Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
2.
Development ; 144(7): 1221-1234, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28174249

ABSTRACT

Mouse embryonic stem (ES) cells are locked into self-renewal by shielding from inductive cues. Release from this ground state in minimal conditions offers a system for delineating developmental progression from naïve pluripotency. Here, we examine the initial transition process. The ES cell population behaves asynchronously. We therefore exploited a short-half-life Rex1::GFP reporter to isolate cells either side of exit from naïve status. Extinction of ES cell identity in single cells is acute. It occurs only after near-complete elimination of naïve pluripotency factors, but precedes appearance of lineage specification markers. Cells newly departed from the ES cell state display features of early post-implantation epiblast and are distinct from primed epiblast. They also exhibit a genome-wide increase in DNA methylation, intermediate between early and late epiblast. These findings are consistent with the proposition that naïve cells transition to a distinct formative phase of pluripotency preparatory to lineage priming.


Subject(s)
Cell Tracking , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Lineage , Cell Self Renewal , DNA Methylation/genetics , Down-Regulation , Embryo, Mammalian/cytology , Embryonic Stem Cells/metabolism , Genes, Reporter , Germ Layers/cytology , Kinetics , Mice , Pluripotent Stem Cells/metabolism , Stem Cell Transplantation , Transcription Factors/metabolism , Transcription, Genetic
3.
Nat Methods ; 13(3): 229-232, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26752769

ABSTRACT

We report scM&T-seq, a method for parallel single-cell genome-wide methylome and transcriptome sequencing that allows for the discovery of associations between transcriptional and epigenetic variation. Profiling of 61 mouse embryonic stem cells confirmed known links between DNA methylation and transcription. Notably, the method revealed previously unrecognized associations between heterogeneously methylated distal regulatory elements and transcription of key pluripotency genes.


Subject(s)
Embryonic Stem Cells/physiology , Epigenesis, Genetic/genetics , High-Throughput Nucleotide Sequencing/methods , Regulatory Elements, Transcriptional/genetics , Transcription Factors/genetics , Animals , Base Sequence , Cells, Cultured , Mice , Molecular Sequence Data
4.
PLoS Biol ; 13(12): e1002330, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26717410

ABSTRACT

During pregnancy, the ETS transcription factor ELF5 establishes the milk-secreting alveolar cell lineage by driving a cell fate decision of the mammary luminal progenitor cell. In breast cancer, ELF5 is a key transcriptional determinant of tumor subtype and has been implicated in the development of insensitivity to anti-estrogen therapy. In the mouse mammary tumor virus-Polyoma Middle T (MMTV-PyMT) model of luminal breast cancer, induction of ELF5 levels increased leukocyte infiltration, angiogenesis, and blood vessel permeability in primary tumors and greatly increased the size and number of lung metastasis. Myeloid-derived suppressor cells, a group of immature neutrophils recently identified as mediators of vasculogenesis and metastasis, were recruited to the tumor in response to ELF5. Depletion of these cells using specific Ly6G antibodies prevented ELF5 from driving vasculogenesis and metastasis. Expression signatures in luminal A breast cancers indicated that increased myeloid cell invasion and inflammation were correlated with ELF5 expression, and increased ELF5 immunohistochemical staining predicted much shorter metastasis-free and overall survival of luminal A patients, defining a group who experienced unexpectedly early disease progression. Thus, in the MMTV-PyMT mouse mammary model, increased ELF5 levels drive metastasis by co-opting the innate immune system. As ELF5 has been previously implicated in the development of antiestrogen resistance, this finding implicates ELF5 as a defining factor in the acquisition of the key aspects of the lethal phenotype in luminal A breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Lung Neoplasms/secondary , Lung/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Animals , Breast Neoplasms/immunology , Breast Neoplasms/physiopathology , Breast Neoplasms/virology , Capillary Permeability , Cell Proliferation , DNA-Binding Proteins , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hemorrhage/etiology , Hemorrhage/prevention & control , Humans , Leukocytes/immunology , Leukocytes/pathology , Lung/blood supply , Lung/immunology , Lung/pathology , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Lymphocyte Depletion , Mice, Transgenic , Myeloid Cells/immunology , Myeloid Cells/pathology , Neoplasm Proteins/genetics , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/prevention & control , Neutrophil Infiltration , Polyomavirus/pathogenicity , Proto-Oncogene Proteins c-ets/genetics , Recombinant Fusion Proteins/metabolism , Survival Analysis , Transcription Factors , Tumor Burden
5.
Nat Methods ; 11(8): 817-820, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25042786

ABSTRACT

We report a single-cell bisulfite sequencing (scBS-seq) method that can be used to accurately measure DNA methylation at up to 48.4% of CpG sites. Embryonic stem cells grown in serum or in 2i medium displayed epigenetic heterogeneity, with '2i-like' cells present in serum culture. Integration of 12 individual mouse oocyte datasets largely recapitulated the whole DNA methylome, which makes scBS-seq a versatile tool to explore DNA methylation in rare cells and heterogeneous populations.


Subject(s)
Epigenesis, Genetic , Genome , Sulfites/chemistry , Animals , DNA Methylation , Mice
6.
Breast Cancer Res ; 18(1): 4, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26738740

ABSTRACT

BACKGROUND: E74-like factor 5 (ELF5) is an epithelial-specific member of the E26 transforming sequence (ETS) transcription factor family and a critical regulator of cell fate in the placenta, pulmonary bronchi, and milk-producing alveoli of the mammary gland. ELF5 also plays key roles in malignancy, particularly in basal-like and endocrine-resistant forms of breast cancer. Almost all genes undergo alternative transcription or splicing, which increases the diversity of protein structure and function. Although ELF5 has multiple isoforms, this has not been considered in previous studies of ELF5 function. METHODS: RNA-sequencing data for 6757 samples from The Cancer Genome Atlas were analyzed to characterize ELF5 isoform expression in multiple normal tissues and cancers. Extensive in vitro analysis of ELF5 isoforms, including a 116-gene quantitative polymerase chain reaction panel, was performed in breast cancer cell lines. RESULTS: ELF5 isoform expression was found to be tissue-specific due to alternative promoter use but altered in multiple cancer types. The normal breast expressed one main isoform, while in breast cancer there were subtype-specific alterations in expression. Expression of other ETS factors was also significantly altered in breast cancer, with the basal-like subtype demonstrating a distinct ETS expression profile. In vitro inducible expression of the full-length isoforms 1 and 2, as well as isoform 3 (lacking the Pointed domain) had similar phenotypic and transcriptional effects. CONCLUSIONS: Alternative promoter use, conferring differential regulatory responses, is the main mechanism governing ELF5 action rather than differential transcriptional activity of the isoforms. This understanding of expression and function at the isoform level is a vital first step in realizing the potential of transcription factors such as ELF5 as prognostic markers or therapeutic targets in cancer.


Subject(s)
Alternative Splicing/genetics , DNA-Binding Proteins/genetics , Neoplasms/genetics , Protein Isoforms/genetics , Proto-Oncogene Proteins c-ets/genetics , Animals , DNA-Binding Proteins/biosynthesis , Female , Gene Expression Regulation, Neoplastic/genetics , High-Throughput Nucleotide Sequencing , Humans , Mammary Glands, Human/pathology , Neoplasms/pathology , Organ Specificity , Pregnancy , Promoter Regions, Genetic , Protein Isoforms/biosynthesis , Proto-Oncogene Proteins c-ets/biosynthesis , Transcription Factors
7.
Development ; 140(7): 1397-401, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23462470

ABSTRACT

Progesterone-RankL paracrine signaling has been proposed as a driver of stem cell expansion in the mammary gland, and Elf5 is essential for the differentiation of mammary epithelial progenitor cells. We demonstrate that Elf5 expression is induced by progesterone and that Elf5 and progesterone cooperate to promote alveolar development. The progesterone receptor and Elf5 are expressed in a mutually exclusive pattern, and we identify RankL as the paracrine mediator of the effects of progesterone on Elf5 expression in CD61+ progenitor cells and their consequent differentiation. Blockade of RankL action prevented progesterone-induced side branching and the expansion of Elf5(+) mature luminal cells. These findings describe a mechanism by which steroid hormones can produce the expansion of steroid hormone receptor-negative mammary epithelial cells.


Subject(s)
DNA-Binding Proteins/genetics , Mammary Glands, Animal/drug effects , Progesterone/pharmacology , RANK Ligand/pharmacology , Stem Cells/metabolism , Transcription Factors/genetics , Animals , DNA-Binding Proteins/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/physiology , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , RANK Ligand/metabolism , RANK Ligand/physiology , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Receptors, Progesterone/physiology , Stem Cells/physiology , Transcription Factors/metabolism , Up-Regulation/genetics , Up-Regulation/physiology
8.
Nature ; 468(7320): 98-102, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-20881962

ABSTRACT

Breast cancer is one of the most common cancers in humans and will on average affect up to one in eight women in their lifetime in the United States and Europe. The Women's Health Initiative and the Million Women Study have shown that hormone replacement therapy is associated with an increased risk of incident and fatal breast cancer. In particular, synthetic progesterone derivatives (progestins) such as medroxyprogesterone acetate (MPA), used in millions of women for hormone replacement therapy and contraceptives, markedly increase the risk of developing breast cancer. Here we show that the in vivo administration of MPA triggers massive induction of the key osteoclast differentiation factor RANKL (receptor activator of NF-κB ligand) in mammary-gland epithelial cells. Genetic inactivation of the RANKL receptor RANK in mammary-gland epithelial cells prevents MPA-induced epithelial proliferation, impairs expansion of the CD49f(hi) stem-cell-enriched population, and sensitizes these cells to DNA-damage-induced cell death. Deletion of RANK from the mammary epithelium results in a markedly decreased incidence and delayed onset of MPA-driven mammary cancer. These data show that the RANKL/RANK system controls the incidence and onset of progestin-driven breast cancer.


Subject(s)
Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Progestins/adverse effects , RANK Ligand/metabolism , Animals , Apoptosis/radiation effects , Cell Differentiation , Cell Proliferation/drug effects , DNA Damage , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Female , Gamma Rays , Integrin alpha6/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Medroxyprogesterone Acetate/administration & dosage , Medroxyprogesterone Acetate/adverse effects , Mice , NF-kappa B/metabolism , Osteoclasts/cytology , Phosphoproteins/analysis , Phosphoproteins/immunology , Progestins/administration & dosage , RANK Ligand/deficiency , RANK Ligand/genetics , Receptor Activator of Nuclear Factor-kappa B/deficiency , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism
9.
PLoS Biol ; 10(12): e1001461, 2012.
Article in English | MEDLINE | ID: mdl-23300383

ABSTRACT

We have previously shown that during pregnancy the E-twenty-six (ETS) transcription factor ELF5 directs the differentiation of mammary progenitor cells toward the estrogen receptor (ER)-negative and milk producing cell lineage, raising the possibility that ELF5 may suppress the estrogen sensitivity of breast cancers. To test this we constructed inducible models of ELF5 expression in ER positive luminal breast cancer cells and interrogated them using transcript profiling and chromatin immunoprecipitation of DNA followed by DNA sequencing (ChIP-Seq). ELF5 suppressed ER and FOXA1 expression and broadly suppressed ER-driven patterns of gene expression including sets of genes distinguishing the luminal molecular subtype. Direct transcriptional targets of ELF5, which included FOXA1, EGFR, and MYC, accurately classified a large cohort of breast cancers into their intrinsic molecular subtypes, predicted ER status with high precision, and defined groups with differential prognosis. Knockdown of ELF5 in basal breast cancer cell lines suppressed basal patterns of gene expression and produced a shift in molecular subtype toward the claudin-low and normal-like groups. Luminal breast cancer cells that acquired resistance to the antiestrogen Tamoxifen showed greatly elevated levels of ELF5 and its transcriptional signature, and became dependent on ELF5 for proliferation, compared to the parental cells. Thus ELF5 provides a key transcriptional determinant of breast cancer molecular subtype by suppression of estrogen sensitivity in luminal breast cancer cells and promotion of basal characteristics in basal breast cancer cells, an action that may be utilised to acquire antiestrogen resistance.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Estrogens/pharmacology , Proto-Oncogene Proteins c-ets/metabolism , Animals , Binding Sites , Breast Neoplasms/classification , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , DNA, Neoplasm/metabolism , DNA-Binding Proteins , Female , Gene Expression Regulation, Neoplastic/drug effects , Genome, Human/genetics , Humans , Mice , Models, Biological , Phenotype , Protein Binding/drug effects , Protein Binding/genetics , Proto-Oncogene Proteins c-ets/genetics , Sequence Analysis, DNA , Transcription Factors , Transcription, Genetic/drug effects
10.
Clin Epigenetics ; 15(1): 150, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37705055

ABSTRACT

BACKGROUND: Acute myeloid leukaemia (AML) is a deadly disease characterised by the uncontrolled proliferation of immature myeloid cells within the bone marrow. Altered regulation of DNA methylation is an important epigenetic driver of AML, where the hypoxic bone marrow microenvironment can help facilitate leukaemogenesis. Thus, interactions between epigenetic regulation and hypoxia signalling will have important implications for AML development and treatment. MAIN BODY: This review summarises the importance of DNA methylation and the hypoxic bone marrow microenvironment in the development, progression, and treatment of AML. Here, we focus on the role hypoxia plays on signalling and the subsequent regulation of DNA methylation. Hypoxia is likely to influence DNA methylation through altered metabolic pathways, transcriptional control of epigenetic regulators, and direct effects on the enzymatic activity of epigenetic modifiers. DNA methylation may also prevent activation of hypoxia-responsive genes, demonstrating bidirectional crosstalk between epigenetic regulation and the hypoxic microenvironment. Finally, we consider the clinical implications of these interactions, suggesting that reduced cell cycling within the hypoxic bone marrow may decrease the efficacy of hypomethylating agents. CONCLUSION: Hypoxia is likely to influence AML progression through complex interactions with DNA methylation, where the therapeutic efficacy of hypomethylating agents may be limited within the hypoxic bone marrow. To achieve optimal outcomes for AML patients, future studies should therefore consider co-treatments that can promote cycling of AML cells within the bone marrow or encourage their dissociation from the bone marrow.


Subject(s)
DNA Methylation , Leukemia, Myeloid, Acute , Humans , Epigenesis, Genetic , Leukemia, Myeloid, Acute/genetics , Signal Transduction , Hypoxia/genetics , Tumor Microenvironment
11.
Stem Cells ; 29(10): 1611-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21823211

ABSTRACT

Recent characterization of mammary stem and progenitor cells has improved our understanding of the transcriptional network that coordinates mammary development; however, little is known about the mechanisms that enforce lineage commitment and prevent transdifferentiation in the mammary gland. The E-twenty six transcription factor Elf5 forces the differentiation of mammary luminal progenitor cells to establish the milk producing alveolar lineage. Methylation of the Elf5 promoter has been proposed to act as a lineage gatekeeper during embryonic development. We used bisulphite sequencing to investigate in detail whether Elf5 promoter methylation plays a role in lineage commitment during mammary development. An increase in Elf5 expression was associated with decreasing Elf5 promoter methylation in differentiating HC11 mammary cells. Similarly, purified mammary epithelial cells from mice had increased Elf5 expression and decreased promoter methylation during pregnancy. Finally, analysis of epithelial subpopulations revealed that the Elf5 promoter is methylated and silenced in the basal, stem cell-containing population relative to luminal cells. These results demonstrate that Elf5 promoter methylation is lineage-specific and developmentally regulated in the mammary gland in vivo, and suggest that loss of Elf5 methylation specifies the mammary luminal lineage, while continued Elf5 methylation maintains the stem cell and myoepithelial lineages.


Subject(s)
Cell Lineage , DNA Methylation , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Promoter Regions, Genetic , Transcription Factors/metabolism , Animals , Cell Differentiation , DNA-Binding Proteins/genetics , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Developmental , Male , Mammary Glands, Animal/cytology , Mice , Pregnancy , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sequence Analysis, DNA , Stem Cells/cytology , Transcription Factors/genetics , Transfection
12.
Hemasphere ; 6(6): e734, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35651714

ABSTRACT

Drug resistance and treatment failure in pediatric acute lymphoblastic leukemia (ALL) are in part driven by tumor heterogeneity and clonal evolution. Although bulk tumor genomic analyses have provided some insight into these processes, single-cell sequencing has emerged as a powerful technique to profile individual cells in unprecedented detail. Since the introduction of single-cell RNA sequencing, we now have the capability to capture not only transcriptomic, but also genomic, epigenetic, and proteomic variation between single cells separately and in combination. This rapidly evolving field has the potential to transform our understanding of the fundamental biology of pediatric ALL and guide the management of ALL patients to improve their clinical outcome. Here, we discuss the impact single-cell sequencing has had on our understanding of tumor heterogeneity and clonal evolution in ALL and provide examples of how single-cell technology can be integrated into the clinic to inform treatment decisions for children with high-risk disease.

13.
Sci Rep ; 12(1): 5776, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35388081

ABSTRACT

Global changes in DNA methylation are observed in development and disease, and single-cell analyses are highlighting the heterogeneous regulation of these processes. However, technical challenges associated with single-cell analysis of DNA methylation limit these studies. We present single-cell transposable element methylation sequencing (scTEM-seq) for cost-effective estimation of average DNA methylation levels. By targeting high-copy SINE Alu elements, we achieve amplicon bisulphite sequencing with thousands of loci covered in each scTEM-seq library. Parallel transcriptome analysis is also performed to link global DNA methylation estimates with gene expression. We apply scTEM-seq to KG1a acute myeloid leukaemia (AML) cells, and primary AML cells. Our method reveals global DNA methylation heterogeneity induced by decitabine treatment of KG1a cells associated with altered expression of immune process genes. We also compare global DNA methylation estimates to expression of transposable elements and find a predominance of negative correlations. Finally, we observe co-ordinated upregulation of many transposable elements in a sub-set of decitabine treated cells. By linking global DNA methylation heterogeneity with transcription, scTEM-seq will refine our understanding of epigenetic regulation in cancer and beyond.


Subject(s)
DNA Transposable Elements , Leukemia, Myeloid, Acute , DNA Methylation , DNA Transposable Elements/genetics , Decitabine/pharmacology , Epigenesis, Genetic , Humans , Leukemia, Myeloid, Acute/genetics , Single-Cell Analysis
14.
Epigenetics Chromatin ; 15(1): 26, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35843975

ABSTRACT

Embryonic development is dependent on the maternal supply of proteins through the oocyte, including factors setting up the adequate epigenetic patterning of the zygotic genome. We previously reported that one such factor is the epigenetic repressor SMCHD1, whose maternal supply controls autosomal imprinted expression in mouse preimplantation embryos and mid-gestation placenta. In mouse preimplantation embryos, X chromosome inactivation is also an imprinted process. Combining genomics and imaging, we show that maternal SMCHD1 is required not only for the imprinted expression of Xist in preimplantation embryos, but also for the efficient silencing of the inactive X in both the preimplantation embryo and mid-gestation placenta. These results expand the role of SMCHD1 in enforcing the silencing of Polycomb targets. The inability of zygotic SMCHD1 to fully restore imprinted X inactivation further points to maternal SMCHD1's role in setting up the appropriate chromatin environment during preimplantation development, a critical window of epigenetic remodelling.


Subject(s)
Chromosomal Proteins, Non-Histone , RNA, Long Noncoding , X Chromosome Inactivation , Animals , Blastocyst/physiology , Chromatin/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Embryonic Development , Genomic Imprinting , Mice , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , X Chromosome
15.
Funct Integr Genomics ; 10(1): 87-95, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19830464

ABSTRACT

Murine milk protein gene expression requires insulin, hydrocortisone, and prolactin; however, the role of insulin is not well understood. This study, therefore, examined the requirement of insulin for milk protein synthesis. Mammary explants were cultured in various combinations of the lactogenic hormones and global changes in gene expression analysed using Affymetrix microarray. The expression of 164 genes was responsive to insulin, and 18 were involved in protein synthesis at the level of transcription and posttranscription, as well as amino acid uptake and metabolism. The folate receptor gene was increased by fivefold, highlighting a potentially important role for the hormone in folate metabolism, a process that is emerging to be central for protein synthesis. Interestingly, gene expression of two milk protein transcription factors, Stat5a and Elf5, previously identified as key components of prolactin signalling, both showed an essential requirement for insulin. Subsequent experiments in HCll cells confirmed that Stat5a and Elf5 gene expression could be induced in the absence of prolactin but in the presence of insulin. Whereas prolactin plays an essential role in phosphorylating and activating Stat5a, gene expression is only induced when insulin is present. This indicates insulin plays a crucial role in the transcription of the milk protein genes.


Subject(s)
Insulin/pharmacology , Lactation/metabolism , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Milk Proteins/biosynthesis , Protein Biosynthesis/drug effects , Tissue Culture Techniques , Animals , Cattle , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Lactation/drug effects , Mice , Models, Genetic , Oligonucleotide Array Sequence Analysis , Protein Biosynthesis/genetics , Reproducibility of Results , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Cancers (Basel) ; 12(11)2020 Oct 26.
Article in English | MEDLINE | ID: mdl-33114584

ABSTRACT

Myelodysplastic syndrome (MDS) is a malignancy that disrupts normal blood cell production and commonly affects our ageing population. MDS patients are diagnosed using an invasive bone marrow biopsy and high-risk MDS patients are treated with hypomethylating agents (HMAs) such as decitabine and azacytidine. However, these therapies are only effective in 50% of patients, and many develop resistance to therapy, often resulting in bone marrow failure or leukemic transformation. Therefore, there is a strong need for less invasive, diagnostic tests for MDS, novel markers that can predict response to therapy and/or patient prognosis to aid treatment stratification, as well as new and effective therapeutics to enhance patient quality of life and survival. Epigenetic modifiers such as DNA methylation, long non-coding RNAs (lncRNAs) and micro-RNAs (miRNAs) are perturbed in MDS blasts and the bone marrow micro-environment, influencing disease progression and response to therapy. This review focusses on the potential utility of epigenetic modifiers in aiding diagnosis, prognosis, and predicting treatment response in MDS, and touches on the need for extensive and collaborative research using single-cell technologies and multi-omics to test the clinical utility of epigenetic markers for MDS patients in the future.

17.
Epigenomics ; 12(13): 1139-1151, 2020 07.
Article in English | MEDLINE | ID: mdl-32790506

ABSTRACT

Cancer is a disease of global epigenetic dysregulation. Mutations in epigenetic regulators are common events in multiple cancer types and epigenetic therapies are emerging as a treatment option in several malignancies. A major challenge for the clinical management of cancer is the heterogeneous nature of this disease. Cancers are composed of numerous cell types and evolve over time. This heterogeneity confounds decisions regarding treatment and promotes disease relapse. The emergence of single-cell epigenomic technologies has introduced the exciting possibility of linking genetic and transcriptional heterogeneity in the context of cancer biology. The next challenge is to leverage these tools for improved patient outcomes. Here we consider how single-cell epigenomic technologies may address the current challenges faced by cancer clinicians.


Subject(s)
Epigenesis, Genetic , Epigenomics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Single-Cell Analysis , Epigenome , High-Throughput Nucleotide Sequencing , Humans , Neoplasm Metastasis , Neoplasms/therapy
18.
Elife ; 92020 11 13.
Article in English | MEDLINE | ID: mdl-33186096

ABSTRACT

Genomic imprinting establishes parental allele-biased expression of a suite of mammalian genes based on parent-of-origin specific epigenetic marks. These marks are under the control of maternal effect proteins supplied in the oocyte. Here we report epigenetic repressor Smchd1 as a novel maternal effect gene that regulates the imprinted expression of ten genes in mice. We also found zygotic SMCHD1 had a dose-dependent effect on the imprinted expression of seven genes. Together, zygotic and maternal SMCHD1 regulate three classic imprinted clusters and eight other genes, including non-canonical imprinted genes. Interestingly, the loss of maternal SMCHD1 does not alter germline DNA methylation imprints pre-implantation or later in gestation. Instead, what appears to unite most imprinted genes sensitive to SMCHD1 is their reliance on polycomb-mediated methylation as germline or secondary imprints, therefore we propose that SMCHD1 acts downstream of polycomb imprints to mediate its function.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Genomic Imprinting/genetics , Animals , Blastocyst , Chromosomal Proteins, Non-Histone/genetics , DNA Methylation , Embryo, Mammalian/metabolism , Female , Gene Expression Regulation, Developmental/physiology , Genotype , Green Fluorescent Proteins , Male , Mice , Neural Stem Cells
19.
Cancer Cell ; 36(6): 660-673.e11, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31821784

