms , Transcription Factors , A549 Cells , Adenocarcinoma of Lung/pathology , Cell Line, Tumor , Cell Movement , DNA-Binding Proteins/metabolism , Humans , Lung Neoplasms/metabolism , Neoplasm Invasiveness , Signal Transduction , Transcription Factors/metabolism , Transforming Growth Factor beta1/metabolism
2.
Int J Mol Sci ; 23(17)2022 Aug 23.
Article in English | MEDLINE | ID: mdl-36076953

ABSTRACT

Disruption of the skin microbial balance can exacerbate certain skin diseases and affect prognosis and treatment. Changes in the distribution and prevalence of certain microbial species on the skin, such as Staphylococcus aureus (SA), can impact the development of severe atopic dermatitis (AD) or psoriasis (Pso). A dysfunctional skin barrier develops in AD and Pso due to SA colonization, resulting in keratinization and chronic or progressive chronic inflammation. Disruption of the skin barrier following SA colonization can elevate the production of T helper 2 (Th2)-derived cytokines, which can cause an imbalance in Th1, Th2, and Th17 cells. This study examined the ability of potential therapeutic skin microbiomes, such as Cutibacterium avidum R-CH3 and Staphylococcus hominis R9, to inhibit SA biofilm formation and restore skin barrier function-related genes through the activation of the aryl hydrocarbon receptor (AhR) and the nuclear factor erythroid-2-related factor 2 (Nrf2) downstream target. We observed that IL-4/IL-13-induced downregulation of FLG, LOR, and IVL induced by SA colonization could be reversed by dual AhR/Nrf2 activation. Further, OVOL1 expression may be modulated by functional microbiomes via dual AhR/Nrf2 activation. Our results suggest that our potential therapeutic skin microbiomes can prevent SA-derived Th2-biased skin barrier disruption via IL-13 and IL-4-dependent FLG deregulation, STAT3 activation, and AhR-mediated STAT6 expression.


Subject(s)
Microbiota , Psoriasis , Receptors, Aryl Hydrocarbon , Staphylococcus aureus , Humans , Immunity , Interleukin-13/metabolism , Interleukin-4/metabolism , Intermediate Filament Proteins/genetics , Keratinocytes/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Psoriasis/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Skin/metabolism , Skin/microbiology , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism
3.
J Biol Chem ; 294(1): 299-313, 2019 01 04.
Article in English | MEDLINE | ID: mdl-30409904

ABSTRACT

Even in the face of physiological DNA damage or expression of the tumor suppressor protein p53, B cell CLL/lymphoma 6 (BCL6) increases proliferation and antagonizes apoptotic responses in B cells. BCL6 represses TP53 transcription and also appears to inactivate p53 at the protein level, and additional findings have suggested negative mutual regulation between BCL6 and p53. Here, using Bcl6-/- knockout mice, HEK293A and HCT116 p53-/- cells, and site-directed mutagenesis, we found that BCL6 interacts with p53 and thereby inhibits acetylation of Lys-132 in p53 by E1A-binding protein p300 (p300), a modification that normally occurs upon DNA damage-induced cellular stress and whose abrogation by BCL6 diminished transcriptional activation of p53 target genes, including that encoding caspase-1. Conversely, we also found that BCL6 protein is degraded via p53-induced, caspase-mediated proteolytic cleavage, and the formation of a BCL6-p53-caspase-1 complex. Our results suggest that p53 may block oncogenic transformation by decreasing BCL6 stability via caspase-1 up-regulation, whereas aberrant BCL6 expression inactivates transactivation of p53 target genes, either by inhibiting p53 acetylation by p300 or repressing TP53 gene transcription. These findings have implications for B cell development and lymphomagenesis.


Subject(s)
B-Lymphocytes/metabolism , Caspase 1/blood , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Enzymologic , Proto-Oncogene Proteins c-bcl-6/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , B-Lymphocytes/pathology , Caspase 1/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , HCT116 Cells , HEK293 Cells , Humans , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6/genetics , Tumor Suppressor Protein p53/genetics
4.
Biochim Biophys Acta ; 1859(11): 1429-1439, 2016 11.
Article in English | MEDLINE | ID: mdl-27646874

ABSTRACT

Matrix metalloproteinases (MMPs) are zinc-containing endopeptidases that play roles in cell proliferation, migration, differentiation, angiogenesis, and apoptosis. The expression of MMP gene is tightly regulated and shows cell- and tissue-specific expression patterns. Despite their differential expression, MMP genes have AP-1 (activator protein-1) binding elements within their promoters. Interestingly, c-JUN phosphorylation by cytokine signaling decreased its interaction with NCoR, but increased its interaction with p300, resulting in activation of MMP gene transcription. Here, we found that Zbtb7c (Kr-pok) is a critical component of a transcriptional repressor complex containing c-Jun and NCoR. c-Jun, bound at AP-1, interacts with Zbtb7c, which in turn recruits an NCoR/Hdac3 complex to repress several Mmp (-8, -10, -13, and -16) genes. The molecular interaction between c-Jun and Zbtb7c also prevents phosphorylation of c-Jun by p-Jnk, However, Zbtb7c phosphorylation by p-Jnk (induced by TNFα), and its (Zbtb7c) subsequent degradation by the ubiquitin-mediated proteasomal pathway, leads to c-Jun phosphorylation by p-Jnk. Promoter-bound p-c-Jun then recruits the coactivator p300 to upregulate Mmp gene. Overall, these findings show that Zbtb7c is a key molecule that recruits an NCoR/Hdac3 complex to inhibit phosphorylation of c-Jun, and thereby repress Mmp gene expression.


Subject(s)
Matrix Metalloproteinases/genetics , Proteins/genetics , Transcription, Genetic , Amino Acid Sequence , Animals , Humans , Intracellular Signaling Peptides and Proteins , Mice , NIH 3T3 Cells , Promoter Regions, Genetic , Proteins/chemistry , Proteolysis , Sequence Homology, Amino Acid , Tumor Necrosis Factor-alpha/administration & dosage , Ubiquitination
5.
Nucleic Acids Res ; 43(3): 1609-25, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25609694

ABSTRACT

The NF-κB is found in almost all animal cell types and is involved in a myriad of cellular responses. Aberrant expression of NF-κB has been linked to cancer, inflammatory diseases and improper development. Little is known about transcriptional regulation of the NF-κB family member gene RelA/p65. Sp1 plays a key role in the expression of the RelA/p65 gene. ZBTB2 represses transcription of the gene by inhibiting Sp1 binding to a Sp1-binding GC-box in the RelA/p65 proximal promoter (bp, -31 to -21). Moreover, recent studies revealed that RelA/p65 directly binds to the peroxisome proliferator-activated receptor-γ coactivator1α (PGC1α) to decrease transcriptional activation of the PGC1α target gene PDK4, whose gene product inhibits pyruvate dehydrogenase (PDH), a key regulator of TCA cycle flux. Accordingly, we observed that RelA/p65 repression by ZBTB2 indirectly results in increased PDK4 expression, which inhibits PDH. Consequently, in cells with ectopic ZBTB2, the concentrations of pyruvate and lactate were higher than those in normal cells, indicating changes in glucose metabolism flux favoring glycolysis over the TCA cycle. Knockdown of ZBTB2 in mouse xenografts decreased tumor growth. ZBTB2 may increase cell proliferation by reprogramming glucose metabolic pathways to favor glycolysis by upregulating PDK4 expression via repression of RelA/p65 expression.


