Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 19(8): 809-820, 2018 08.
Article in English | MEDLINE | ID: mdl-29967452

ABSTRACT

Regulatory factor X 7 (Rfx7) is an uncharacterized transcription factor belonging to a family involved in ciliogenesis and immunity. Here, we found that deletion of Rfx7 leads to a decrease in natural killer (NK) cell maintenance and immunity in vivo. Genomic approaches showed that Rfx7 coordinated a transcriptional network controlling cell metabolism. Rfx7-/- NK lymphocytes presented increased size, granularity, proliferation, and energetic state, whereas genetic reduction of mTOR activity mitigated those defects. Notably, Rfx7-deficient NK lymphocytes were rescued by interleukin 15 through engagement of the Janus kinase (Jak) pathway, thus revealing the importance of this signaling for maintenance of such spontaneously activated NK cells. Rfx7 therefore emerges as a novel transcriptional regulator of NK cell homeostasis and metabolic quiescence.


Subject(s)
Interleukin-15/metabolism , Killer Cells, Natural/metabolism , Regulatory Factor X1/metabolism , Animals , Cell Proliferation , Cell Survival , Cells, Cultured , Chimera , Energy Metabolism , Gene Regulatory Networks , Immunity, Cellular/genetics , Immunity, Innate/genetics , Janus Kinases/metabolism , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Regulatory Factor X1/genetics , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
2.
Mol Microbiol ; 121(4): 727-741, 2024 04.
Article in English | MEDLINE | ID: mdl-38183361

ABSTRACT

Adhesion to mucosal surfaces is a critical step in many bacterial and fungal infections. Here, using a mouse model of oral infection by the human fungal pathobiont Candida albicans, we report the identification of a novel regulator of C. albicans adhesion to the oral mucosa. The regulator is a member of the regulatory factor X (RFX) family of transcription factors, which control cellular processes ranging from genome integrity in model yeasts to tissue differentiation in vertebrates. Mice infected with the C. albicans rfx1 deletion mutant displayed increased fungal burden in tongues compared to animals infected with the reference strain. High-resolution imaging revealed RFX1 transcripts being expressed by C. albicans cells during infection. Concomitant with the increase in fungal burden, the rfx1 mutant elicited an enhanced innate immune response. Transcriptome analyses uncovered HWP1, a gene encoding an adhesion protein that mediates covalent attachment to buccal cells, as a major RFX1-regulated locus. Consistent with this result, we establish that C. albicans adhesion to oral cells is modulated by RFX1 in an HWP1-dependent manner. Our findings expand the repertoire of biological processes controlled by the RFX family and illustrate a mechanism whereby C. albicans can adjust adhesion to the oral epithelium.


Subject(s)
Candida albicans , Fungal Proteins , Regulatory Factor X1 , Animals , Humans , Candida albicans/genetics , Epithelium/microbiology , Fungal Proteins/genetics , Fungal Proteins/metabolism , Mouth Mucosa/microbiology , Regulatory Factor X1/genetics , Regulatory Factor X1/metabolism
3.
BMC Cancer ; 22(1): 1019, 2022 Sep 26.
Article in English | MEDLINE | ID: mdl-36163022

ABSTRACT

PURPOSE: To investigate the expression and prognostic value of c-Jun in hypopharyngeal squamous cell carcinoma (HPSCC). METHODS: A retrospective study was performed on a cohort of 99 HPSCC patients. The expression of c-Jun and phosphorylated-c-Jun (p-c-Jun) was evaluated via immunohistochemistry (IHC) staining. Overall survival (OS) and progression-free survival (PFS) were assessed using Kaplan-Meier method and multivariate Cox regression analysis. RESULTS: The high expression of c-Jun and p-c-Jun in HPSCC accounted for 60.61% and 16.16%, respectively. High expression of c-Jun was closely associated with cT stage (p = 0.0401), tumor size (p = 0.0276), number of lymph node metastases (p = 0.0205) and pathological differentiation (p = 0.0108). The expression of c-Junhigh (p = 0.0043), p-c-Junhigh (p = 0.0376) and c-Junhigh/p-c-Junhigh were closely associated with poor OS. The Cox proportional multivariate hazard model revealed that lymphovascular invasion and c-Jun expression were independent influencing factors of OS in HPSCC patients. CONCLUSION: Our findings suggest that c-Jun is a reliable prognostic factors in HPSCC patients.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Hypopharyngeal Neoplasms , Carcinoma, Squamous Cell/pathology , Follow-Up Studies , Humans , Hypopharyngeal Neoplasms/pathology , Immunologic Factors , Prognosis , Proto-Oncogene Proteins c-jun , Regulatory Factor X1 , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck
4.
Nucleic Acids Res ; 48(16): 9019-9036, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32725242

ABSTRACT

Cilia assembly is under strict transcriptional control during animal development. In vertebrates, a hierarchy of transcription factors (TFs) are involved in controlling the specification, differentiation and function of multiciliated epithelia. RFX TFs play key functions in the control of ciliogenesis in animals. Whereas only one RFX factor regulates ciliogenesis in C. elegans, several distinct RFX factors have been implicated in this process in vertebrates. However, a clear understanding of the specific and redundant functions of different RFX factors in ciliated cells remains lacking. Using RNA-seq and ChIP-seq approaches we identified genes regulated directly and indirectly by RFX1, RFX2 and RFX3 in mouse ependymal cells. We show that these three TFs have both redundant and specific functions in ependymal cells. Whereas RFX1, RFX2 and RFX3 occupy many shared genomic loci, only RFX2 and RFX3 play a prominent and redundant function in the control of motile ciliogenesis in mice. Our results provide a valuable list of candidate ciliary genes. They also reveal stunning differences between compensatory processes operating in vivo and ex vivo.


Subject(s)
Cilia/physiology , Ependyma/cytology , Gene Expression Regulation, Developmental , Regulatory Factor X Transcription Factors/physiology , Regulatory Factor X1/physiology , Animals , Cilia/genetics , Mice , Mice, Inbred C57BL
5.
Int Heart J ; 63(4): 763-772, 2022 Jul 30.
Article in English | MEDLINE | ID: mdl-35831154

ABSTRACT

Atherosclerosis (AS) is a common etiology of cardiovascular disease. As an emerging functional biomarker, circular RNAs (circRNAs) are involved in various diseases, including cardiovascular disease. However, the mechanism of action of circ_0030042 in AS has not been reported.Human umbilical vein endothelial cells (HUVECs) stimulated by ox-LDL served as a cellular model of AS. Gene expression was detected using quantitative real-time polymerase chain reaction. The influence of circ_0030042 on cell viability, proliferation, and apoptosis was verified using Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine, and flow cytometry assays. An enzyme-linked immunosorbent assay was performed to measure the contents of tumor necrosis factor-α, interleukin (IL) -6, and IL-1ß. Western blot assay was utilized to determine the protein levels of Bax, Bcl-2, PCNA, and regulatory factor X 7 (RFX7). The interrelationship between miR-616-3p and circ_0030042 or RFX7 was validated using dual-luciferase reporter, RNA immunoprecipitation, and RNA pull-down assays.The expression of circ_0030042 was downregulated in ox-LDL-induced HUVECs. It was found that overexpression of circ_0030042 facilitated cell proliferation, repressed apoptosis, and reduced the level of inflammatory factors in HUVECs. Circ_0030042 and miR-616-3p had a targeting relationship, and the miR-616-3p mimic eliminated the effects of overexpressed circ_0030042 on ox-LDL-induced HUVECs. RFX7 was a downstream gene of miR-616-3p and was lowly expressed in ox-LDL-induced HUVECs. The miR-616-3p inhibitor stimulated cell proliferation, arrested apoptosis, and caused a decline in the levels of inflammatory factors, whereas knockdown of RFX7 abolished the effects.Circ_0030042 weakened ox-LDL-induced HUVEC injury by regulating the miR-616-3p/RFX7 pathway, thereby influencing AS progression. Circ_0030042 is likely to be a potential biomarker for the future treatment of patients with AS.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , MicroRNAs , Apoptosis , Atherosclerosis/metabolism , Cardiovascular Diseases/metabolism , Cell Proliferation , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , Regulatory Factor X1/metabolism
6.
J Cell Physiol ; 236(10): 6884-6896, 2021 10.
Article in English | MEDLINE | ID: mdl-33655492

