Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Nat Immunol ; 22(9): 1093-1106, 2021 09.
Article in English | MEDLINE | ID: mdl-34282331

ABSTRACT

Neutrophils display distinct gene expression patters depending on their developmental stage, activation state and tissue microenvironment. To determine the transcription factor networks that shape these responses in a mouse model, we integrated transcriptional and chromatin analyses of neutrophils during acute inflammation. We showed active chromatin remodeling at two transition stages: bone marrow-to-blood and blood-to-tissue. Analysis of differentially accessible regions revealed distinct sets of putative transcription factors associated with control of neutrophil inflammatory responses. Using ex vivo and in vivo approaches, we confirmed that RUNX1 and KLF6 modulate neutrophil maturation, whereas RELB, IRF5 and JUNB drive neutrophil effector responses and RFX2 and RELB promote survival. Interfering with neutrophil activation by targeting one of these factors, JUNB, reduced pathological inflammation in a mouse model of myocardial infarction. Therefore, our study represents a blueprint for transcriptional control of neutrophil responses in acute inflammation and opens possibilities for stage-specific therapeutic modulation of neutrophil function in disease.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Inflammation/immunology , Neutrophils/immunology , Transcriptional Activation/genetics , Animals , CHO Cells , Cell Line , Core Binding Factor Alpha 2 Subunit/metabolism , Cricetulus , Female , Interferon Regulatory Factors/metabolism , Kruppel-Like Factor 6/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Regulatory Factor X Transcription Factors/metabolism , Transcription Factor RelB/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics
2.
Development ; 151(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38722098

ABSTRACT

During development, the gastrointestinal tract undergoes patterning along its anterior-posterior axis to define regions with distinct organs and functions. A new paper in Development derives human intestinal organoids from an individual with duodenal defects and a compound heterozygous variant in the gene encoding the transcription factor RFX6. By studying these organoids, the authors identify novel roles for RFX6 in intestinal patterning. To learn more about the story behind the paper, we caught up with first author J. Guillermo Sanchez and corresponding author Jim Wells, an endowed professor in the Division of Developmental Biology at Cincinnati Children's Hospital, USA, where he is also the Director for Basic Research in the Division of Endocrinology.


Subject(s)
Developmental Biology , Humans , History, 21st Century , History, 20th Century , Developmental Biology/history , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/metabolism , Organoids/metabolism , Body Patterning/genetics
3.
Development ; 151(9)2024 05 01.
Article in English | MEDLINE | ID: mdl-38587174

ABSTRACT

The gastrointestinal (GI) tract is complex and consists of multiple organs with unique functions. Rare gene variants can cause congenital malformations of the human GI tract, although the molecular basis of these has been poorly studied. We identified a patient with compound-heterozygous variants in RFX6 presenting with duodenal malrotation and atresia, implicating RFX6 in development of the proximal intestine. To identify how mutations in RFX6 impact intestinal patterning and function, we derived induced pluripotent stem cells from this patient to generate human intestinal organoids (HIOs). We identified that the duodenal HIOs and human tissues had mixed regional identity, with gastric and ileal features. CRISPR-mediated correction of RFX6 restored duodenal identity. We then used gain- and loss-of-function and transcriptomic approaches in HIOs and Xenopus embryos to identify that PDX1 is a downstream transcriptional target of RFX6 required for duodenal development. However, RFX6 had additional PDX1-independent transcriptional targets involving multiple components of signaling pathways that are required for establishing early regional identity in the GI tract. In summary, we have identified RFX6 as a key regulator in intestinal patterning that acts by regulating transcriptional and signaling pathways.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins , Organoids , Regulatory Factor X Transcription Factors , Trans-Activators , Humans , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/metabolism , Animals , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Organoids/metabolism , Organoids/embryology , Duodenum/metabolism , Duodenum/embryology , Intestines/embryology , Intestinal Atresia/genetics , Induced Pluripotent Stem Cells/metabolism , Body Patterning/genetics , Signal Transduction/genetics , Mutation/genetics
4.
PLoS Biol ; 20(2): e3001538, 2022 02.
Article in English | MEDLINE | ID: mdl-35143476