Subject(s)
Protein Serine-Threonine Kinases/genetics , Repressor Proteins/physiology , Transcription Factor RelA/genetics , Transcription, Genetic , Base Sequence , Cell Line , DNA Primers , Humans , Promoter Regions, Genetic , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Real-Time Polymerase Chain Reaction , Sp1 Transcription Factor/metabolism
6.
Proc Natl Acad Sci U S A ; 111(42): 15078-83, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25288747

ABSTRACT

An unresolved issue in genotoxic stress response is identification of induced regulatory proteins and how these activate tumor suppressor p53 to determine appropriate cell responses. Transcription factor KAISO was previously described to repress transcription following binding to methylated DNA. In this study, we show that KAISO is induced by DNA damage in p53-expressing cells and then interacts with the p53-p300 complex to increase acetylation of p53 K320 and K382 residues, although decreasing K381 acetylation. Moreover, the p53 with this particular acetylation pattern shows increased DNA binding and potently induces cell cycle arrest and apoptosis by activating transcription of CDKN1A (cyclin-dependent kinase inhibitor 1) and various apoptotic genes. Analogously, in Kaiso KO mouse embryonic fibroblast cells, p53-to-promoter binding and up-regulation of p21 and apoptosis gene expression is significantly compromised. KAISO may therefore be a critical regulator of p53-mediated cell cycle arrest and apoptosis in response to various genotoxic stresses in mammalian cells.


Subject(s)
Apoptosis , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Animals , Cell Cycle , Cell Line , Cell Proliferation , DNA/chemistry , DNA Damage , DNA Methylation , E1A-Associated p300 Protein/metabolism , Female , Fibroblasts/cytology , HCT116 Cells , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Protein Binding , Response Elements
7.
Biochim Biophys Acta ; 1849(9): 1170-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26183023

ABSTRACT

KAISO, a member of the POK protein family, is induced by DNA-damaging agents to enhance apoptosis in a p53-dependent manner. Previously, we found that p53 interacts with KAISO, and acetylation of p53 lysine residues by p300 is modulated by KAISO. APAF1, the core molecule of the apoptosome, is transcriptionally activated by KAISO only in cells expressing p53, which binds to APAF1 promoter p53-response elements (p53REs). APAF1 transcriptional upregulation is further enhanced by KAISO augmentation of p53 binding to the APAF1 promoter distal p53RE#1 (bp, -765 to -739). Interestingly, a NF-κB response element, located close to the p53RE#1, mediates APAF1 transcriptional repression by affecting interaction between KAISO and p53. Ectopic RelA/p65 expression led to depletion of nuclear KAISO, with KAISO being mainly detected in the cytoplasm. RelA/p65 cytoplasmic sequestration of KAISO prevents its nuclear interaction with p53, decreasing APAF1 transcriptional activation by a p53-KAISO-p300 complex in cells exposed to genotoxic stresses. While KAISO enhances p53-dependent apoptosis by increasing APAF1 gene expression, RelA/p65 decreases apoptosis by blocking interaction between KAISO and p53. These findings have relevance to the phenomenon of cancer cells' diminished apoptotic capacity and the onset of chemotherapy resistance.


Subject(s)
Apoptotic Protease-Activating Factor 1/genetics , Transcription Factor RelA/physiology , Transcription Factors/physiology , Transcriptional Activation/physiology , Tumor Suppressor Protein p53/physiology , Cell Line , Cell Proliferation/physiology , Cytoplasm/metabolism , Humans , Promoter Regions, Genetic , Protein Binding , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
8.
Nucleic Acids Res ; 42(18): 11447-61, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25245946

ABSTRACT

ZNF509 is unique among POK family proteins in that four isoforms are generated by alternative splicing. Short ZNF509 (ZNF509S1, -S2 and -S3) isoforms contain one or two out of the seven zinc-fingers contained in long ZNF509 (ZNF509L). Here, we investigated the functions of ZNF509 isoforms in response to DNA damage, showing isoforms to be induced by p53. Intriguingly, to inhibit proliferation of HCT116 and HEK293 cells, we found that ZNF509L activates p21/CDKN1A transcription, while ZNF509S1 induces RB. ZNF509L binds to the p21/CDKN1A promoter either alone or by interacting with MIZ-1 to recruit the co-activator p300 to activate p21/CDKN1A transcription. In contrast, ZNF509S1 binds to the distal RB promoter to interact and interfere with the MIZF repressor, resulting in derepression and transcription of RB. Immunohistochemical analysis revealed that ZNF509 is highly expressed in normal epithelial cells, but was completely repressed in tumor tissues of the colon, lung and skin, indicating a possible role as a tumor suppressor.


Subject(s)
Cell Cycle Checkpoints , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA-Binding Proteins/metabolism , Retinoblastoma Protein/genetics , Transcription Factors/metabolism , Transcriptional Activation , Cell Line , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , DNA Damage , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/metabolism , Neoplasms/metabolism , Promoter Regions, Genetic , Protein Isoforms/biosynthesis , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Retinoblastoma Protein/biosynthesis , Stress, Physiological/genetics , Transcription Factors/biosynthesis , Transcription Factors/chemistry , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Zinc Fingers , p300-CBP Transcription Factors/metabolism
9.
J Biol Chem ; 289(7): 4018-31, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24382891

ABSTRACT

HKR3 (Human Krüppel-related 3) is a novel POK (POZ-domain Krüppel-like zinc-finger) family transcription factor. Recently, some of the POK (POZ-domain Krüppel-like zinc finger) family proteins have been shown to play roles in cell cycle arrest, apoptosis, cell proliferation, and oncogenesis. We investigated whether HKR3, an inhibitor of cell proliferation and an uncharacterized POK family protein, could regulate the cell cycle by controlling expression of genes within the p53 pathway (ARF-MDM2-TP53-p21WAF/CDKN1A). HKR3 potently activated the transcription of the tumor suppressor gene ARF by acting on the proximal promoter region (bp, -149∼+53), which contains Sp1 and FBI-1 binding elements (FREs). HKR3 interacted with the co-activator p300 to activate ARF transcription, which increased the acetylation of histones H3 and H4 within the proximal promoter. Oligonucleotide pull-down assays and ChIP assays revealed that HKR3 interferes with the binding of the proto-oncogenic transcription repressor FBI-1 to proximal FREs, thus derepressing ARF transcription.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Response Elements/physiology , Transcription Factors/metabolism , Transcription, Genetic/physiology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
10.
J Biol Chem ; 289(27): 18625-40, 2014 Jul 04.
Article in English | MEDLINE | ID: mdl-24821727

ABSTRACT

Promyelocytic leukemia zinc finger (PLZF) is a transcription repressor that was initially isolated as a fusion protein with retinoic acid receptor α. PLZF is aberrantly overexpressed in various human solid tumors, such as clear cell renal carcinoma, glioblastoma, and seminoma. PLZF causes cellular transformation of NIH3T3 cells and increases cell proliferation in several cell types. PLZF also increases tumor growth in the mouse xenograft tumor model. PLZF may stimulate cell proliferation by controlling expression of the genes of the p53 pathway (ARF, TP53, and CDKN1A). We found that PLZF can directly repress transcription of CDKN1A encoding p21, a negative regulator of cell cycle progression. PLZF binds to the proximal Sp1-binding GC-box 5/6 and the distal p53-responsive elements of the CDKN1A promoter to repress transcription. Interestingly, PLZF interacts with Sp1 or p53 and competes with Sp1 or p53. PLZF interacts with corepressors, such as mSin3A, NCoR, and SMRT, thereby deacetylates Ac-H3 and Ac-H4 histones at the CDKN1A promoter, which indicated the involvement of the corepressor·HDACs complex in transcription repression by PLZF. Also, PLZF represses transcription of TP53 and also decreases p53 protein stability by ubiquitination. PLZF may act as a potential proto-oncoprotein in various cell types.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/metabolism , Repressor Proteins/metabolism , Acetylation , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Gene Knockdown Techniques , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Mice , Promoter Regions, Genetic/genetics , Promyelocytic Leukemia Zinc Finger Protein , Repressor Proteins/deficiency , Repressor Proteins/genetics , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
11.
Nucleic Acids Res ; 41(13): 6403-20, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23658227

ABSTRACT

The tumour-suppressor gene CDKN1A (encoding p21Waf/Cip1) is thought to be epigenetically repressed in cancer cells. FBI-1 (ZBTB7A) is a proto-oncogenic transcription factor repressing the alternative reading frame and p21WAF/CDKN1A genes of the p53 pathway. FBI-1 interacts directly with MBD3 (methyl-CpG-binding domain protein 3) in the nucleus. We demonstrated that FBI-1 binds both non-methylated and methylated DNA and that MBD3 is recruited to the CDKN1A promoter through its interaction with FBI-1, where it enhances transcriptional repression by FBI-1. FBI-1 also interacts with the co-repressors nuclear receptor corepressor (NCoR), silencing mediator for retinoid and thyroid receptors (SMRT) and BCL-6 corepressor (BCoR) to repress transcription. MBD3 regulates a molecular interaction between the co-repressor and FBI-1. MBD3 decreases the interaction between FBI-1 and NCoR/SMRT but increases the interaction between FBI-1 and BCoR. Because MBD3 is a subunit of the Mi-2 autoantigen (Mi-2)/nucleosome remodelling and histone deacetylase (NuRD)-HDAC complex, FBI-1 recruits the Mi-2/NuRD-HDAC complex via MBD3. BCoR interacts with the Mi-2/NuRD-HDAC complex, DNMTs and HP1. MBD3 and BCoR play a significant role in the recruitment of the Mi-2/NuRD-HDAC complex- and the NuRD complex-associated proteins, DNMTs and HP. By recruiting DNMTs and HP1, Mi-2/NuRD-HDAC complex appears to play key roles in epigenetic repression of CDKN1A by DNA methylation.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Methylation , DNA-Binding Proteins/metabolism , Gene Silencing , Transcription Factors/metabolism , Cell Line , Cells, Cultured , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , HEK293 Cells , Histones/metabolism , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Transcription, Genetic
12.
J Biol Chem ; 288(10): 7053-64, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23329847

ABSTRACT

The human POZ domain and Krüppel-like zinc finger (POK) family proteins play important roles in the regulation of apoptosis, cell proliferation, differentiation, development, oncogenesis, and tumor suppression. A novel POK family transcription factor, BTB/POZ and zinc finger domains factor on chromosome 1 (BOZF-1; also called ZBTB8A), contains a POZ domain and two C2H2-type Krüppel-like zinc fingers and is localized at nuclear speckles. Compared with paired normal tissues, BOZF1 expression is increased in cancer tissues of the prostate, breast, and cervix. BOZF1 repressed the transcription of p21WAF/CDKN1A by acting on the proximal promoter concentrated with Sp1-binding GC boxes. BOZF1 competed with Sp1 in binding to GC boxes 1-5/6 of the CDKN1A proximal promoter. In addition, BOZF1 interacted with p53 and decreased the acetylation of p53 by p300, which reduced the DNA binding activity of p53 at the far distal p53-binding element. BOZF1 blocked the two major molecular events that are important in both constitutive and inducible transcription activation of CDKN1A. BOZF1 is unique in that it bound to all the proximal GC boxes to repress transcription, and it inhibited p53 acetylation without affecting p53 stability. BOZF1 might be a novel proto-oncoprotein that stimulates cell proliferation.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Kruppel-Like Transcription Factors/metabolism , Sp1 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Animals , Binding Sites/genetics , Binding, Competitive , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation , HCT116 Cells , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/genetics , Mice , NIH 3T3 Cells , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sp1 Transcription Factor/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/genetics
13.
Adv Sci (Weinh) ; 10(17): e2204378, 2023 06.
Article in English | MEDLINE | ID: mdl-37097643

ABSTRACT

Immune checkpoint inhibitor (ICI) clinically benefits cancer treatment. However, the ICI responses are only achieved in a subset of patients, and the underlying mechanisms of the limited response remain unclear. 160 patients with non-small cell lung cancer treated with anti-programmed cell death protein-1 (anti-PD-1) or anti-programmed death ligand-1 (anti-PD-L1) are analyzed to understand the early determinants of response to ICI. It is observed that high levels of intracellular adhesion molecule-1 (ICAM-1) in tumors and plasma of patients are associated with prolonged survival. Further reverse translational studies using murine syngeneic tumor models reveal that soluble ICAM-1 (sICAM-1) is a key molecule that increases the efficacy of anti-PD-1 via activation of cytotoxic T cells. Moreover, chemokine (CXC motif) ligand 13 (CXCL13) in tumors and plasma is correlated with the level of ICAM-1 and ICI efficacy, suggesting that CXCL13 might be involved in the ICAM-1-mediated anti-tumor pathway. Using sICAM-1 alone and in combination with anti-PD-1 enhances anti-tumor efficacy in anti-PD-1-responsive tumors in murine models. Notably, combinatorial therapy with sICAM-1 and anti-PD-1 converts anti-PD-1-resistant tumors to responsive ones in a preclinical study. These findings provide a new immunotherapeutic strategy for treating cancers using ICAM-1.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Mice , Animals , CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Intercellular Adhesion Molecule-1
14.
J Lipid Res ; 53(4): 755-66, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22331133

ABSTRACT

Kr-pok (kidney cancer-related POZ domain and Krüppel-like protein) is a new proto-oncogenic POZ-domain transcription factor. Fatty acid synthase gene (FASN) encodes one of the key enzymes in fatty acids synthesis and is the only enzyme that synthesizes fatty acids in cancer cells. Sp1 and SREBP-1c are the two major transcription activators of FASN. We investigated whether Kr-pok modulates transcription of the FASN. FASN expression is significantly decreased in Kr-pok knockout murine embryonic fibroblasts. Coimmunoprecipitation, GST fusion protein pull-down, and immunocytochemistry assays show that the zinc-finger domain of Kr-pok interacts directly with the bZIP DNA binding domain of SREBP-1. Electrophoretic mobility shift assay, oligonucleotide pull-down, and chromatin immunoprecipitation assays showed that Kr-pok changes the transcription factor binding dynamics of Sp1 and SREBP-1c to the SRE/E-box elements of the proximal promoter. We found that Kr-pok expression increased during 3T3-L1 preadipocyte differentiation and that FASN expression is decreased by the knockdown of Kr-pok. Kr-pok facilitates the SREBP-1c-mediated preadipocyte differentiation and/or fatty acid synthesis. Kr-pok may act as an important regulator of fatty acid synthesis and may induce rapid cancer cell proliferation by increasing palmitate synthesis.


Subject(s)
Fatty Acid Synthase, Type I/metabolism , Promoter Regions, Genetic , Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , 3T3-L1 Cells , Animals , Cell Dedifferentiation , Cell Differentiation , Cell Proliferation , Doxycycline/pharmacology , Electrophoretic Mobility Shift Assay , Enzyme Activation , Fatty Acid Synthase, Type I/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HCT116 Cells , HEK293 Cells , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Male , Mice , Pregnancy , Protein Binding , Protein Interaction Domains and Motifs , Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Transcriptional Activation
15.
Cancers (Basel) ; 13(5)2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33668827

ABSTRACT

Colorectal cancer (CRC) is among the leading causes of cancer-related death in the world. The development of CRC is associated with smoking, diet, and microbial exposure. Previous studies have shown that dysbiosis of the gut microbiome affects cancer development, because it leads to inflammation and genotoxicity. Supplementation with specific microbiota induces anti-tumor effects by enhancing of anti-tumor immunity. Here, we observed that supplementation with either of two B. breve strains reduces tumor growth in MC38 colon carcinoma-bearing mice. Interestingly, only one B. breve strain boosted the efficacy of cancer therapeutics, including oxaliplatin and PD-1 blockade. Extensive immune profiling and transcriptomic analysis revealed that the boosting B. breve strain augments lymphocyte-mediated anti-cancer immunity. Our results suggest that supplementation with B. breve strains could potentially be used as a strategy to enhance the efficacy of CRC therapeutics.

16.
Exp Mol Med ; 53(5): 917-932, 2021 05.
Article in English | MEDLINE | ID: mdl-34017061

ABSTRACT

Zbtb7c is a proto-oncoprotein that controls the cell cycle and glucose, glutamate, and lipid metabolism. Zbtb7c expression is increased in the liver and white adipose tissues of aging or high-fat diet-fed mice. Knockout or knockdown of Zbtb7c gene expression inhibits the adipocyte differentiation of 3T3-L1 cells and decreases adipose tissue mass in aging mice. We found that Zbtb7c was a potent transcriptional repressor of SIRT1 and that SIRT1 was derepressed in various tissues of Zbtb7c-KO mice. Mechanistically, Zbtb7c interacted with p53 and bound to the proximal promoter p53RE1 and p53RE2 to repress the SIRT1 gene, in which p53RE2 was particularly critical. Zbtb7c induced p53 to interact with the corepressor mSin3A-HADC1 complex at p53RE. By repressing the SIRT1 gene, Zbtb7c increased the acetylation of Pgc-1α and Pparγ, which resulted in repression or activation of Pgc-1α or Pparγ target genes involved in lipid metabolism. Our study provides a molecular target that can overexpress SIRT1 protein in the liver, pancreas, and adipose tissues, which can be beneficial in the treatment of diabetes, obesity, longevity, etc.


Subject(s)
Aging/genetics , Aging/metabolism , Diet, High-Fat , Intracellular Signaling Peptides and Proteins/genetics , Obesity/etiology , Obesity/metabolism , Sirtuin 1/genetics , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , Biomarkers , Diet, High-Fat/adverse effects , Disease Models, Animal , Disease Susceptibility , Gene Expression Regulation , Gene Expression Regulation, Enzymologic , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism , Mice , Mice, Knockout , Obesity/pathology , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protein Binding , Response Elements , Sirtuin 1/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Nat Microbiol ; 6(3): 277-288, 2021 03.
Article in English | MEDLINE | ID: mdl-33432149

ABSTRACT

The gut microbiome can influence the development of tumours and the efficacy of cancer therapeutics1-5; however, the multi-omics characteristics of antitumour bacterial strains have not been fully elucidated. In this study, we integrated metagenomics, genomics and transcriptomics of bacteria, and analyses of mouse intestinal transcriptome and serum metabolome data to reveal an additional mechanism by which bacteria determine the efficacy of cancer therapeutics. In gut microbiome analyses of 96 samples from patients with non-small-cell lung cancer, Bifidobacterium bifidum was abundant in patients responsive to therapy. However, when we treated syngeneic mouse tumours with commercial strains of B. bifidum to establish relevance for potential therapeutic uses, only specific B. bifidum strains reduced tumour burden synergistically with PD-1 blockade or oxaliplatin treatment by eliciting an antitumour host immune response. In mice, these strains induced tuning of the immunological background by potentiating the production of interferon-γ, probably through the enhanced biosynthesis of immune-stimulating molecules and metabolites.


Subject(s)
Bifidobacterium bifidum/physiology , Immune Checkpoint Inhibitors/therapeutic use , Probiotics/therapeutic use , Tumor Burden/drug effects , Animals , Bifidobacterium bifidum/classification , Bifidobacterium bifidum/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/pathology , Drug Therapy, Combination , Gastrointestinal Microbiome , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/microbiology , Lung Neoplasms/pathology , Metabolome/drug effects , Mice , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Probiotics/administration & dosage , Species Specificity , Transcriptome/drug effects , Tryptophan/metabolism
18.
J Biol Chem ; 284(27): 17935-46, 2009 Jul 03.
Article in English | MEDLINE | ID: mdl-19380588