ABSTRACT

Cellular communication network factor (CCN) family members are multifunctional matricellular proteins that manipulate and integrate extracellular signals. In our previous studies investigating the role of CCN family members in cellular metabolism, we found three members that might be under the regulation of energy metabolism. In this study, we confirmed that CCN2 and CCN3 are the only members that are tightly regulated by glycolysis in human chondrocytic cells. Interestingly, CCN3 was induced under a variety of impaired glycolytic conditions. This CCN3 induction was also observed in two breast cancer cell lines with a distinct phenotype, suggesting a basic role of CCN3 in cellular metabolism. Reporter gene assays indicated a transcriptional regulation mediated by an enhancer in the proximal promoter region. As a result of analyses in silico, we specified regulatory factor binding to the X-box 1 (RFX1) as a candidate that mediated the transcriptional activation by impaired glycolysis. Indeed, the inhibition of glycolysis induced the expression of RFX1, and RFX1 silencing nullified the CCN3 induction by impaired glycolysis. Subsequent experiments with an anti-CCN3 antibody indicated that CCN3 supported the survival of chondrocytes under impaired glycolysis. Consistent with these findings in vitro, abundant CCN3 production by chondrocytes in the deep zones of developing epiphysial cartilage, which are located far away from the synovial fluid, was confirmed in vivo. Our present study uncovered that RFX1 is the mediator that enables CCN3 induction upon cellular starvation, which may eventually assist chondrocytes in retaining their viability, even when there is an energy supply shortage.


Subject(s)
Chondrocytes/metabolism , Glycolysis , Nephroblastoma Overexpressed Protein/metabolism , Regulatory Factor X1/metabolism , Animals , Cell Line, Tumor , Cell Survival , Chondrocytes/drug effects , Gene Expression Regulation , Gestational Age , Glycolysis/drug effects , Humans , Joints/embryology , Joints/metabolism , Mice, Inbred BALB C , Nephroblastoma Overexpressed Protein/genetics , Regulatory Factor X1/genetics , Sodium Fluoride/pharmacology
7.
Biochem Biophys Res Commun ; 509(4): 1015-1020, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30654936

ABSTRACT

RFX proteins are a family of conserved DNA binding proteins involved in various, essential cellular and developmental processes. RFX1 is a ubiquitously expressed, dual-activity transcription factor capable of both activation and repression of target genes. The exact mechanism by which RFX1 regulates its target is not known yet. In this work, we show that the C-terminal repression domain of RFX1 interacts with the Serine/Threonine protein phosphatase PP1c, and that interaction with RFX1 can target PP1c to specific sites in the genome. Given that PP1c was shown to de-phosphorylate several transcription factors, as well as the regulatory C-terminal domain of RNA Polymerase II the recruitment of PP1c to promoters may be a mechanism by which RFX1 regulates the target genes.


Subject(s)
Promoter Regions, Genetic , Protein Phosphatase 1/metabolism , Regulatory Factor X1/metabolism , Animals , Biological Transport , Catalytic Domain , Gene Expression Regulation , Humans , Phosphorylation , Protein Binding , Protein Domains , Transcription Factors/metabolism
8.
Evol Dev ; 20(6): 233-243, 2018 11.
Article in English | MEDLINE | ID: mdl-30259625

ABSTRACT

Cilia are complex organelles involved in sensory perception and motility with intraflagellar transport (IFT) proteins being essential for cilia assembly and function, but little is known about cilia in an evo-devo context. For example, recent comparisons revealed conservation and divergence of IFT components in the regulation of social feeding behaviors between the nematodes Caenorhabditis elegans and Pristionchus pacificus. Here, we focus on the P. pacificus RFX transcription factor daf-19, the master regulator of ciliogenesis in C. elegans. Two CRISPR/Cas9-induced Ppa-daf-19 mutants lack ciliary structures in amphid neurons and display chemosensory defects. In contrast to IFT mutants, Ppa-daf-19 mutants do not exhibit social behavior. However, they show weak locomotive responses to shifts in oxygen concentration, suggesting partial impairment in sensing or responding to oxygen. To identify targets of Ppa-daf-19 regulation we compared the transcriptomes of Ppa-daf-19 and wild-type animals and performed a bioinformatic search for the X-box RFX binding-site across the genome. The regulatory network of Ppa-DAF-19 involves IFT genes but also many taxonomically restricted genes. We identified a conserved X-box motif as the putative binding site, which was validated for the Ppa-dyf-1 gene. Thus, Ppa-DAF-19 controls ciliogenesis, influences oxygen-induced behaviors and displays a high turnover of its regulatory network.


Subject(s)
Regulatory Factor X1/genetics , Rhabditida/cytology , Rhabditida/genetics , Transcription Factors/genetics , Animals , Cilia/metabolism , Oxygen/metabolism , Regulatory Factor X1/metabolism , Rhabditida/classification , Rhabditida/metabolism , Social Behavior , Transcription Factors/metabolism
9.
Clin Immunol ; 169: 1-7, 2016 08.
Article in English | MEDLINE | ID: mdl-27283392

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease caused by complex interactions between genes and the environment. The expression level of transcription factor regulatory factor X 1 (RFX1) is reduced in T cells from SLE patients. RFX1 can regulate epigenetic modifications of CD70 and CD11a and plays an important role in the development of SLE. However, the mechanisms that mediate reduction of RFX1 in SLE are unclear. Here, we demonstrate that RFX1 protein expression can be tightly regulated by polyubiquitination-mediated proteosomal degradation via STIP1 homology and U-box containing protein 1 (STUB1). The E3 ligase STUB1 is upregulated in CD4(+)T cells of SLE patients compared to healthy subjects. Overexpression of STUB1 in CD4(+)T cells leads to upregulation of levels of CD70 and CD11a in T cells. The modulation of STUB1 activity may provide a novel therapeutic approach for SLE.


Subject(s)
Lupus Erythematosus, Systemic/metabolism , Regulatory Factor X1/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Adult , CD11a Antigen/genetics , CD11a Antigen/metabolism , CD27 Ligand/genetics , CD27 Ligand/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Female , Gene Expression , HEK293 Cells , Humans , Immunoblotting , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Male , Proteasome Endopeptidase Complex/metabolism , RNA Interference , Regulatory Factor X1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics
10.
Mol Reprod Dev ; 83(7): 606-14, 2016 07.
Article in English | MEDLINE | ID: mdl-27228460

ABSTRACT

Formation and maintenance of testis cords during embryogenesis are essential for establishing testicular structure and function in adults. At least five genes (Wt1, Dhh, Sox8/Sox9, and Dax1) appear to be required for the maintenance of testis cord integrity in mice. Here, we report that RFX1 is specifically expressed in fetal Sertoli cells. Mouse embryos conditionally deficient in Rfx1 (Rfx1(flox/flox) , Amh-Cre) possessed disrupted testis cords, as the basal lamina lining was fragmented or completely absent in some areas of the testes. Spermatogenesis was blocked, leading to complete infertility. Expression of integrin alpha-6 was significantly decreased in Rfx1-deficient testes compared to control testes; indeed, luciferase and chromatin immunoprecipitation assays indicated that RFX1 directly activates transcription of Itga6 (the gene coding for integrin alpha-6). Taken together, RFX1 transcriptionally targets Itga6 in Sertoli cells, thereby, helping maintain the integrity of the basal lamina during testis cord development. Mol. Reprod. Dev. 83: 606-614, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental/physiology , Integrin alpha6/biosynthesis , Regulatory Factor X1/metabolism , Sertoli Cells/metabolism , Transcription, Genetic/physiology , Animals , Embryo, Mammalian/cytology , Integrin alpha6/genetics , Male , Mice , Mice, Knockout , Regulatory Factor X1/genetics , Sertoli Cells/cytology , Spermatogenesis/physiology
11.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 41(11): 1117-1123, 2016 Nov 28.
Article in Zh | MEDLINE | ID: mdl-27932754

ABSTRACT

OBJECTIVE: To construct overexpression lentivirus vector for human regulatory factor X1 (RFX1) gene, and to explore its effect on proliferation of F98 cell line.
 Methods: Huamn RFX1 gene was amplified by polymerase reaction. Gene amplification products were inserted into lentivirus vector pITA, and the lentivirus vector pITA-RFX1 was constructed. The constructed vector was verified by agarose gel electrophoresis and DNA sequencing. Lentivirus vector pITA-RFX1 and virus packaging plasmids were cotransfected into 293T cells, and then transfected into F98 cells. RFX1 protein expression were detected by Western blot and laser confocal before and after transfection. Flow cytometry and cell counting kit-8 were used to detect cellular proliferation.
 Results: Agarose gel electrophoresis and DNA sequencing showed that recombinant lentivirus plasmids pITA-RFX1 were constructed successfully. After transfection of pITA-RFX1, the RFX1 protein were over-expressed, which significantly inhibited the proliferation of F98 cells.
 Conclusion: The overexpression lentivirus vector for RFX1 was constrcted successfully, and the up-regulation of RFX1 can prevent the proliferation of glioblastoma cells.


Subject(s)
Glioblastoma/genetics , Glioblastoma/physiopathology , Regulatory Factor X1/physiology , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Genetic Vectors , HEK293 Cells , Humans , Lentivirus/genetics , Plasmids , Rats , Regulatory Factor X1/genetics , Transfection , Up-Regulation
12.
Carcinogenesis ; 35(12): 2807-14, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25322871

ABSTRACT

Regulatory factor X-1 (RFX-1) is a transcription factor that has been linked to negative regulation of tumor progression; however, its biological function and signaling cascades are unknown. Here, we performed several studies to elucidate the roles of RFX-1 in the regulation of SHP-1 in hepatocellular carcinoma (HCC) cells. Overexpression of RFX-1 resulted in the activation of SHP-1 and repressed colony formation of HCC cells. In addition, by a mouse xenograft model, we demonstrated that RFX-1 overexpression also inhibited the tumor growth of HCC cells in vivo, suggesting that RFX-1 is of potential interest for small-molecule-targeted therapy. We also found that SC-2001, a bipyrrole molecule, induced apoptosis in HCC cells through activating RFX-1 expression. SC-2001 induced RFX-1 translocation from the cytosol to nucleus, bound to the SHP-1 promoter, and activated SHP-1 transcription. In a xenograft model, knockdown of RFX-1 reversed the antitumor effect of SC-2001. Notably, SC-2001 is much more potent than sorafenib, a clinically approved drug for HCC, in in vitro and in vivo assays. Our study confirmed that RFX-1 acts as a tumor suppressor in HCC and might be a new target for HCC therapy. The findings of this study also provide a new lead compound for targeted therapy via the activation of the RFX-1/SHP-1 pathway.


Subject(s)
Carcinoma, Hepatocellular/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , STAT3 Transcription Factor/antagonists & inhibitors , Transcription Factors/metabolism , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Chromatin Immunoprecipitation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Flow Cytometry , Humans , Immunoenzyme Techniques , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Luciferases/metabolism , Male , Mice , Mice, Nude , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Promoter Regions, Genetic/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Pyrroles/pharmacology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Regulatory Factor X Transcription Factors , Regulatory Factor X1 , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Sorafenib , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Breast Cancer Res Treat ; 146(1): 71-84, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24903225

ABSTRACT

Interfering oncogenic STAT3 signaling is a promising anti-cancer strategy. We examined the efficacy and drug mechanism of an obatoclax analog SC-2001, a novel STAT3 inhibitor, in human breast cancer cells. Human breast cancer cell lines were used for in vitro studies. Apoptosis was examined by both flow cytometry and western blot. Signaling pathways were assessed by western blot. In vivo efficacy of SC-2001 was tested in xenograft nude mice. SC-2001 inhibited cell growth and induced apoptosis in association with downregulation of p-STAT3 (Tyr 705) in breast cancer cells. STAT3-regulated proteins, including Mcl-1, survivin, and cyclin D1, were repressed by SC-2001. Over-expression of STAT3 in MDA-MB-468 cells protected cells from SC-2001-induced apoptosis. Moreover, SC-2001 enhanced the expression of protein tyrosine phosphatase SHP-1, a negative regulator of STAT3. Furthermore, the enhanced SHP-1 expression, in conjunction with increased SHP-1 phosphatase activity, was mediated by upregulated transcription by RFX-1. Chromatin immunoprecipitation assay revealed that SC-2001 increased the binding capacity of RFX-1 to the SHP-1 promoter. Knockdown of either RFX-1 or SHP-1 reduced SC-2001-induced apoptosis, whereas ectopic expression of RFX-1 increased SHP-1 expression and enhanced the apoptotic effect of SC-2001. Importantly, SC-2001 suppressed tumor growth in association with enhanced RFX-1 and SHP-1 expression and p-STAT3 downregulation in MDA-MB-468 xenograft tumors. SC-2001 induced apoptosis in breast cancer cells, an effect that was mediated by RFX-1 upregulated SHP-1 expression and SHP-1-dependent STAT3 inactivation. Our study indicates targeting STAT3 signaling pathway may be a useful approach for the development of targeted agents for anti-breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Pyrroles/pharmacology , STAT3 Transcription Factor/metabolism , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Pyrroles/administration & dosage , Regulatory Factor X Transcription Factors , Regulatory Factor X1 , Signal Transduction/drug effects , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
PLoS Genet ; 7(9): e1002277, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21935353

ABSTRACT

Understanding how silent genes can be competent for activation provides insight into development as well as cellular reprogramming and pathogenesis. We performed genomic location analysis of the pioneer transcription factor FoxA in the adult mouse liver and found that about one-third of the FoxA bound sites are near silent genes, including genes without detectable RNA polymerase II. Virtually all of the FoxA-bound silent sites are within conserved sequences, suggesting possible function. Such sites are enriched in motifs for transcriptional repressors, including for Rfx1 and type II nuclear hormone receptors. We found one such target site at a cryptic "shadow" enhancer 7 kilobases (kb) downstream of the Cdx2 gene, where Rfx1 restricts transcriptional activation by FoxA. The Cdx2 shadow enhancer exhibits a subset of regulatory properties of the upstream Cdx2 promoter region. While Cdx2 is ectopically induced in the early metaplastic condition of Barrett's esophagus, its expression is not necessarily present in progressive Barrett's with dysplasia or adenocarcinoma. By contrast, we find that Rfx1 expression in the esophageal epithelium becomes gradually extinguished during progression to cancer, i.e, expression of Rfx1 decreased markedly in dysplasia and adenocarcinoma. We propose that this decreased expression of Rfx1 could be an indicator of progression from Barrett's esophagus to adenocarcinoma and that similar analyses of other transcription factors bound to silent genes can reveal unanticipated regulatory insights into oncogenic progression and cellular reprogramming.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/pathology , DNA-Binding Proteins/genetics , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/metabolism , Homeodomain Proteins/genetics , Transcription Factors/genetics , Transcriptional Activation , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Barrett Esophagus/metabolism , Base Sequence , Binding Sites , CDX2 Transcription Factor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Enhancer Elements, Genetic , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Gene Silencing , Genome , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Humans , Liver/metabolism , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Regulatory Factor X Transcription Factors , Regulatory Factor X1
15.
Int Immunopharmacol ; 130: 111751, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38402833

ABSTRACT

BACKGROUND AND AIMS: Atherosclerosis (AS) is a continuously low-grade inflammatory disease, and monocyte-derived macrophages play a vital role in AS pathogenesis. Regulatory factor X1 (RFX1) has been reported to participate in differentiation of various cells. Our previous report showed that RFX1 expression in CD14+ monocytes from AS patients was decreased and closely related to AS development. Macrophages mostly derive from monocytes and play an important role in AS plaque formation and stability. However, the functions of RFX1 in the formation of macrophage-derived foam cells and consequent AS development are unclear. METHODS: We explored the effects of RFX1 on oxidation low lipoprotein (ox-LDL)-stimulated foam cell formation and CD36 expression by increasing or silencing Rfx1 expression in mouse peritoneal macrophages (PMAs). The ApoE-/-Rfx1f/f or ApoE-/-Rfx1f/f Lyz2-Cre mice fed a high-fat diet for 24 weeks were used to further examine the effect of RFX1 on AS pathogenesis. We then performed dual luciferase reporter assays to study the regulation of RFX1 for CD36 transcription. RESULTS: Our results demonstrate that RFX1 expression was significantly reduced in ox-LDL induced foam cells and negatively correlated with lipid uptake in macrophages. Besides, Rfx1 deficiency in myeloid cells aggravated atherosclerotic lesions in ApoE-/- mice. Mechanistically, RFX1 inhibited CD36 expression by directly regulating CD36 transcription in macrophages. CONCLUSIONS: The reduction of RFX1 expression in macrophages is a vital determinant for foam cell formation and the initiation of AS, proving a potential novel approach for the treatment of AS disease.


Subject(s)
Atherosclerosis , CD36 Antigens , Foam Cells , Animals , Humans , Mice , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Foam Cells/cytology , Foam Cells/metabolism , Lipoproteins, LDL/metabolism , Regulatory Factor X1/metabolism , CD36 Antigens/metabolism
16.
J Biol Chem ; 287(27): 22730-9, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22582395

ABSTRACT

Regulatory factor X (RFX) proteins are transcription factors. Seven mammalian RFX proteins have been identified. RFX1 is the prototype RFX. However, its biological functions are not known. Here, RFX1 overexpression reduced fetal bovine serum-stimulated proliferation of SH-SY5Y cells, a human neuroblastoma cell line. This inhibition is associated with decreased transforming growth factor ß2 (TGFß2) and phospho-extracellular signal-regulated kinase (ERK). Exogenous TGFß2 increased cell proliferation and phospho-ERK in cells overexpressing RFX1. An anti-TGFß2 antibody and PD98059, an ERK activation inhibitor, inhibited SH-SY5Y cell proliferation. TGFß2 promoter activity was decreased in cells overexpressing RFX1. Chromosome immunoprecipitation assay showed that RFX1 bound the TGFß2 promoter. RFX1 down-regulation increased TGFß2 in SH-SY5Y and HCN-1A cells, a normal human neuronal cell line. More importantly, TGFß2 concentrations were negatively correlated with RFX1 levels in human medulloblastoma tissues with a R(2) of 0.464. These results suggest that RFX1 reduces cell proliferation through inhibiting the TGFß2-ERK signaling pathway. RFX1 blocks TGFß2 expression through its direct action on TGFß2 transcription. This effect also appears in human brain tumor tissues. Because TGFß is known to be involved in cancer development, our results provide initial evidence to suggest that RFX1 may play an important role in human tumor biology.


Subject(s)
Cerebellar Neoplasms/physiopathology , DNA-Binding Proteins/physiology , Medulloblastoma/physiopathology , Neurons/cytology , Neurons/physiology , Transcription Factors/physiology , Transforming Growth Factor beta2/genetics , Cell Division/physiology , Cell Line, Tumor , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic/physiology , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , Neuroblastoma , RNA, Small Interfering/genetics , Regulatory Factor X Transcription Factors , Regulatory Factor X1 , Transcription, Genetic/physiology
17.
Biochim Biophys Acta Mol Cell Res ; 1870(7): 119510, 2023 10.
Article in English | MEDLINE | ID: mdl-37301270

ABSTRACT

Aberrant expression of multidrug resistance (MDR) proteins is one of the features of cancer stem cells (CSCs) that make them escape chemotherapy. A well-orchestrated regulation of multiple MDRs by different transcription factors in cancer cells confers this drug resistance. An in silico analysis of the major MDR genes revealed a possible regulation by RFX1 and Nrf2. Previous reports also noted that Nrf2 is a positive regulator of MDR genes in NT2 cells. But we, for the first time, report that Regulatory factor X1 (RFX1), a pleiotropic transcription factor, negatively regulates the major MDR genes, Abcg2, Abcb1, Abcc1, and Abcc2, in NT2 cells. The levels of RFX1 in undifferentiated NT2 cells were found to be very low, which significantly increased upon RA-induced differentiation. Ectopic expression of RFX1 reduced the levels of transcripts corresponding to MDRs and stemness-associated genes. Interestingly, Bexarotene, an RXR agonist that acts as an inhibitor of Nrf2-ARE signaling, could increase the transcription of RFX1. Further analysis revealed that the RFX1 promoter has binding sites for RXRα, and upon Bexarotene exposure RXRα could bind and activate the RFX1 promoter. Bexarotene, alone or in combination with Cisplatin, could inhibit many cancer/CSC-associated properties in NT2 cells. Also, it significantly reduced the expression of drug resistance proteins and made the cells sensitive towards Cisplatin. Our study proves that RFX1 could be a potent molecule to target MDRs, and Bexarotene can induce RXRα-mediated RFX1 expression, therefore, would be a better chemo-assisting drug during therapy.


Subject(s)
Carcinoma , Drug Resistance, Neoplasm , Regulatory Factor X1 , Humans , Bexarotene/pharmacology , Cisplatin/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , NF-E2-Related Factor 2/genetics , Regulatory Factor X Transcription Factors , Regulatory Factor X1/drug effects , Regulatory Factor X1/metabolism , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects
18.
JCI Insight ; 8(20)2023 Oct 23.
Article in English | MEDLINE | ID: mdl-37733446

ABSTRACT

Abnormal macrophage polarization is generally present in autoimmune diseases. Overwhelming M1 macrophage activation promotes the continuous progression of inflammation, which is one of the reasons for the development of autoimmune diseases. However, the underlying mechanism is still unclear. Here we explore the function of Regulatory factor X1 (RFX1) in macrophage polarization by constructing colitis and lupus-like mouse models. Both in vivo and in vitro experiments confirmed that RFX1 can promote M1 and inhibit M2 macrophage polarization. Furthermore, we found that RFX1 promoted DNA demethylation of macrophage polarization-related genes by increasing APOBEC3A/Apobec3 expression. We identified a potential RFX1 inhibitor, adenosine diphosphate (ADP), providing a potential strategy for treating autoimmune diseases.


Subject(s)
Autoimmune Diseases , Macrophage Activation , Animals , Mice , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , DNA Demethylation , Inflammation/metabolism , Macrophages/metabolism , Regulatory Factor X1/metabolism
19.
Neuron ; 111(4): 481-492.e8, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36577402

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is a paradigmatic neurodegenerative disease in that it is caused by a mutation in a broadly expressed protein, ATXN1; however, only select populations of cells degenerate. The interaction of polyglutamine-expanded ATXN1 with the transcriptional repressor CIC drives cerebellar Purkinje cell pathogenesis; however, the importance of this interaction in other vulnerable cells remains unknown. Here, we mutated the 154Q knockin allele of Atxn1154Q/2Q mice to prevent the ATXN1-CIC interaction globally. This normalized genome-wide CIC binding; however, it only partially corrected transcriptional and behavioral phenotypes, suggesting the involvement of additional factors in disease pathogenesis. Using unbiased proteomics, we identified three ATXN1-interacting transcription factors: RFX1, ZBTB5, and ZKSCAN1. We observed altered expression of RFX1 and ZKSCAN1 target genes in SCA1 mice and patient-derived iNeurons, highlighting their potential contributions to disease. Together, these data underscore the complexity of mechanisms driving cellular vulnerability in SCA1.


Subject(s)
Spinocerebellar Ataxias , Mice , Animals , Ataxin-1/genetics , Spinocerebellar Ataxias/metabolism , Purkinje Cells/metabolism , Alleles , Mutation/genetics , Cerebellum/metabolism , Regulatory Factor X1/genetics , Regulatory Factor X1/metabolism
20.
Int J Biol Sci ; 19(15): 4744-4762, 2023.
Article in English | MEDLINE | ID: mdl-37781522

ABSTRACT

Background: Hypoxia plays an important role in the lung metastasis of hepatocellular carcinoma (HCC). However, the process by which hypoxia promotes the formation of a pre-metastatic niche (PMN) and its underlying mechanism remain unclear. Methods: Exosomes derived from normoxic and hypoxic HCC cells were collected to induce fibroblast activation in vitro and PMN formation in vivo. The micro RNA (miR) profiles of the exosomes were sequenced to identify differentially expressed miRNAs. Gain- and loss-of-function analyses were performed to investigate miR-4508 function. Dual-luciferase, western blotting, and real-time reverse transcription-PCR analyses were used to identify the direct targets of miR-4508 and its downstream signaling pathways. To demonstrate the roles of hypoxic tumor-derived exosomes (H-TDEs) and miR-4508 in the lung metastasis of liver cancer, H22 tumor cells were injected through the tail vein of mice. Blood plasma-derived exosomes from patients with HCC who underwent transarterial chemoembolization (TACE) were applied to determine clinical correlations. Results: We demonstrated that H-TDEs activated lung fibroblasts and facilitated PMN formation, thereby promoting lung metastasis in mice. Screening for upregulated exosomal miRNAs revealed that miR-4508 and its target, regulatory factor X1 (RFX1), were involved in H-TDE-induced lung PMN formation. Moreover, miR-4508 was significantly upregulated in plasma exosomes derived from patients with HCC after TACE. We confirmed that the p38 MAPK-NF-κB signaling pathway is involved in RFX1 knockdown-induced fibroblast activation and PMN formation. In addition, IL17A, a downstream target of RFX1, was identified as a link between RFX1 knockdown and p38 MAPK activation in fibroblasts. Conclusion: Hypoxia enhances the release of TDEs enriched with miR-4508, thereby promoting lung PMN formation by targeting the RFX1-IL17A-p38 MAPK-NF-κB pathway. These findings highlight a novel mechanism underlying hypoxia-induced pulmonary metastasis of HCC.


Subject(s)
Carcinoma, Hepatocellular , Chemoembolization, Therapeutic , Exosomes , Liver Neoplasms , Lung Neoplasms , MicroRNAs , Humans , Animals , Mice , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , NF-kappa B/metabolism , Regulatory Factor X1/genetics , Exosomes/genetics , Exosomes/metabolism , Cell Line, Tumor , MicroRNAs/genetics , MicroRNAs/metabolism , Lung Neoplasms/metabolism , Hypoxia/metabolism , Lung/metabolism , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Interleukin-17/genetics
SELECTION OF CITATIONS
SEARCH DETAIL