ABSTRACT

Within the immune system, microRNAs (miRNAs) exert key regulatory functions. However, what are the mRNA targets regulated by miRNAs and how miRNAs are transcriptionally regulated themselves remain for the most part unknown. We found that in primary human memory T helper lymphocytes, miR-150 was the most abundantly expressed miRNA, and its expression decreased drastically upon activation, suggesting regulatory roles. Constitutive MIR150 gene expression required the RFX family of transcription factors, and its activation-induced down-regulation was linked to their reduced expression. By performing miRNA pull-down and sequencing experiments, we identified PDGFA-associated protein 1 (PDAP1) as one main target of miR-150 in human T lymphocytes. PDAP1 acted as an RNA-binding protein (RBP), and its CRISPR/Cas-9-mediated deletion revealed that it prominently contributed to the regulation of T-cell proliferation. Overall, using an integrated approach involving quantitative analysis, unbiased genomics, and genome editing, we identified RFX factors, miR-150, and the PDAP1 RBP as the components of a regulatory axis that restrains proliferation of primary human T lymphocytes.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation/genetics , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , Regulatory Factor X Transcription Factors/genetics , 3' Untranslated Regions/genetics , Blotting, Western , CD4-Positive T-Lymphocytes/cytology , Cells, Cultured , Chromatin Immunoprecipitation Sequencing/methods , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Lymphocyte Activation/genetics , Regulatory Factor X Transcription Factors/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
5.
Diabetologia ; 67(8): 1642-1662, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38743124

ABSTRACT

AIMS/HYPOTHESIS: Regulatory factor X 6 (RFX6) is crucial for pancreatic endocrine development and differentiation. The RFX6 variant p.His293LeufsTer7 is significantly enriched in the Finnish population, with almost 1:250 individuals as a carrier. Importantly, the FinnGen study indicates a high predisposition for heterozygous carriers to develop type 2 and gestational diabetes. However, the precise mechanism of this predisposition remains unknown. METHODS: To understand the role of this variant in beta cell development and function, we used CRISPR technology to generate allelic series of pluripotent stem cells. We created two isogenic stem cell models: a human embryonic stem cell model; and a patient-derived stem cell model. Both were differentiated into pancreatic islet lineages (stem-cell-derived islets, SC-islets), followed by implantation in immunocompromised NOD-SCID-Gamma mice. RESULTS: Stem cell models of the homozygous variant RFX6-/- predictably failed to generate insulin-secreting pancreatic beta cells, mirroring the phenotype observed in Mitchell-Riley syndrome. Notably, at the pancreatic endocrine stage, there was an upregulation of precursor markers NEUROG3 and SOX9, accompanied by increased apoptosis. Intriguingly, heterozygous RFX6+/- SC-islets exhibited RFX6 haploinsufficiency (54.2% reduction in protein expression), associated with reduced beta cell maturation markers, altered calcium signalling and impaired insulin secretion (62% and 54% reduction in basal and high glucose conditions, respectively). However, RFX6 haploinsufficiency did not have an impact on beta cell number or insulin content. The reduced insulin secretion persisted after in vivo implantation in mice, aligning with the increased risk of variant carriers to develop diabetes. CONCLUSIONS/INTERPRETATION: Our allelic series isogenic SC-islet models represent a powerful tool to elucidate specific aetiologies of diabetes in humans, enabling the sensitive detection of aberrations in both beta cell development and function. We highlight the critical role of RFX6 in augmenting and maintaining the pancreatic progenitor pool, with an endocrine roadblock and increased cell death upon its loss. We demonstrate that RFX6 haploinsufficiency does not affect beta cell number or insulin content but does impair function, predisposing heterozygous carriers of loss-of-function variants to diabetes. DATA AVAILABILITY: Ultra-deep bulk RNA-seq data for pancreatic differentiation stages 3, 5 and 7 of H1 RFX6 genotypes are deposited in the Gene Expression Omnibus database with accession code GSE234289. Original western blot images are deposited at Mendeley ( https://data.mendeley.com/datasets/g75drr3mgw/2 ).


Subject(s)
Haploinsufficiency , Insulin-Secreting Cells , Regulatory Factor X Transcription Factors , Insulin-Secreting Cells/metabolism , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/metabolism , Animals , Humans , Mice , Cell Differentiation/genetics , Mice, Inbred NOD , Mice, SCID , Genetic Predisposition to Disease , Female , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Nerve Tissue Proteins
6.
J Neurooncol ; 165(3): 487-497, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38057505

ABSTRACT

PURPOSE: Spinal ependymoma (SE) is a rare tumor that is most commonly low-grade and tends to recur when complete tumor resection is not feasible. We investigated the molecular mechanism induces stem cell features in SE. METHODS: Immunohistochemical staining was conducted to analyze the expression of RFX2 in tumor tissues of SE patients at different stages. The expression of tumor stemness markers (Netsin and CD133) was analyzed using western blot analysis and IF, and the efficiency of sphere formation in SE cells was analyzed. The biological activities of SE cells were analyzed by EdU proliferation assay, TUNEL, wound healing, and Transwell assays. The regulatory relationship of RFX2 on PAF1 was verified by ChIP-qPCR and the dual-luciferase assay. SE cells were injected into the spinal cord of nude mice for in vivo assays. RESULTS: RFX2 was higher in the tumor tissues of SE-III patients than in the tumor tissues of SE-I patients. RFX2 knockdown reduced the expression of tumor stemness markers in SE cells and inhibited the sphere formation efficiency. Moreover, RFX2 knockdown ameliorated the malignant progression of SE in nude mice, as manifested by prolonged survival and alleviated SE tumor infiltration. RFX2 bound to the PAF1 promoter to induce its transcription. Overexpression of PAF1 overturned the effects of RFX2 knockdown on stem cell features and biological activities of SE cells, thereby reducing survival in mice. CONCLUSIONS: RFX2 activates PAF1 transcription, which promotes tumor stemness of SE cells and leads to the malignant progression of SE.


Subject(s)
Ependymoma , Epigenesis, Genetic , Humans , Animals , Mice , Mice, Nude , Cell Line, Tumor , Neoplasm Recurrence, Local/pathology , Ependymoma/pathology , Cell Proliferation , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/pathology , Regulatory Factor X Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Nucleic Acids Res ; 49(13): 7437-7456, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34197623

ABSTRACT

Despite its prominence, the mechanisms through which the tumor suppressor p53 regulates most genes remain unclear. Recently, the regulatory factor X 7 (RFX7) emerged as a suppressor of lymphoid neoplasms, but its regulation and target genes mediating tumor suppression remain unknown. Here, we identify a novel p53-RFX7 signaling axis. Integrative analysis of the RFX7 DNA binding landscape and the RFX7-regulated transcriptome in three distinct cell systems reveals that RFX7 directly controls multiple established tumor suppressors, including PDCD4, PIK3IP1, MXD4, and PNRC1, across cell types and is the missing link for their activation in response to p53 and stress. RFX7 target gene expression correlates with cell differentiation and better prognosis in numerous cancer types. Interestingly, we find that RFX7 sensitizes cells to Doxorubicin by promoting apoptosis. Together, our work establishes RFX7's role as a ubiquitous regulator of cell growth and fate determination and a key node in the p53 transcriptional program.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , Genes, Tumor Suppressor , Regulatory Factor X Transcription Factors/metabolism , Stress, Physiological/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Cell Differentiation/genetics , Cell Line, Tumor , DNA/metabolism , Doxorubicin/pharmacology , Humans , Mice , Neoplasms/genetics , Neoplasms/mortality , Prognosis , Promoter Regions, Genetic , Regulatory Factor X Transcription Factors/physiology , Signal Transduction , Trans-Activators/metabolism , Transcriptome
8.
J Biol Chem ; 296: 100291, 2021.
Article in English | MEDLINE | ID: mdl-33453283

ABSTRACT

Androglobin (ADGB) represents the latest addition to the globin superfamily in metazoans. The chimeric protein comprises a calpain domain and a unique circularly permutated globin domain. ADGB expression levels are most abundant in mammalian testis, but its cell-type-specific expression, regulation, and function have remained unexplored. Analyzing bulk and single-cell mRNA-Seq data from mammalian tissues, we found that-in addition to the testes-ADGB is prominently expressed in the female reproductive tract, lungs, and brain, specifically being associated with cell types forming motile cilia. Correlation analysis suggested coregulation of ADGB with FOXJ1, a crucial transcription factor of ciliogenesis. Investigating the transcriptional regulation of the ADGB gene, we characterized its promoter using epigenomic datasets, exogenous promoter-dependent luciferase assays, and CRISPR/dCas9-VPR-mediated activation approaches. Reporter gene assays revealed that FOXJ1 indeed substantially enhanced luciferase activity driven by the ADGB promoter. ChIP assays confirmed binding of FOXJ1 to the endogenous ADGB promoter region. We dissected the minimal sequence required for FOXJ1-dependent regulation and fine mapped the FOXJ1 binding site to two evolutionarily conserved regions within the ADGB promoter. FOXJ1 overexpression significantly increased endogenous ADGB mRNA levels in HEK293 and MCF-7 cells. Similar results were observed upon RFX2 overexpression, another key transcription factor in ciliogenesis. The complex transcriptional regulation of the ADGB locus was illustrated by identifying a distal enhancer, responsible for synergistic regulation by RFX2 and FOXJ1. Finally, cell culture studies indicated an ADGB-dependent increase in the number of ciliated cells upon overexpression of the full-length protein, confirming a ciliogenesis-associated role of ADGB in mammals.


Subject(s)
Calmodulin-Binding Proteins/genetics , Cilia/genetics , Forkhead Transcription Factors/genetics , Globins/genetics , Regulatory Factor X Transcription Factors/genetics , Transcriptome , Animals , Binding Sites , Brain/cytology , Brain/growth & development , Brain/metabolism , Calmodulin-Binding Proteins/metabolism , Cattle , Cilia/metabolism , Enhancer Elements, Genetic , Female , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Gene Ontology , Globins/metabolism , HEK293 Cells , HeLa Cells , Humans , Lung/cytology , Lung/growth & development , Lung/metabolism , MCF-7 Cells , Male , Molecular Sequence Annotation , Ovary/cytology , Ovary/growth & development , Ovary/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Regulatory Factor X Transcription Factors/metabolism , Sequence Analysis, RNA , Testis/cytology , Testis/growth & development , Testis/metabolism
9.
Fish Shellfish Immunol ; 121: 370-379, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35051562

ABSTRACT

Regulatory factor X 5 (RFX 5) is a member of the RFX family, and it forms the transcription factor complex RFX with RFXANK/B and RFXAP. The RFX complex can activate MHC expression by binding to the MHC promoter. However, the regulate mechanism of RFX in fish species is not been fully elucidated. In this study, we investigated the transcriptional regulation of Epinephelus akaara RFX5 (EaRFX5) on EaMHCI, and its effect on immune pathways. The genomic sequence of EaRFX5 was 35,774 bp and consisted of ten exons and nine introns. The length of EaRFX5 ORF sequence is 2,160 bp, encoding 719 amino acids. By qRT-PCR, EaRFX5 was detected constitutively expressed in twelve selected tissues, showing a wide range of expression. EaRFX5 expression parttern in response to poly (I:C), LPS, Zymosan A, SGIV, and NNV challenges showed that EaRFX5 plays a differentiated immunomodulatory role in response to various stimuli in different tissues, and EaRFX5 was most significantly upregulated in the kidney after challenge with SGIV. Subcellular localization assays showed that EaRFX5 is a typical nuclear protein. Based on the in vitro overexpression experiments, EaRFX5 appeared to promote the expression of EaMHCIa gene, interferon signalling pathway and inflammatory cytokine. Luciferase reporter assay showed that the -267 bp to +82 bp region of EaMHCIa promoter was the core region where EaRFX5 modulated. Additionally, point mutations and electrophoretic mobility shift assays indicating M3 is the EaRFX5 binding sites in the EaMHCIa promoter. These results contribute to elucidating the function of EaRFX5 in fish immune response, and provide the first evidence of positive regulation of MHCIa expression by RFX5 in fish.


Subject(s)
Bass , Fish Proteins/metabolism , Regulatory Factor X Transcription Factors/metabolism , Animals , Bass/genetics , Bass/immunology , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genes, MHC Class I
10.
Int J Mol Sci ; 23(5)2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35269888

ABSTRACT

X-box binding protein 1 (XBP1) is a member of the CREB/ATF basic region leucine zipper family transcribed as the unspliced isoform (XBP1-u), which, upon exposure to endoplasmic reticulum stress, is spliced into its spliced isoform (XBP1-s). XBP1-s interacts with the cAMP response element of major histocompatibility complex class II gene and plays critical role in unfolded protein response (UPR) by regulating the transcriptional activity of genes involved in UPR. XBP1-s is also involved in other physiological pathways, including lipid metabolism, insulin metabolism, and differentiation of immune cells. Its aberrant expression is closely related to inflammation, neurodegenerative disease, viral infection, and is crucial for promoting tumor progression and drug resistance. Meanwhile, recent studies reported that the function of XBP1-u has been underestimated, as it is not merely a precursor of XBP1-s. Instead, XBP-1u is a critical factor involved in various biological pathways including autophagy and tumorigenesis through post-translational regulation. Herein, we summarize recent research on the biological functions of both XBP1-u and XBP1-s, as well as their relation to diseases.


Subject(s)
Neurodegenerative Diseases , Endoplasmic Reticulum Stress/genetics , Humans , Protein Isoforms/genetics , Protein Isoforms/metabolism , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/metabolism , Unfolded Protein Response , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
11.
Proc Natl Acad Sci U S A ; 115(36): E8403-E8412, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30127002

ABSTRACT

Defects in cilia have been associated with an expanding human disease spectrum known as ciliopathies. Regulatory Factor X 3 (RFX3) is one of the major transcription factors required for ciliogenesis and cilia functions. In addition, RFX3 regulates pancreatic islet cell differentiation and mature ß-cell functions. However, how RFX3 protein is regulated at the posttranslational level remains poorly understood. Using chemical reporters of protein fatty acylation and mass spectrometry analysis, here we show that RFX3 transcriptional activity is regulated by S-fatty acylation at a highly conserved cysteine residue in the dimerization domain. Surprisingly, RFX3 undergoes enzyme-independent, "self-catalyzed" auto-fatty acylation and displays preferences for 18-carbon stearic acid and oleic acid. The fatty acylation-deficient mutant of RFX3 shows decreased homodimerization; fails to promote ciliary gene expression, ciliogenesis, and elongation; and impairs Hedgehog signaling. Our findings reveal a regulation of RFX3 transcription factor and link fatty acid metabolism and protein lipidation to the regulation of ciliogenesis.


Subject(s)
Lipoylation , Oleic Acid/metabolism , Regulatory Factor X Transcription Factors/metabolism , Stearic Acids/metabolism , Acylation , Animals , Cilia/genetics , Cilia/metabolism , Ciliopathies/genetics , Ciliopathies/metabolism , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Regulatory Factor X Transcription Factors/genetics
12.
Biochem Biophys Res Commun ; 523(1): 263-266, 2020 02 26.
Article in English | MEDLINE | ID: mdl-31864703

ABSTRACT

RFX7 is an important member in the RFX family of DNA binding proteins and plays critical roles in natural killer cell-mediated immunity and neuron development. Our previous work identified ANKRA2 and RFXANK as the potential binding partners of RFX7. Here we present two structures of a RFX7 fragment, with one bound with the ANKRA2 ankyrin domain and the other bound to the RFXANK ankyrin domain. Our structural analysis reveals that both ANKRA2 and RFXANK recognize the PXLPXL motif of RFX7 and its flanking sequences via extensive hydrophobic interactions. Detailed structural comparison also provides an explanation for the different RFX7 binding affinities of ANKRA2 and RFXANK. Thus our work would provide clue to the understanding the roles of ANKRA2 and RFXANK in the RFX7-associated signaling pathway.


Subject(s)
Ankyrins/chemistry , Ankyrins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Regulatory Factor X Transcription Factors/chemistry , Regulatory Factor X Transcription Factors/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation , Regulatory Factor X Transcription Factors/genetics , Signal Transduction
13.
PLoS Genet ; 13(1): e1006538, 2017 01.
Article in English | MEDLINE | ID: mdl-28103240

ABSTRACT

Cooperative transcription factor binding at cis-regulatory sites in the genome drives robust eukaryotic gene expression, and many such sites must be coordinated to produce coherent transcriptional programs. The transcriptional program leading to motile cilia formation requires members of the DNA-binding forkhead (Fox) and Rfx transcription factor families and these factors co-localize to cilia gene promoters, but it is not clear how many cilia genes are regulated by these two factors, whether these factors act directly or indirectly, or how these factors act with specificity in the context of a 3-dimensional genome. Here, we use genome-wide approaches to show that cilia genes reside at the boundaries of topological domains and that these areas have low enhancer density. We show that the transcription factors Foxj1 and Rfx2 binding occurs in the promoters of more cilia genes than other known cilia transcription factors and that while Rfx2 binds directly to promoters and enhancers equally, Foxj1 prefers direct binding to enhancers and is stabilized at promoters by Rfx2. Finally, we show that Rfx2 and Foxj1 lie at the anchor endpoints of chromatin loops, suggesting that target genes are activated when Foxj1 bound at distal sites is recruited via a loop created by Rfx2 binding at both sites. We speculate that the primary function of Rfx2 is to stabilize distal enhancers with proximal promoters by operating as a scaffolding factor, bringing key regulatory domains bound by Foxj1 into close physical proximity and enabling coordinated cilia gene expression.


Subject(s)
Chromatin/metabolism , Forkhead Transcription Factors/metabolism , Regulatory Factor X Transcription Factors/metabolism , Transcriptional Activation , Xenopus Proteins/metabolism , Animals , Chromatin/chemistry , Cilia/metabolism , Promoter Regions, Genetic , Protein Binding , Skin/cytology , Skin/metabolism , Species Specificity , Xenopus
14.
Proc Natl Acad Sci U S A ; 114(9): 2301-2306, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28193859

ABSTRACT

Genome-wide association studies (GWAS) have identified >100 independent SNPs that modulate the risk of type 2 diabetes (T2D) and related traits. However, the pathogenic mechanisms of most of these SNPs remain elusive. Here, we examined genomic, epigenomic, and transcriptomic profiles in human pancreatic islets to understand the links between genetic variation, chromatin landscape, and gene expression in the context of T2D. We first integrated genome and transcriptome variation across 112 islet samples to produce dense cis-expression quantitative trait loci (cis-eQTL) maps. Additional integration with chromatin-state maps for islets and other diverse tissue types revealed that cis-eQTLs for islet-specific genes are specifically and significantly enriched in islet stretch enhancers. High-resolution chromatin accessibility profiling using assay for transposase-accessible chromatin sequencing (ATAC-seq) in two islet samples enabled us to identify specific transcription factor (TF) footprints embedded in active regulatory elements, which are highly enriched for islet cis-eQTL. Aggregate allelic bias signatures in TF footprints enabled us de novo to reconstruct TF binding affinities genetically, which support the high-quality nature of the TF footprint predictions. Interestingly, we found that T2D GWAS loci were strikingly and specifically enriched in islet Regulatory Factor X (RFX) footprints. Remarkably, within and across independent loci, T2D risk alleles that overlap with RFX footprints uniformly disrupt the RFX motifs at high-information content positions. Together, these results suggest that common regulatory variations have shaped islet TF footprints and the transcriptome and that a confluent RFX regulatory grammar plays a significant role in the genetic component of T2D predisposition.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease , Genome, Human , Islets of Langerhans/metabolism , Quantitative Trait Loci , Transcriptome , Alleles , Base Sequence , Binding Sites , Chromatin/chemistry , Chromatin/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Epigenesis, Genetic , Gene Expression Profiling , Genetic Variation , Genome-Wide Association Study , Genomic Imprinting , Humans , Islets of Langerhans/pathology , Polymorphism, Single Nucleotide , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/metabolism
15.
Yi Chuan ; 42(8): 760-774, 2020 Aug 20.
Article in English | MEDLINE | ID: mdl-32952112

ABSTRACT

Gene expression and three-dimensional (3D) genome organization are thought to be closely related. The protocadherin (Pcdh) clusters are central for neuronal self-avoidance in brain development and have been used as model genes to explore the role of 3D genome structures in gene regulation. Transcription factor RFX5 (regulatory factor x 5) is a member of the winged-helix subfamily of the helix-turn-helix superfamily proteins. The RFX5 protein contains four domains: oligomerization, DNA-binding, helical, and transactivation domains. RFX5 plays an important role in regulating expression of the major histocompatibility complex class II (MHC II) gene complex. Here we report a role of RFX5 in the regulation of clustered Pcdh genes. We first knocked out the RFX5 gene by using CRISPR/Cas9 DNA-fragment editing in HEC-1-B cell line. By RNA-seq experiments, we found that RFX5 deletion results in a significant increase of expression levels of the Pcdhα6, Pcdhα12 and Pcdhαc2 genes. By ChIP-nexus experiments, we found that RFX5 deletion leads to the increased binding of CTCF and RAD21 in the Pcdhα cluster. Finally, through quantitative high-resolution chromosome conformation capture copy (QHR-4C) experiments, we found that RFX5 deletion results in a significant increase of long-distance chromatin interactions between the HS5-1 enhancer and its target promoters in the Pcdhα cluster. In conclusion, RFX5 regulates gene expression of the Pcdhα cluster through higher-order chromatin structure.


Subject(s)
Cadherins , Gene Expression Regulation , Regulatory Factor X Transcription Factors , Transcription Factors , Cadherins/genetics , Cell Line, Tumor , Gene Expression Regulation/genetics , Gene Knockout Techniques , Humans , Promoter Regions, Genetic/genetics , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/metabolism , Regulatory Sequences, Nucleic Acid
16.
BMC Genomics ; 20(1): 718, 2019 Sep 18.
Article in English | MEDLINE | ID: mdl-31533632

ABSTRACT

BACKGROUND: The work of the FANTOM5 Consortium has brought forth a new level of understanding of the regulation of gene transcription and the cellular processes involved in creating diversity of cell types. In this study, we extended the analysis of the FANTOM5 Cap Analysis of Gene Expression (CAGE) transcriptome data to focus on understanding the genetic regulators involved in mouse cerebellar development. RESULTS: We used the HeliScopeCAGE library sequencing on cerebellar samples over 8 embryonic and 4 early postnatal times. This study showcases temporal expression pattern changes during cerebellar development. Through a bioinformatics analysis that focused on transcription factors, their promoters and binding sites, we identified genes that appear as strong candidates for involvement in cerebellar development. We selected several candidate transcriptional regulators for validation experiments including qRT-PCR and shRNA transcript knockdown. We observed marked and reproducible developmental defects in Atf4, Rfx3, and Scrt2 knockdown embryos, which support the role of these genes in cerebellar development. CONCLUSIONS: The successful identification of these novel gene regulators in cerebellar development demonstrates that the FANTOM5 cerebellum time series is a high-quality transcriptome database for functional investigation of gene regulatory networks in cerebellar development.


Subject(s)
Cerebellum/growth & development , Gene Expression Profiling , Nucleotide Motifs/genetics , Transcription, Genetic/genetics , Activating Transcription Factor 4/deficiency , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Cerebellum/embryology , Cerebellum/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Regulatory Factor X Transcription Factors/deficiency , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Biochem Biophys Res Commun ; 508(2): 556-562, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30509498

ABSTRACT

Regulatory Factor X-box binding transcriptional factor 6 (Rfx6) plays an important role in the differentiation and development of pancreas in mammals. However, the direct target genes of Rfx6 to regulate this process were largely unknown. The present study aimed to investigate the function of Rfx6 on regulating pancreatic differentiation and development in a physiologically-relevant context. We performed the chromatin immunoprecipitation followed by the next generation sequencing analysis (ChIP-seq) using whole pancreatic tissue harvested from C57/BL6 adult mice to find target genes of Rfx6. We captured 4146 unique peaks in the genome region of the adult murine pancreas. Among all these binding peaks, a majority were located in intron or intergenic regions. We further annotated all peaks to their nearest gene, and over 1000 genes were captured as Rfx6-binding genes in the pancreas. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analysis found that Rfx6-binding genes to be associated with the pancreas developmental process. A portion of selected ChIP-seq targets related with pancreas differentiation including Pdx1, Neurod1, Hnf1a, Nkx6-1, St18 and Shox2 were selected and validated as true targets by independent qPCR experiments. In addition, Rfx6 can directly bind to upstream of MiR-145, MiR-195, and possibly other non-protein-coding functional RNAs to control adult mouse pancreatic differentiation. Interestingly, our study revealed that Rfx6 played an important role in insulin translation by binding to the Eif2ak1, Upf1, and Eif5. Our data provide direct target genes of Rfx6 during pancreas development and point to Rfx6 as a potential therapeutic target for improving insulin protein content.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Insulin/biosynthesis , Pancreas/growth & development , Regulatory Factor X Transcription Factors/genetics , Animals , Cell Differentiation , Gene Expression Regulation , Insulin/metabolism , Mice , Mice, Inbred C57BL , Organogenesis , Pancreas/chemistry , Protein Binding , Regulatory Factor X Transcription Factors/metabolism
18.
BMC Med Genet ; 19(1): 22, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29439679

ABSTRACT

BACKGROUND: Maturity-onset diabetes of the young (MODY) is an early-onset, autosomal dominant form of non-insulin dependent diabetes. Genetic diagnosis of MODY can transform patient management. Earlier data on the genetic predisposition to MODY have come primarily from familial studies in populations of European origin. METHODS: In this study, we carried out a comprehensive genomic analysis of 289 individuals from India that included 152 clinically diagnosed MODY cases to identify variants in known MODY genes. Further, we have analyzed exome data to identify putative MODY relevant variants in genes previously not implicated in MODY. Functional validation of MODY relevant variants was also performed. RESULTS: We found MODY 3 (HNF1A; 7.2%) to be most frequently mutated followed by MODY 12 (ABCC8; 3.3%). They together account for ~ 11% of the cases. In addition to known MODY genes, we report the identification of variants in RFX6, WFS1, AKT2, NKX6-1 that may contribute to development of MODY. Functional assessment of the NKX6-1 variants showed that they are functionally impaired. CONCLUSIONS: Our findings showed HNF1A and ABCC8 to be the most frequently mutated MODY genes in south India. Further we provide evidence for additional MODY relevant genes, such as NKX6-1, and these require further validation.


Subject(s)
Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease/epidemiology , Adolescent , Adult , Cohort Studies , Exome , Female , Gene Library , Genomics , Glycated Hemoglobin/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , India/epidemiology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/metabolism , Sequence Analysis, DNA , Sulfonylurea Receptors/genetics , Sulfonylurea Receptors/metabolism , Young Adult
19.
Breast Cancer Res ; 19(1): 104, 2017 Sep 06.
Article in English | MEDLINE | ID: mdl-28877752

ABSTRACT

BACKGROUND: The primary cilium is a microtubule-based and nonmotile organelle functioning as a cellular antenna that is involved in the regulation of cell proliferation, differentiation, and migration. In breast cancer cells, the primary cilium is a structure that decreases in incidence with increasing degrees of transformation and may be biologically more important in estrogen receptor (ERα)-negative breast cancer cells. Split ends (SPEN) is an ERα corepressor that we have identified as a tumor suppressor protein in ERα-positive breast cancer cells whose hormone-independent roles in breast cancer have never been explored. METHODS: We determined the hormone-independent transcriptional program regulated by the ERα cofactor SPEN in breast cancer using DNA microarrays. The biological functions regulated by SPEN independently of hormones were studied in vitro in ERα-positive and ERα-negative breast cancer cells. Finally, we examined the clinical relevance of SPEN expression in cohorts of breast cancer samples with outcome data. RESULTS: We found that SPEN is coexpressed with a number of genes involved in ciliary biology, including the ciliogenic transcription factor RFX3, in a hormone-independent manner. SPEN reexpression in T47D cells containing a nonsense mutation in SPEN restored the primary cilium, whereas its knockdown in MCF10A and Hs578T cells considerably decreased primary cilia levels. We also report that SPEN regulates migration in breast cells, but only in those harboring primary cilia, and that KIF3A silencing, a critical factor in primary cilia, partially reverses SPEN's effects, suggesting that SPEN may coordinate cellular movement through primary cilia-dependent mechanisms. Finally, we found that high SPEN RNA levels were predictive of early metastasis in two independent cohorts of 77 (HR 2.25, P = 0.03) and 170 (HR = 2.23, P = 0.004) patients with ERα-negative breast cancer. CONCLUSIONS: Together, our data demonstrate a role for SPEN in the regulation of primary cilia formation and cell migration in breast cancer cells, which may collectively explain why its expression is associated with time to metastasis in cohorts of patients with ERα-negative breast cancers.


Subject(s)
Breast Neoplasms/genetics , Cell Movement/genetics , Cilia/metabolism , Homeodomain Proteins/genetics , Nuclear Proteins/genetics , Biomarkers, Tumor , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , DNA-Binding Proteins , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Humans , Neoplasm Metastasis , Nuclear Proteins/metabolism , RNA, Small Interfering/genetics , RNA-Binding Proteins , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/metabolism
20.
FASEB J ; 30(10): 3578-3587, 2016 10.
Article in English | MEDLINE | ID: mdl-27451412

ABSTRACT

DYX1C1, DCDC2, and KIAA0319 are three of the most replicated dyslexia candidate genes (DCGs). Recently, these DCGs were implicated in functions at the cilium. Here, we investigate the regulation of these DCGs by Regulatory Factor X transcription factors (RFX TFs), a gene family known for transcriptionally regulating ciliary genes. We identify conserved X-box motifs in the promoter regions of DYX1C1, DCDC2, and KIAA0319 and demonstrate their functionality, as well as the ability to recruit RFX TFs using reporter gene and electrophoretic mobility shift assays. Furthermore, we uncover a complex regulation pattern between RFX1, RFX2, and RFX3 and their significant effect on modifying the endogenous expression of DYX1C1 and DCDC2 in a human retinal pigmented epithelial cell line immortalized with hTERT (hTERT-RPE1). In addition, induction of ciliogenesis increases the expression of RFX TFs and DCGs. At the protein level, we show that endogenous DYX1C1 localizes to the base of the cilium, whereas DCDC2 localizes along the entire axoneme of the cilium, thereby validating earlier localization studies using overexpression models. Our results corroborate the emerging role of DCGs in ciliary function and characterize functional noncoding elements, X-box promoter motifs, in DCG promoter regions, which thus can be targeted for mutation screening in dyslexia and ciliopathies associated with these genes.-Tammimies, K., Bieder, A., Lauter, G., Sugiaman-Trapman, D., Torchet, R., Hokkanen, M.-E., Burghoorn, J., Castrén, E., Kere, J., Tapia-Páez, I., Swoboda, P. Ciliary dyslexia candidate genes DYX1C1 and DCDC2 are regulated by Regulatory Factor (RF) X transcription factors through X-box promoter motifs.


Subject(s)
Cilia/metabolism , Dyslexia/metabolism , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Regulatory Factor X Transcription Factors/metabolism , Animals , Binding Sites/genetics , Caenorhabditis elegans , Cells, Cultured , Cytoskeletal Proteins , Genes, Reporter , Humans
SELECTION OF CITATIONS
SEARCH DETAIL