ABSTRACT

We found that ZBTB2, a POK family transcription factor, is a potent repressor of the ARF-HDM2-p53-p21 pathway important in cell cycle regulation. ZBTB2 repressed transcription of the ARF, p53, and p21 genes, but activated the HDM2 gene. In particular, ZBTB2 repressed transcription of the p21 gene by acting on the two distal p53 binding elements and the proximal Sp1 binding GC-box 5/6 elements. ZBTB2 directly interacted with Sp1 via its POZ domain and zinc fingers, which was important in the repression of transcription activation by Sp1. ZBTB2 and Sp1 competed with each other in binding to the GC-box 5/6 elements and the two p53 binding elements. ZBTB2 directly interacted with p53 via its zinc fingers, inhibiting p53 binding and repressing transcription activation by p53. The POZ domain, required for transcription repression, interacted with corepressors such as BCoR, NCoR, and SMRT. The interactions deacetylated histones Ac-H3 and -H4 at the proximal promoter. Although ectopic ZBTB2 stimulated cell proliferation, knock-down of ZBTB2 expression decreased cell proliferation and DNA synthesis. Overall, our data suggest that ZBTB2 is a potential proto-oncogenic master control gene of the p53 pathway and, in particular, is a potent transcription repressor of the cell cycle arrest gene p21 by inhibiting p53 and Sp1.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , ADP-Ribosylation Factor 1/genetics , ADP-Ribosylation Factor 1/metabolism , Acetylation , Animals , Binding, Competitive/physiology , Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins/chemistry , Drosophila , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Kidney/cytology , Mice , Mice, Inbred Strains , Promoter Regions, Genetic/physiology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Messenger/metabolism , Repressor Proteins/chemistry , S Phase/physiology , Sp1 Transcription Factor/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/physiology , Two-Hybrid System Techniques
19.
Sci Rep ; 10(1): 7620, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32376896

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a major cellular process in which epithelial cells lose cell polarity and cell-cell adhesion and become motility and invasiveness by transforming into mesenchymal cells. Catechol is one of the natural compounds present in fruits and vegetables and has various pharmacological and physiological activities including anti-carcinogenic effects. However, the effects of catechol on EMT has not been reported. Epidermal growth factor (EGF) is one of the growth factors and is known to play a role in inducing EMT. The present study showed that catechol suppressed not only the morphological changes to the mesenchymal phenotype of epithelial HCC cells, but also the reduction of E-cadherin and the increment of Vimentin, which are typical hallmark of EMT. In addition, catechol suppressed EMT-related steps such as migration, invasion, anoikis resistance acquisition, and stem cell-like characterization through the EGFR-AKT-ERK signaling pathway during liver cancer metastasis. Therefore, these results suggest that catechol may be able to regulate the early metastasis of liver cancer in vitro.


Subject(s)
Carcinoma, Hepatocellular/pathology , Catechols/pharmacology , Epidermal Growth Factor/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Liver Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Enzyme Activation/drug effects , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Matrix Metalloproteinase 2/metabolism , Neoplasm Invasiveness , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Snail Family Transcription Factors/metabolism
20.
Cell Physiol Biochem ; 23(4-6): 359-70, 2009.
Article in English | MEDLINE | ID: mdl-19471103

ABSTRACT

FBI-1, a member of the POK (POZ and Kruppel) family of transcription factors, plays a role in differentiation, oncogenesis, and adipogenesis. eEF1A is a eukaryotic translation elongation factor involved in several cellular processes including embryogenesis, oncogenic transformation, cell proliferation, and cytoskeletal organization. CCS-3, a potential cervical cancer suppressor, is an isoform of eEF1A. We found that eEF1A forms a complex with FBI-1 by co-immunoprecipitation, SDS-PAGE, and MALDI-TOF Mass analysis of the immunoprecipitate. GST fusion protein pull-downs showed that FBI-1 directly interacts with eEF1A and CCS-3 via the zinc finger and POZ-domain of FBI-1. FBI-1 co-localizes with either eEF1A or CCS-3 at the nuclear periplasm. CCS-3 enhances transcriptional repression of the p21CIP1 gene (hereafter referred to as p21) by FBI-1. The POZ-domain of FBI-1 interacts with the co-repressors, SMRT and BCoR. We found that CCS-3 also interacts with the co-repressors independently. The molecular interaction between the co-repressors and CCS-3 at the POZ-domain of FBI-1 appears to enhance FBI-1 mediated transcriptional repression. Our data suggest that CCS-3 may be important in cell differentiation, tumorigenesis, and oncogenesis by interacting with the proto-oncogene FBI-1 and transcriptional co-repressors.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins/metabolism , Peptide Elongation Factor 1/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Cell Differentiation , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA-Binding Proteins/genetics , Humans , Immunoprecipitation , Molecular Sequence Data , Peptide Elongation Factor 1/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Proto-Oncogene Mas , Transcription Factors/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL