Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
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
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.
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
4.
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
5.
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
6.
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
7.
Cell Res ; 32(9): 801-813, 2022 09.
Article in English | MEDLINE | ID: mdl-35428874

ABSTRACT

Chromatin remodeling is essential for epigenome reprogramming after fertilization. However, the underlying mechanisms of chromatin remodeling remain to be explored. Here, we investigated the dynamic changes in nucleosome occupancy and positioning in pronucleus-stage zygotes using ultra low-input MNase-seq. We observed distinct features of inheritance and reconstruction of nucleosome positioning in both paternal and maternal genomes. Genome-wide de novo nucleosome occupancy in the paternal genome was observed as early as 1 h after the injection of sperm into ooplasm. The nucleosome positioning pattern was continually rebuilt to form nucleosome-depleted regions (NDRs) at promoters and transcription factor (TF) binding sites with differential dynamics in paternal and maternal genomes. NDRs formed more quickly on the promoters of genes involved in zygotic genome activation (ZGA), and this formation is closely linked to histone acetylation, but not transcription elongation or DNA replication. Importantly, we found that NDR establishment on the binding motifs of specific TFs might be associated with their potential pioneer functions in ZGA. Further investigations suggested that the predicted factors MLX and RFX1 played important roles in regulating minor and major ZGA, respectively. Our data not only elucidate the nucleosome positioning dynamics in both male and female pronuclei following fertilization, but also provide an efficient method for identifying key transcription regulators during development.


Subject(s)
Nucleosomes , Zygote , Animals , Chromatin Assembly and Disassembly , Female , Fertilization/genetics , Male , Mice , Nucleosomes/metabolism , Regulatory Factor X1/genetics , Regulatory Factor X1/metabolism , Semen/metabolism , Zygote/metabolism
8.
Cell Rep ; 39(2): 110661, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35417689

ABSTRACT

Cilia are important for the interaction with environments and the proper function of tissues. While the basic structure of cilia is well conserved, ciliated cells have various functions. To understand the distinctive identities of ciliated cells, the identification of cell-specific proteins and its regulation is essential. Here, we report the mechanism that confers a specific identity on IL2 neurons in Caenorhabditis elegans, neurons important for the dauer larva-specific nictation behavior. We show that DAF-19M, an isoform of the sole C. elegans RFX transcription factor DAF-19, heads a regulatory subroutine, regulating target genes through an X-box motif variant under the control of terminal selector proteins UNC-86 and CFI-1 in IL2 neurons. Considering the conservation of DAF-19M module in IL2 neurons for nictation and in male-specific neurons for mating behavior, we propose the existence of an evolutionarily adaptable, hard-wired genetic module for distinct behaviors that share the feature "recognizing the environment."


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Regulatory Factor X1 , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Interleukin-2/metabolism , Male , Regulatory Factor X1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Cell Mol Immunol ; 19(5): 619-633, 2022 05.
Article in English | MEDLINE | ID: mdl-35301470

ABSTRACT

Neutrophils are derived from bone marrow hematopoietic stem cells (HSCs) and are the largest population among circulating white blood cells in humans, acting as the first line of defense against invading pathogens. Whether neutrophils can be generated by transdifferentiation strategies is unknown. Here, we show that thymidine induces the conversion of mouse fibroblasts to neutrophils. Induced neutrophils (iNeus) showed antibacterial effects and did not undergo malignant transformation in vivo. Importantly, iNeu transplantation cured neutropenia in mice in vivo. Mechanistically, thymidine mediates iNeu conversion by enhancing Tet3 activity. Tet3 initiates the expression of the neutrophil fate decision factors Cebpδ and Rfx1 that drive the transdifferentiation of mouse fibroblasts to neutrophils. Therefore, the induction of functional neutrophils by chemicals may provide a potential therapeutic strategy for patients with neutropenia patients and infectious diseases.Fibroblasts; Neutrophils; Thymidine; Transdifferentiation; Tet3.


Subject(s)
Dioxygenases , Neutropenia , Animals , Dioxygenases/metabolism , Fibroblasts/metabolism , Humans , Mice , Neutropenia/metabolism , Neutropenia/pathology , Neutrophils/metabolism , Regulatory Factor X1/metabolism , Thymidine/metabolism
10.
Int J Biol Sci ; 17(15): 4093-4107, 2021.
Article in English | MEDLINE | ID: mdl-34803485

ABSTRACT

Diabetic nephropathy (DN) has become the common and principal microvascular complication of diabetes that could lead to end-stage renal disease. It was reported endothelial-to-mesenchymal transition (EndMT) in glomeruli plays an important role in DN. Enolase1 (ENO1) and Lysine Methyltransferase 5A (KMT5A) were found to modulate epithelial-to-mesenchymal transition in some situations. In the present study, we speculated KMT5A regulates ENO1 transcript, thus participating in hyperglycemia-induced EndMT in glomeruli of DN. Our study represented vimentin, αSMA and ENO1 expression elevated, and CD31 expression decreased in glomeruli of DN participants and rats. In vitro, high glucose induced EndMT by increase of ENO1 levels. Moreover, high glucose downregulated KMT5A levels and increased regulatory factor X1 (RFX1) levels. KMT5A upregulation or si-RFX1 decreased high glucose-induced ENO1 expression and EndMT. RFX1 overexpression- or sh-KMT5A-induced EndMT was attenuated by si-ENO1. Further, the association between KMT5A and RFX1 was verified. Furthermore, histone H4 lysine20 methylation (the direct target of KMT5A) and RFX1 positioned on ENO1 promoter region. sh-KMT5A enhanced positive action of RFX1 on ENO1 promoter activity. KMT5A reduction and RFX1 upregulation were verified in glomeruli of DN patients and rats. KMT5A associated with RFX1 to modulate ENO1, thus involved in hyperglycemia-mediated EndMT in glomeruli of DN.


Subject(s)
Diabetic Nephropathies/metabolism , Gene Expression Regulation/drug effects , Glucose/toxicity , Histone-Lysine N-Methyltransferase/metabolism , Adult , Animals , Biomarkers, Tumor , Blood Glucose , DNA-Binding Proteins , Epithelial-Mesenchymal Transition , Female , Histone-Lysine N-Methyltransferase/genetics , Human Umbilical Vein Endothelial Cells , Humans , Hyperglycemia/metabolism , Kidney Glomerulus , Male , Middle Aged , Phosphopyruvate Hydratase , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA, Messenger/genetics , Rats , Regulatory Factor X1/genetics , Regulatory Factor X1/metabolism , Tumor Suppressor Proteins , Up-Regulation
11.
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
12.
Nat Commun ; 12(1): 1789, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33741976

ABSTRACT

Sensory perception and metabolic homeostasis are known to deteriorate with ageing, impairing the health of aged animals, while mechanisms underlying their deterioration remain poorly understood. The potential interplay between the declining sensory perception and the impaired metabolism during ageing is also barely explored. Here, we report that the intraflagellar transport (IFT) in the cilia of sensory neurons is impaired in the aged nematode Caenorhabditis elegans due to a daf-19/RFX-modulated decrease of IFT components. We find that the reduced IFT in sensory cilia thus impairs sensory perception with ageing. Moreover, we demonstrate that whereas the IFT-dependent decrease of sensory perception in aged worms has a mild impact on the insulin/IGF-1 signalling, it remarkably suppresses AMP-activated protein kinase (AMPK) signalling across tissues. We show that upregulating daf-19/RFX effectively enhances IFT, sensory perception, AMPK activity and autophagy, promoting metabolic homeostasis and longevity. Our study determines an ageing pathway causing IFT decay and sensory perception deterioration, which in turn disrupts metabolism and healthy ageing.


Subject(s)
Aging , Caenorhabditis elegans/metabolism , Cilia/metabolism , Flagella/metabolism , Sensory Receptor Cells/physiology , Signal Transduction/physiology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Animals, Genetically Modified , Biological Transport , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Longevity/genetics , Perception/physiology , RNA Interference , Regulatory Factor X1/genetics , Regulatory Factor X1/metabolism , Sensory Receptor Cells/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
13.
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
14.
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
15.
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
16.
Eur J Surg Oncol ; 44(7): 1087-1093, 2018 07.
Article in English | MEDLINE | ID: mdl-29764705

ABSTRACT

OBJECTIVE: Regulatory factor X1 (RFX1) deletion has been reported to be correlated with poor prognosis of some types of cancer. The present study aimed to investigate the prognostic value of RFX1 in HCC, especially in small hepatocellular carcinoma. METHODS: Immunohistochemical assay was used to investigate RFX1 expression in 221 HCC tissues and another validation cohort of 71 small HCC samples. We also performed in vitro experiments to investigate if RFX1 regulated invasive capacity of HCC cells and expression of epithelial-mesenchymal transition (EMT) markers. RESULTS: We found that RFX1 expression was significantly lower in HCC tissues compared to the corresponding non-tumor tissues. Further survival analysis suggested that the downregulation of RFX1 correlated with poor prognosis and a high recurrence risk in HCC patients, particularly in small HCC patients. Furthermore, another validation cohort of small HCC samples confirmed that downregulation of RFX1 in HCC tissues predicted high recurrence risk and poor prognosis for early stage HCC patients. In vitro studies suggested that knocking down RFX1 facilitated HCC cell invasion, while overexpression of RFX1 reduced the invasion of HCC cells. Western blot assays also indicated that RFX1 regulated expression of some EMT markers. Knocking down RFX1 decreased E-cadherin and increased vimentin expression, while RFX1 overexpression enhanced E-cadherin and decreased vimentin expression. CONCLUSIONS: Our study demonstrated that RFX1 downregulation is a new predictive marker of high recurrence risk and poor prognosis of HCC; It has potential to help guide treatment for postoperative HCC patients, especially for small HCC patients.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Regulatory Factor X1/metabolism , Antigens, CD , Blotting, Western , Cadherins/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cohort Studies , Down-Regulation , Epithelial-Mesenchymal Transition , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , In Vitro Techniques , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , Proportional Hazards Models , Survival Analysis , Tumor Burden , Vimentin/metabolism
17.
Cell Death Dis ; 9(4): 437, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29740017

ABSTRACT

Degeneration or loss of inner ear hair cells (HCs) is irreversible and results in sensorineural hearing loss (SHL). Human-induced pluripotent stem cells (hiPSCs) have been employed in disease modelling and cell therapy. Here, we propose a transcription factor (TF)-driven approach using ATOH1 and regulatory factor of x-box (RFX) genes to generate HC-like cells from hiPSCs. Our results suggest that ATOH1/RFX1/RFX3 could significantly increase the differentiation capacity of iPSCs into MYO7AmCherry-positive cells, upregulate the mRNA expression levels of HC-related genes and promote the differentiation of HCs with more mature stereociliary bundles. To model the molecular and stereociliary structural changes involved in HC dysfunction in SHL, we further used ATOH1/RFX1/RFX3 to differentiate HC-like cells from the iPSCs from patients with myoclonus epilepsy associated with ragged-red fibres (MERRF) syndrome, which is caused by A8344G mutation of mitochondrial DNA (mtDNA), and characterised by myoclonus epilepsy, ataxia and SHL. Compared with isogenic iPSCs, MERRF-iPSCs possessed ~42-44% mtDNA with A8344G mutation and exhibited significantly elevated reactive oxygen species (ROS) production and CAT gene expression. Furthermore, MERRF-iPSC-differentiated HC-like cells exhibited significantly elevated ROS levels and MnSOD and CAT gene expression. These MERRF-HCs that had more single cilia with a shorter length could be observed only by using a non-TF method, but those with fewer stereociliary bundle-like protrusions than isogenic iPSCs-differentiated-HC-like cells could be further observed using ATOH1/RFX1/RFX3 TFs. We further analysed and compared the whole transcriptome of M1ctrl-HCs and M1-HCs after treatment with ATOH1 or ATOH1/RFX1/RFX3. We revealed that the HC-related gene transcripts in M1ctrl-iPSCs had a significantly higher tendency to be activated by ATOH1/RFX1/RFX3 than M1-iPSCs. The ATOH1/RFX1/RFX3 TF-driven approach for the differentiation of HC-like cells from iPSCs is an efficient and promising strategy for the disease modelling of SHL and can be employed in future therapeutic strategies to treat SHL patients.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Hair Cells, Auditory, Inner/metabolism , MERRF Syndrome/pathology , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X1/genetics , Adolescent , Basic Helix-Loop-Helix Transcription Factors/metabolism , Catalase/genetics , Catalase/metabolism , Cilia/physiology , DNA, Mitochondrial/genetics , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Female , Hair Cells, Auditory, Inner/cytology , Humans , Induced Pluripotent Stem Cells/cytology , MERRF Syndrome/complications , Myosin VIIa/genetics , Myosin VIIa/metabolism , Point Mutation , Reactive Oxygen Species/metabolism , Regulatory Factor X Transcription Factors/metabolism , Regulatory Factor X1/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
18.
Cancer Med ; 7(5): 2021-2033, 2018 05.
Article in English | MEDLINE | ID: mdl-29601674

ABSTRACT

Cytotoxic chemotherapy drugs, including doxorubicin, can directly promote hepatitis B virus (HBV) replication, but the mechanism has not been fully clarified. This study investigated the potential mechanism underlying the cytotoxic chemotherapy-mediated direct promotion of HBV replication. We found that HBV replication and regulatory factor X box 1 gene (RFX1) expression were simultaneously promoted by doxorubicin treatment. The amount of RFX1 bound to the HBV enhancer I was significantly increased under doxorubicin treatment. Furthermore, the activity of doxorubicin in promoting HBV replication was significantly attenuated when the expression of endogenous RFX1 was knocked down, and the EP element of HBV enhancer I, an element that mediated the binding of RFX1 and HBV enhancer I, was mutated. In addition, two different sequences of the conserved EP element were found among HBV genotypes A-D, and doxorubicin could promote the replication of HBV harboring either of the conserved EP elements. Here, a novel pathway in which doxorubicin promoted HBV replication via RFX1 was identified, and it might participate in doxorubicin-induced HBV reactivation. These findings would be helpful in preventing HBV reactivation during anticancer chemotherapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Hepatitis B virus/growth & development , Regulatory Factor X1/metabolism , Virus Activation/drug effects , Cell Line, Tumor , Hep G2 Cells , Hepatitis B Surface Antigens/analysis , Hepatitis B e Antigens/analysis , Humans , Regulatory Factor X1/biosynthesis
19.
Nat Commun ; 9(1): 583, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29422534

ABSTRACT

Epigenetic modifications affect the differentiation of T cell subsets and the pathogenesis of autoimmune diseases, but many mechanisms of epigenetic regulation of T cell differentiation are unclear. Here we show reduced expression of the transcription factor RFX1 in CD4+ T cells from patients with systemic lupus erythematosus, which leads to IL-17A overexpression through increased histone H3 acetylation and decreased DNA methylation and H3K9 tri-methylation. Conditional deletion of Rfx1 in mice exacerbates experimental autoimmune encephalomyelitis and pristane-induced lupus-like syndrome and increases induction of Th17 cells. In vitro, Rfx1 deficiency increases the differentiation of naive CD4+ T cells into Th17 cells, but this effect can be reversed by forced expression of Rfx1. Importantly, RFX1 functions downstream of STAT3 and phosphorylated STAT3 can inhibit RFX1 expression, highlighting a non-canonical pathway that regulates differentiation of Th17 cells. Collectively, our findings identify a unique role for RFX1 in Th17-related autoimmune diseases.


Subject(s)
Epigenesis, Genetic , Interleukin-6/pharmacology , Lupus Erythematosus, Systemic/immunology , Regulatory Factor X1/metabolism , STAT3 Transcription Factor/metabolism , Th17 Cells/drug effects , Adolescent , Adult , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , HEK293 Cells , Humans , Jurkat Cells , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Phosphorylation , Regulatory Factor X1/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , Th17 Cells/immunology , Th17 Cells/metabolism , Young Adult
20.
Sci Rep ; 8(1): 210, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29317724

ABSTRACT

Adeno-associated virus (AAV) transduction efficiency depends on the way in which cellular proteins process viral genomes in the nucleus. In this study, we have investigated the binding of nuclear proteins to the double stranded D (dsD) sequence of the AAV inverted terminal repeat (ITRs) by electromobility shift assay. We present here several lines of evidence that transcription factors belonging to the RFX protein family bind specifically and selectively to AAV2 and AAV1 dsD sequences. Using supershift experiments, we characterize complexes containing RFX1 homodimers and RFX1/RFX3 heterodimers. Following transduction of HEK-293 cells, the AAV genome can be pulled-down by RFX1 and RFX3 antibodies. Moreover, our data suggest that RFX proteins which interact with transcriptional enhancers of several mammalian DNA viruses, can act as regulators of AAV mediated transgene expression.


Subject(s)
Dependovirus/genetics , Regulatory Factor X Transcription Factors/metabolism , Regulatory Factor X1/metabolism , Transduction, Genetic , Dependovirus/metabolism , HEK293 Cells , Humans , Protein Binding , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X1/genetics , Terminal Repeat Sequences
SELECTION OF CITATIONS
SEARCH DETAIL
...