Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Publication year range
1.
Immunity ; 56(1): 58-77.e11, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36521495

ABSTRACT

Obesity-induced chronic liver inflammation is a hallmark of nonalcoholic steatohepatitis (NASH)-an aggressive form of nonalcoholic fatty liver disease. However, it remains unclear how such a low-grade, yet persistent, inflammation is sustained in the liver. Here, we show that the macrophage phagocytic receptor TREM2, induced by hepatocyte-derived sphingosine-1-phosphate, was required for efferocytosis of lipid-laden apoptotic hepatocytes and thereby maintained liver immune homeostasis. However, prolonged hypernutrition led to the production of proinflammatory cytokines TNF and IL-1ß in the liver to induce TREM2 shedding through ADAM17-dependent proteolytic cleavage. Loss of TREM2 resulted in aberrant accumulation of dying hepatocytes, thereby further augmenting proinflammatory cytokine production. This ultimately precipitated a vicious cycle that licensed chronic inflammation to drive simple steatosis transition to NASH. Therefore, impaired macrophage efferocytosis is a previously unrecognized key pathogenic event that enables chronic liver inflammation in obesity. Blocking TREM2 cleavage to restore efferocytosis may represent an effective strategy to treat NASH.


Subject(s)
Non-alcoholic Fatty Liver Disease , Overnutrition , Humans , Non-alcoholic Fatty Liver Disease/pathology , Overnutrition/pathology , Liver/pathology , Inflammation/pathology , Obesity/pathology , Membrane Glycoproteins , Receptors, Immunologic
2.
Mol Cell ; 82(6): 1225-1238.e6, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35196517

ABSTRACT

The long-range interactions of cis-regulatory elements (cREs) play a central role in gene regulation. cREs can be characterized as accessible chromatin sequences. However, it remains technically challenging to comprehensively identify their spatial interactions. Here, we report a new method HiCAR (Hi-C on accessible regulatory DNA), which utilizes Tn5 transposase and chromatin proximity ligation, for the analysis of open-chromatin-anchored interactions with low-input cells. By applying HiCAR in human embryonic stem cells and lymphoblastoid cells, we demonstrate that HiCAR identifies high-resolution chromatin contacts with an efficiency comparable with that of in situ Hi-C over all distance ranges. Interestingly, we found that the "poised" gene promoters exhibit silencer-like function to repress the expression of distal genes via promoter-promoter interactions. Lastly, we applied HiCAR to 30,000 primary human muscle stem cells and demonstrated that HiCAR is capable of analyzing chromatin accessibility and looping using low-input primary cells and clinical samples.


Subject(s)
Chromatin , Regulatory Sequences, Nucleic Acid , Chromatin/genetics , DNA , Gene Expression Regulation , Humans , Promoter Regions, Genetic
3.
Nature ; 623(7987): 633-642, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37938770

ABSTRACT

Trimethylation of histone H3 lysine 9 (H3K9me3) is crucial for the regulation of gene repression and heterochromatin formation, cell-fate determination and organismal development1. H3K9me3 also provides an essential mechanism for silencing transposable elements1-4. However, previous studies have shown that canonical H3K9me3 readers (for example, HP1 (refs. 5-9) and MPP8 (refs. 10-12)) have limited roles in silencing endogenous retroviruses (ERVs), one of the main transposable element classes in the mammalian genome13. Here we report that trinucleotide-repeat-containing 18 (TNRC18), a poorly understood chromatin regulator, recognizes H3K9me3 to mediate the silencing of ERV class I (ERV1) elements such as LTR12 (ref. 14). Biochemical, biophysical and structural studies identified the carboxy-terminal bromo-adjacent homology (BAH) domain of TNRC18 (TNRC18(BAH)) as an H3K9me3-specific reader. Moreover, the amino-terminal segment of TNRC18 is a platform for the direct recruitment of co-repressors such as HDAC-Sin3-NCoR complexes, thus enforcing optimal repression of the H3K9me3-demarcated ERVs. Point mutagenesis that disrupts the TNRC18(BAH)-mediated H3K9me3 engagement caused neonatal death in mice and, in multiple mammalian cell models, led to derepressed expression of ERVs, which affected the landscape of cis-regulatory elements and, therefore, gene-expression programmes. Collectively, we describe a new H3K9me3-sensing and regulatory pathway that operates to epigenetically silence evolutionarily young ERVs and exert substantial effects on host genome integrity, transcriptomic regulation, immunity and development.


Subject(s)
Endogenous Retroviruses , Gene Silencing , Histones , Intracellular Signaling Peptides and Proteins , Lysine , Retroelements , Animals , Humans , Mice , Chromatin/genetics , Chromatin/metabolism , Co-Repressor Proteins/metabolism , Endogenous Retroviruses/genetics , Epigenesis, Genetic , Gene Expression Profiling , Genome/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lysine/metabolism , Methylation , Protein Domains , Retroelements/genetics , Terminal Repeat Sequences/genetics , Animals, Newborn , Cell Line
4.
Nature ; 611(7935): 387-398, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36289338

ABSTRACT

Acute myeloid leukaemia (AML) represents a set of heterogeneous myeloid malignancies, and hallmarks include mutations in epigenetic modifiers, transcription factors and kinases1-5. The extent to which mutations in AML drive alterations in chromatin 3D structure and contribute to myeloid transformation is unclear. Here we use Hi-C and whole-genome sequencing to analyse 25 samples from patients with AML and 7 samples from healthy donors. Recurrent and subtype-specific alterations in A/B compartments, topologically associating domains and chromatin loops were identified. RNA sequencing, ATAC with sequencing and CUT&Tag for CTCF, H3K27ac and H3K27me3 in the same AML samples also revealed extensive and recurrent AML-specific promoter-enhancer and promoter-silencer loops. We validated the role of repressive loops on their target genes by CRISPR deletion and interference. Structural variation-induced enhancer-hijacking and silencer-hijacking events were further identified in AML samples. Hijacked enhancers play a part in AML cell growth, as demonstrated by CRISPR screening, whereas hijacked silencers have a downregulating role, as evidenced by CRISPR-interference-mediated de-repression. Finally, whole-genome bisulfite sequencing of 20 AML and normal samples revealed the delicate relationship between DNA methylation, CTCF binding and 3D genome structure. Treatment of AML cells with a DNA hypomethylating agent and triple knockdown of DNMT1, DNMT3A and DNMT3B enabled the manipulation of DNA methylation to revert 3D genome organization and gene expression. Overall, this study provides a resource for leukaemia studies and highlights the role of repressive loops and hijacked cis elements in human diseases.


Subject(s)
Genome, Human , Leukemia, Myeloid, Acute , Humans , Chromatin/genetics , DNA Methylation , Leukemia, Myeloid, Acute/genetics , Genome, Human/genetics , Promoter Regions, Genetic , Enhancer Elements, Genetic , Gene Silencing , Reproducibility of Results , CRISPR-Cas Systems , Sequence Analysis , DNA (Cytosine-5-)-Methyltransferases , Gene Expression Regulation, Leukemic
5.
Mol Cell ; 76(3): 453-472.e8, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31519520

ABSTRACT

MYOD-directed fibroblast trans-differentiation into skeletal muscle provides a unique model to investigate how one transcription factor (TF) reconfigures the three-dimensional chromatin architecture to control gene expression, which is otherwise achieved by the combinatorial activities of multiple TFs. Integrative analysis of genome-wide high-resolution chromatin interactions, MYOD and CTCF DNA-binding profile, and gene expression, revealed that MYOD directs extensive re-wiring of interactions involving cis-regulatory and structural genomic elements, including promoters, enhancers, and insulated neighborhoods (INs). Re-configured INs were hot-spots of differential interactions, whereby MYOD binding to highly constrained sequences at IN boundaries and/or inside INs led to alterations of promoter-enhancer interactions to repress cell-of-origin genes and to activate muscle-specific genes. Functional evidence shows that MYOD-directed re-configuration of chromatin interactions temporally preceded the effect on gene expression and was mediated by direct MYOD-DNA binding. These data illustrate a model whereby a single TF alters multi-loop hubs to drive somatic cell trans-differentiation.


Subject(s)
Cell Transdifferentiation , Cellular Reprogramming , Chromatin Assembly and Disassembly , Chromatin/metabolism , Fibroblasts/metabolism , Muscle Development , MyoD Protein/metabolism , Myoblasts, Skeletal/metabolism , Animals , Binding Sites , Cell Line , Cell Transdifferentiation/genetics , Chromatin/genetics , Female , Gene Expression Regulation, Developmental , Humans , Mice , Muscle Development/genetics , MyoD Protein/genetics , Nucleic Acid Conformation , Phenotype , Protein Binding , Structure-Activity Relationship , Transcription, Genetic
6.
Development ; 149(23)2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36504079

ABSTRACT

There are fundamental differences in how neonatal and adult intestines absorb nutrients. In adults, macromolecules are broken down into simpler molecular components in the lumen of the small intestine, then absorbed. In contrast, neonates are thought to rely on internalization of whole macromolecules and subsequent degradation in the lysosome. Here, we identify the Maf family transcription factors MAFB and c-MAF as markers of terminally differentiated intestinal enterocytes throughout life. The expression of these factors is regulated by HNF4α and HNF4γ, master regulators of enterocyte cell fate. Loss of Maf factors results in a neonatal-specific failure to thrive and loss of macromolecular nutrient uptake. RNA-Seq and CUT&RUN analyses defined an endo-lysosomal program as being downstream of these transcription factors. We demonstrate major transcriptional changes in metabolic pathways, including fatty acid oxidation and increases in peroxisome number, in response to loss of Maf proteins. Finally, we show that loss of BLIMP1, a repressor of adult enterocyte genes, shows highly overlapping changes in gene expression and similar defects in macromolecular uptake. This work defines transcriptional regulators that are necessary for nutrient uptake in neonatal enterocytes.


Subject(s)
Maf Transcription Factors , Nutrients , Mice , Animals , Biological Transport , Cell Differentiation , Transcription Factors/genetics , Proto-Oncogene Proteins c-maf
7.
Nature ; 569(7757): 570-575, 2019 05.
Article in English | MEDLINE | ID: mdl-31019297

ABSTRACT

Precision oncology hinges on linking tumour genotype with molecularly targeted drugs1; however, targeting the frequently dysregulated metabolic landscape of cancer has proven to be a major challenge2. Here we show that tissue context is the major determinant of dependence on the nicotinamide adenine dinucleotide (NAD) metabolic pathway in cancer. By analysing more than 7,000 tumours and 2,600 matched normal samples of 19 tissue types, coupled with mathematical modelling and extensive in vitro and in vivo analyses, we identify a simple and actionable set of 'rules'. If the rate-limiting enzyme of de novo NAD synthesis, NAPRT, is highly expressed in a normal tissue type, cancers that arise from that tissue will have a high frequency of NAPRT amplification and be completely and irreversibly dependent on NAPRT for survival. By contrast, tumours that arise from normal tissues that do not express NAPRT highly are entirely dependent on the NAD salvage pathway for survival. We identify the previously unknown enhancer that underlies this dependence. Amplification of NAPRT is shown to generate a pharmacologically actionable tumour cell dependence for survival. Dependence on another rate-limiting enzyme of the NAD synthesis pathway, NAMPT, as a result of enhancer remodelling is subject to resistance by NMRK1-dependent synthesis of NAD. These results identify a central role for tissue context in determining the choice of NAD biosynthetic pathway, explain the failure of NAMPT inhibitors, and pave the way for more effective treatments.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Amplification , NAD/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Animals , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/metabolism , Cell Death , Cell Line, Tumor , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/metabolism , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasms/enzymology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism
8.
Nature ; 575(7784): 699-703, 2019 11.
Article in English | MEDLINE | ID: mdl-31748743

ABSTRACT

Oncogenes are commonly amplified on particles of extrachromosomal DNA (ecDNA) in cancer1,2, but our understanding of the structure of ecDNA and its effect on gene regulation is limited. Here, by integrating ultrastructural imaging, long-range optical mapping and computational analysis of whole-genome sequencing, we demonstrate the structure of circular ecDNA. Pan-cancer analyses reveal that oncogenes encoded on ecDNA are among the most highly expressed genes in the transcriptome of the tumours, linking increased copy number with high transcription levels. Quantitative assessment of the chromatin state reveals that although ecDNA is packaged into chromatin with intact domain structure, it lacks higher-order compaction that is typical of chromosomes and displays significantly enhanced chromatin accessibility. Furthermore, ecDNA is shown to have a significantly greater number of ultra-long-range interactions with active chromatin, which provides insight into how the structure of circular ecDNA affects oncogene function, and connects ecDNA biology with modern cancer genomics and epigenetics.


Subject(s)
Chromatin/genetics , DNA, Circular/metabolism , Gene Expression Regulation, Neoplastic/genetics , Neoplasms/genetics , Oncogenes/genetics , Cell Line, Tumor , Chromatin/chemistry , DNA, Circular/genetics , Humans , Microscopy, Electron, Scanning , Neoplasms/physiopathology
9.
Nature ; 560(7720): 655-660, 2018 08.
Article in English | MEDLINE | ID: mdl-30135582

ABSTRACT

Mammalian cells are surrounded by neighbouring cells and extracellular matrix (ECM), which provide cells with structural support and mechanical cues that influence diverse biological processes1. The Hippo pathway effectors YAP (also known as YAP1) and TAZ (also known as WWTR1) are regulated by mechanical cues and mediate cellular responses to ECM stiffness2,3. Here we identified the Ras-related GTPase RAP2 as a key intracellular signal transducer that relays ECM rigidity signals to control mechanosensitive cellular activities through YAP and TAZ. RAP2 is activated by low ECM stiffness, and deletion of RAP2 blocks the regulation of YAP and TAZ by stiffness signals and promotes aberrant cell growth. Mechanistically, matrix stiffness acts through phospholipase Cγ1 (PLCγ1) to influence levels of phosphatidylinositol 4,5-bisphosphate and phosphatidic acid, which activates RAP2 through PDZGEF1 and PDZGEF2 (also known as RAPGEF2 and RAPGEF6). At low stiffness, active RAP2 binds to and stimulates MAP4K4, MAP4K6, MAP4K7 and ARHGAP29, resulting in activation of LATS1 and LATS2 and inhibition of YAP and TAZ. RAP2, YAP and TAZ have pivotal roles in mechanoregulated transcription, as deletion of YAP and TAZ abolishes the ECM stiffness-responsive transcriptome. Our findings show that RAP2 is a molecular switch in mechanotransduction, thereby defining a mechanosignalling pathway from ECM stiffness to the nucleus.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Signal Transduction , rap GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Transformation, Neoplastic , Extracellular Matrix/chemistry , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , GTPase-Activating Proteins/metabolism , Germinal Center Kinases , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Nerve Tissue Proteins/metabolism , Phospholipase C gamma/metabolism , Phosphoproteins/metabolism , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transcriptome , YAP-Signaling Proteins , rap GTP-Binding Proteins/genetics
10.
Proc Natl Acad Sci U S A ; 116(37): 18584-18589, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31462499

ABSTRACT

Nearly two-thirds of cancer patients are treated with radiation therapy (RT), often with the intent to achieve complete and permanent tumor regression (local control). RT is the primary treatment modality used to achieve local control for many malignancies, including locally advanced cervical cancer, head and neck cancer, and lung cancer. The addition of concurrent platinum-based radiosensitizing chemotherapy improves local control and patient survival. Enhanced outcomes with concurrent chemoradiotherapy may result from increased direct killing of tumor cells and effects on nontumor cell populations. Many patients treated with concurrent chemoradiotherapy exhibit a decline in neutrophil count, but the effects of neutrophils on radiation therapy are controversial. To investigate the clinical significance of neutrophils in the response to RT, we examined patient outcomes and circulating neutrophil counts in cervical cancer patients treated with definitive chemoradiation. Although pretreatment neutrophil count did not correlate with outcome, lower absolute neutrophil count after starting concurrent chemoradiotherapy was associated with higher rates of local control, metastasis-free survival, and overall survival. To define the role of neutrophils in tumor response to RT, we used genetic and pharmacological approaches to deplete neutrophils in an autochthonous mouse model of soft tissue sarcoma. Neutrophil depletion prior to image-guided focal irradiation improved tumor response to RT. Our results indicate that neutrophils promote resistance to radiation therapy. The efficacy of chemoradiotherapy may depend on the impact of treatment on peripheral neutrophil count, which has the potential to serve as an inexpensive and widely available biomarker.


Subject(s)
Chemoradiotherapy , Neutrophils/immunology , Radiation Tolerance/immunology , Sarcoma/therapy , Uterine Cervical Neoplasms/therapy , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Disease Models, Animal , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Leukocyte Count , Mice , Mice, Transgenic , Middle Aged , Radiation Tolerance/genetics , Retrospective Studies , Sarcoma/blood , Sarcoma/immunology , Uterine Cervical Neoplasms/blood , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/mortality , Whole-Body Irradiation , Young Adult
11.
Nature ; 518(7539): 331-6, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25693564

ABSTRACT

Higher-order chromatin structure is emerging as an important regulator of gene expression. Although dynamic chromatin structures have been identified in the genome, the full scope of chromatin dynamics during mammalian development and lineage specification remains to be determined. By mapping genome-wide chromatin interactions in human embryonic stem (ES) cells and four human ES-cell-derived lineages, we uncover extensive chromatin reorganization during lineage specification. We observe that although self-associating chromatin domains are stable during differentiation, chromatin interactions both within and between domains change in a striking manner, altering 36% of active and inactive chromosomal compartments throughout the genome. By integrating chromatin interaction maps with haplotype-resolved epigenome and transcriptome data sets, we find widespread allelic bias in gene expression correlated with allele-biased chromatin states of linked promoters and distal enhancers. Our results therefore provide a global view of chromatin dynamics and a resource for studying long-range control of gene expression in distinct human cell lineages.


Subject(s)
Cell Differentiation , Chromatin Assembly and Disassembly , Chromatin/chemistry , Chromatin/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/genetics , Alleles , Allelic Imbalance/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Enhancer Elements, Genetic/genetics , Epigenomics , Gene Regulatory Networks , Humans , Promoter Regions, Genetic/genetics , Reproducibility of Results
12.
Nat Methods ; 14(6): 629-635, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28417999

ABSTRACT

Millions of cis-regulatory elements are predicted to be present in the human genome, but direct evidence for their biological function is scarce. Here we report a high-throughput method, cis-regulatory element scan by tiling-deletion and sequencing (CREST-seq), for the unbiased discovery and functional assessment of cis-regulatory sequences in the genome. We used it to interrogate the 2-Mb POU5F1 locus in human embryonic stem cells, and identified 45 cis-regulatory elements. A majority of these elements have active chromatin marks, DNase hypersensitivity, and occupancy by multiple transcription factors, which confirms the utility of chromatin signatures in cis-element mapping. Notably, 17 of them are previously annotated promoters of functionally unrelated genes, and like typical enhancers, they form extensive spatial contacts with the POU5F1 promoter. These results point to the commonality of enhancer-like promoters in the human genome.


Subject(s)
Chromosome Mapping/methods , Genetic Testing/methods , Regulatory Sequences, Nucleic Acid/genetics , Algorithms , Cells, Cultured , Embryonic Stem Cells/physiology , Gene Expression Regulation/genetics , High-Throughput Nucleotide Sequencing , Humans , Sequence Analysis, DNA , Single-Cell Analysis
13.
Genome Res ; 26(3): 397-405, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26813977

ABSTRACT

With <2% of the human genome coding for proteins, a major challenge is to interpret the function of the noncoding DNA. Millions of regulatory sequences have been predicted in the human genome through analysis of DNA methylation, chromatin modification, hypersensitivity to nucleases, and transcription factor binding, but few have been shown to regulate transcription in their native contexts. We have developed a high-throughput CRISPR/Cas9-based genome-editing strategy and used it to interrogate 174 candidate regulatory sequences within the 1-Mbp POU5F1 locus in human embryonic stem cells (hESCs). We identified two classical regulatory elements, including a promoter and a proximal enhancer, that are essential for POU5F1 transcription in hESCs. Unexpectedly, we also discovered a new class of enhancers that contribute to POU5F1 transcription in an unusual way: Disruption of such sequences led to a temporary loss of POU5F1 transcription that is fully restored after a few rounds of cell division. These results demonstrate the utility of high-throughput screening for functional characterization of noncoding DNA and reveal a previously unrecognized layer of gene regulation in human cells.


Subject(s)
CRISPR-Cas Systems , Gene Targeting , Genetic Testing , Phenotype , Cell Line , Enhancer Elements, Genetic , Gene Expression Regulation , Gene Targeting/methods , Genetic Testing/methods , Genome, Human , High-Throughput Screening Assays , Humans , Octamer Transcription Factor-3/genetics , Regulatory Sequences, Nucleic Acid , Sequence Deletion
14.
J Biol Chem ; 289(1): 529-39, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24247238

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma found in children and young adults. It is characterized by the expression of a number of skeletal muscle-specific proteins, including MyoD and muscle α-actin. However, unlike normal myoblasts, RMS cells differentiate poorly both in vivo and in culture. As microRNAs are known to regulate tumorigenesis, intensive efforts have been made to identify microRNAs that are involved in RMS development. In this work, we found that miR-203 was frequently down-regulated by promoter hypermethylation in both RMS cell lines and RMS biopsies and could be reactivated by DNA-demethylating agents. Re-expression of miR-203 in RMS cells inhibited their migration and proliferation and promoted terminal myogenic differentiation. Mechanistically, miR-203 exerts its tumor-suppressive effect by directly targeting p63 and leukemia inhibitory factor receptor in RMS cells, which promotes myogenic differentiation by inhibiting the Notch and the JAK1/STAT1/STAT3 pathways, respectively. Our work reveals that miR-203 functions as a tumor suppressor in RMS development.


Subject(s)
DNA Methylation , DNA, Neoplasm/metabolism , Down-Regulation , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , MicroRNAs/biosynthesis , Promoter Regions, Genetic , Rhabdomyosarcoma/metabolism , Animals , Cell Differentiation/genetics , Cell Line, Tumor , DNA, Neoplasm/genetics , Female , Humans , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Male , Mice , Mice, Nude , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Receptors, OSM-LIF/genetics , Receptors, OSM-LIF/metabolism , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
15.
Ann Hematol ; 94(9): 1505-14, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26051902

ABSTRACT

Acute graft-versus-host disease (aGVHD) is one of the major causes of morbidity and mortality in patients receiving allogeneic hematopoietic cell transplantation (allo-HSCT). MicroRNAs (miRs) were found to have the potential to be the new biomarkers of aGVHD. In this study, we collected samples from 98 patients who underwent allo-HSCT; 63 patients developed aGVHD, and 35 patients did not. Plasma samples were collected at three time points (before aGVHD, at the onset of aGVHD, and after aGVHD) from 52 patients, and the miR-586 expression level was detected by quantitative real-time PCR. We found that the plasma miR-586 level was decreased at the onset of grade I-II aGVHD (P = 0.074). In contrast, when infections were detected, plasma miR-586 level was increased. Moreover, we detected the miR-586 expression level in patients who had infections but did not have aGVHD, and we found that miR-586 was upregulated (P = 0.005). We also compared the plasma miR-586 level at day 7 after transplantation between aGVHD patients and control patients. In the aGVHD group, there was a considerably higher miR-586 expression in comparison with the non-aGVHD group (P < 0.05). A more significant difference between the two groups was found when the patients with infections were excluded (P = 0.004). Furthermore, receive operating characteristic (ROC) analysis indicated that a higher expression level of miR-586 at day 7 could predict impending aGVHD. The optimal cutoff value of miR-586 to predict aGVHD was 2200 copies/µL with a sensitivity of 87.5 % and specificity of 55.0 %, and the area under the curve (AUC) was 0.739 (95 % CI 0.598-0.880, P = 0.004). Our study suggests that miR-586 might participate in the occurrence of aGVHD and could be a putative target for novel aGVHD therapy. The plasma level of miR-586 at day 7 after allo-HSCT would be a potential biomarker for predicting the occurrence of aGVHD.


Subject(s)
Gene Expression Regulation , Graft vs Host Disease/blood , Hematopoietic Stem Cell Transplantation , MicroRNAs/blood , Acute Disease , Adolescent , Adult , Allografts , Biomarkers/blood , Child , Child, Preschool , Female , Humans , Male , Middle Aged
16.
Dev Cell ; 59(5): 676-691.e5, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38290519

ABSTRACT

Regeneration involves gene expression changes explained in part by context-dependent recruitment of transcriptional activators to distal enhancers. Silencers that engage repressive transcriptional complexes are less studied than enhancers and more technically challenging to validate, but they potentially have profound biological importance for regeneration. Here, we identified candidate silencers through a screening process that examined the ability of DNA sequences to limit injury-induced gene expression in larval zebrafish after fin amputation. A short sequence (s1) on chromosome 5 near several genes that reduce expression during adult fin regeneration could suppress promoter activity in stable transgenic lines and diminish nearby gene expression in knockin lines. High-resolution analysis of chromatin organization identified physical associations of s1 with gene promoters occurring preferentially during fin regeneration, and genomic deletion of s1 elevated the expression of these genes after fin amputation. Our study provides methods to identify "tissue regeneration silencer elements" (TRSEs) with the potential to reduce unnecessary or deleterious gene expression during regeneration.


Subject(s)
Silencer Elements, Transcriptional , Zebrafish , Animals , Zebrafish/genetics , Animals, Genetically Modified , Promoter Regions, Genetic
17.
Res Sq ; 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38883785

ABSTRACT

Enchondromas are a common tumor in bone that can occur as multiple lesions in enchondromatosis, which is associated with deformity of the effected bone. These lesions harbor mutations in IDH and driving expression of a mutant Idh1 in Col2 expressing cells in mice causes an enchondromatosis phenotype. In this study we compared growth plates from E18.5 mice expressing a mutant Idh1 with control littermates using single cell RNA sequencing. Data from Col2 expressing cells were analyzed using UMAP and RNA pseudo-time analyses. A unique cluster of cells was identified in the mutant growth plates that expressed genes known to be upregulated in enchondromas. There was also a cluster of cells that was underrepresented in the mutant growth plates that expressed genes known to be important in longitudinal bone growth. Immunofluorescence showed that the genes from the unique cluster identified in the mutant growth plates were expressed in multiple growth plate anatomic zones, and pseudo-time analysis also suggested these cells could arise from multiple growth plate chondrocyte subpopulations. This data identifies subpopulations of cells in control and mutant growth plates, and supports the notion that a mutant Idh1 alters the subpopulations of growth plate chondrocytes, resulting a subpopulation of cells that become enchondromas at the expense of other populations that contribute to longitudinal growth.

18.
iScience ; 27(7): 110187, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38989451

ABSTRACT

Intratumoral heterogeneity is common in cancer, particularly in sarcomas like undifferentiated pleomorphic sarcoma (UPS), where individual cells demonstrate a high degree of cytogenic diversity. Previous studies showed that a small subset of cells within UPS, known as the metastatic clone (MC), as responsible for metastasis. Using a CRISPR-based genomic screen in-vivo, we identified the COMPASS complex member Setd1a as a key regulator maintaining the metastatic phenotype of the MC in murine UPS. Depletion of Setd1a inhibited metastasis development in the MC. Transcriptome and chromatin sequencing revealed COMPASS complex target genes in UPS, such as Cxcl10, downregulated in the MC. Deleting Cxcl10 in non-MC cells increased their metastatic potential. Treating mice with human UPS xenografts with a COMPASS complex inhibitor suppressed metastasis without affecting tumor growth in the primary tumor. Our data identified an epigenetic program in a subpopulation of sarcoma cells that maintains metastatic potential.

19.
J Invest Dermatol ; 2024 May 23.
Article in English | MEDLINE | ID: mdl-38796140

ABSTRACT

UBE2N, a Lys63 ubiquitin-conjugating enzyme, plays critical roles in embryogenesis and immune system development and function. However, its roles in adult epithelial tissue homeostasis and pathogenesis are unclear. We generated conditional mouse models that deleted Ube2n in skin cells in a temporally and spatially controlled manner. We found that Ube2n knockout in the adult skin keratinocytes induced a range of inflammatory skin defects characteristic of psoriatic and actinic keratosis. These included inflammation, epidermal and dermal thickening, parakeratosis, and increased immune cell infiltration as well as signs of edema and blistering. Single-cell transcriptomic analyses and RT-qPCR showed that Ube2n-knockout keratinocytes expressed elevated myeloid cell chemoattractants such as Cxcl1 and Cxcl2 and decreased the homeostatic T lymphocyte chemoattractant Ccl27a. Consistently, the infiltrating immune cells were predominantly myeloid-derived cells, including neutrophils and M1-like macrophages, which expressed high levels of inflammatory cytokines such as Il1ß and Il24. Pharmacological blockade of the IL-1 receptor associated kinases (IRAK1/4) alleviated inflammation, epidermal and dermal thickening, and immune infiltration of the Ube2n-mutant skin. Together, these findings highlight a key role of keratinocyte UBE2N in maintenance of epidermal homeostasis and skin immunity and identify IRAK1/4 as potential therapeutic target for inflammatory skin disorders.

20.
Cell Stem Cell ; 31(6): 921-939.e17, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38692273

ABSTRACT

Nephron progenitor cells (NPCs) self-renew and differentiate into nephrons, the functional units of the kidney. Here, manipulation of p38 and YAP activity allowed for long-term clonal expansion of primary mouse and human NPCs and induced NPCs (iNPCs) from human pluripotent stem cells (hPSCs). Molecular analyses demonstrated that cultured iNPCs closely resemble primary human NPCs. iNPCs generated nephron organoids with minimal off-target cell types and enhanced maturation of podocytes relative to published human kidney organoid protocols. Surprisingly, the NPC culture medium uncovered plasticity in human podocyte programs, enabling podocyte reprogramming to an NPC-like state. Scalability and ease of genome editing facilitated genome-wide CRISPR screening in NPC culture, uncovering genes associated with kidney development and disease. Further, NPC-directed modeling of autosomal-dominant polycystic kidney disease (ADPKD) identified a small-molecule inhibitor of cystogenesis. These findings highlight a broad application for the reported iNPC platform in the study of kidney development, disease, plasticity, and regeneration.


Subject(s)
Nephrons , Organoids , Animals , Organoids/cytology , Organoids/metabolism , Humans , Nephrons/cytology , Mice , Cell Differentiation , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Podocytes/metabolism , Podocytes/cytology , Kidney/pathology , Polycystic Kidney, Autosomal Dominant/pathology , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/genetics , Models, Biological , Gene Editing
SELECTION OF CITATIONS
SEARCH DETAIL