ABSTRACT

Inhibition of the Menin (MEN1) and MLL (MLL1, KMT2A) interaction is a potential therapeutic strategy for MLL-rearranged (MLL-r) leukemia. Structure-based design yielded the potent, highly selective, and orally bioavailable small-molecule inhibitor VTP50469. Cell lines carrying MLL rearrangements were selectively responsive to VTP50469. VTP50469 displaced Menin from protein complexes and inhibited chromatin occupancy of MLL at select genes. Loss of MLL binding led to changes in gene expression, differentiation, and apoptosis. Patient-derived xenograft (PDX) models derived from patients with either MLL-r acute myeloid leukemia or MLL-r acute lymphoblastic leukemia (ALL) showed dramatic reductions of leukemia burden when treated with VTP50469. Multiple mice engrafted with MLL-r ALL remained disease free for more than 1 year after treatment. These data support rapid translation of this approach to clinical trials.


Subject(s)
Chromatin/drug effects , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Proto-Oncogene Proteins/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chromatin/genetics , Gene Expression Regulation, Leukemic/genetics , Gene Rearrangement/drug effects , Gene Rearrangement/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Proto-Oncogene Proteins/genetics , Transcription Factors/drug effects , Transcription Factors/genetics
20.
Biochem J ; 404(1): 141-9, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17212589

ABSTRACT

The calcium-sensing receptor (CaR) mediates feedback control of Ca2+o (extracellular Ca2+) concentration. Although the mechanisms are not fully understood, the CaR couples to several important intracellular signalling enzymes, including PI-PLC (phosphoinositide-specific phospholipase C), leading to Ca2+i (intracellular Ca2+) mobilization, and ERK1/2 (extracellular-signal-regulated kinase 1/2). In addition to Ca2+o, the CaR is activated allosterically by several subclasses of L-amino acids, including the aromatics L-phenylalanine and L-tryptophan. These amino acids enhance the Ca2+o-sensitivity of Ca2+i mobilization in CaR-expressing HEK-293 (human embryonic kidney) cells and normal human parathyroid cells. Furthermore, on a background of a physiological fasting serum L-amino acid mixture, they induce a small, but physiologically significant, enhancement of Ca2+o-dependent suppression of PTH (parathyroid hormone) secretion. The impact of amino acids on CaR-stimulated ERK1/2, however, has not been determined. In the present study, we examined the effects of L-amino acids on Ca2+o-stimulated ERK1/2 phosphorylation as determined by Western blotting and a newly developed quantitative assay (SureFire). L-Amino acids induced a small, but significant, enhancement of Ca2+o-stimulated ERK1/2. In CaR-expressing HEK-293 cells, 10 mM L-phenylalanine lowered the EC50 for Ca2+o from approx. 2.3 to 2.0 mM in the Western blot assay and from 3.4 to 2.9 mM in the SureFire assay. The effect was stereoselective (L>D), and another aromatic amino acid, L-tryptophan, was also effective. The effects of amino acids were investigated further in HEK-293 cells that expressed the CaR mutant S169T. L-Phenylalanine normalized the EC50 for Ca2+o-stimulated Ca2+i mobilization from approx. 12 mM to 5.0 mM and ERK1/2 phosphorylation from approx. 4.6 mM to 2.6 mM. Taken together, the data indicate that L-phenylalanine and other amino acids enhance the Ca2+o-sensitivity of CaR-stimulated ERK1/2 phosphorylation; however, the effect is comparatively small and operates in the form of a fine-tuning mechanism.


Subject(s)
Amino Acids/metabolism , Calcium Channels/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Allosteric Regulation , Amino Acid Substitution , Cell Line , Humans , Kidney , Phenylalanine/metabolism , Phosphorylation , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL