Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
EBioMedicine ; 103: 105096, 2024 May.
Article in English | MEDLINE | ID: mdl-38574408

ABSTRACT

BACKGROUND: Type 2 diabetes (T2D) susceptibility is influenced by genetic and environmental factors. Previous findings suggest DNA methylation as a potential mechanism in T2D pathogenesis and progression. METHODS: We profiled DNA methylation in 248 blood samples from participants of European ancestry from 7 twin cohorts using a methylation sequencing platform targeting regulatory genomic regions encompassing 2,048,698 CpG sites. FINDINGS: We find and replicate 3 previously unreported T2D differentially methylated CpG positions (T2D-DMPs) at FDR 5% in RGL3, NGB and OTX2, and 20 signals at FDR 25%, of which 14 replicated. Integrating genetic variation and T2D-discordant monozygotic twin analyses, we identify both genetic-based and genetic-independent T2D-DMPs. The signals annotate to genes with established GWAS and EWAS links to T2D and its complications, including blood pressure (RGL3) and eye disease (OTX2). INTERPRETATION: The results help to improve our understanding of T2D disease pathogenesis and progression and may provide biomarkers for its complications. FUNDING: Funding acknowledgements for each cohort can be found in the Supplementary Note.


Subject(s)
CpG Islands , DNA Methylation , Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/genetics , Female , Male , Genome-Wide Association Study , Genetic Predisposition to Disease , Middle Aged , Epigenesis, Genetic , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , Diabetes Complications/genetics , Gene Expression Profiling
2.
JCI Insight ; 9(6)2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38319712

ABSTRACT

Dedifferentiation or phenotype switching refers to the transition from a proliferative to an invasive cellular state. We previously identified a 122-gene epigenetic gene signature that classifies primary melanomas as low versus high risk (denoted as Epgn1 or Epgn3). We found that the transcriptomes of the Epgn1 low-risk and Epgn3 high-risk cells are similar to the proliferative and invasive cellular states, respectively. These signatures were further validated in melanoma tumor samples. Examination of the chromatin landscape revealed differential H3K27 acetylation in the Epgn1 low-risk versus Epgn3 high-risk cell lines that corroborated with a differential super-enhancer and enhancer landscape. Melanocytic lineage genes (MITF, its targets and regulators) were associated with super-enhancers in the Epgn1 low-risk state, whereas invasiveness genes were linked with Epgn3 high-risk status. We identified the ITGA3 gene as marked by a super-enhancer element in the Epgn3 invasive cells. Silencing of ITGA3 enhanced invasiveness in both in vitro and in vivo systems, suggesting it as a negative regulator of invasion. In conclusion, we define chromatin landscape changes associated with Epgn1/Epgn3 and phenotype switching during early steps of melanoma progression that regulate transcriptional reprogramming. This super-enhancer and enhancer-driven epigenetic regulatory mechanism resulting in major changes in the transcriptome could be important in future therapeutic targeting efforts.


Subject(s)
Histones , Melanoma , Humans , Histones/genetics , Histones/metabolism , Melanoma/pathology , Cell Dedifferentiation/genetics , Acetylation , Cell Line, Tumor , Chromatin/genetics
3.
J Am Chem Soc ; 145(27): 14932-14944, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37365684

ABSTRACT

With advances in chemically induced proximity technologies, heterobifunctional modalities such as proteolysis targeting chimeras (PROTACs) have been successfully advanced to clinics for treating cancer. However, pharmacologic activation of tumor-suppressor proteins for cancer treatment remains a major challenge. Here, we present a novel Acetylation Targeting Chimera (AceTAC) strategy to acetylate the p53 tumor suppressor protein. We discovered and characterized the first p53Y220C AceTAC, MS78, which recruits histone acetyltransferase p300/CBP to acetylate the p53Y220C mutant. MS78 effectively acetylated p53Y220C lysine 382 (K382) in a concentration-, time-, and p300-dependent manner and suppressed proliferation and clonogenicity of cancer cells harboring the p53Y220C mutation with little toxicity in cancer cells with wild-type p53. RNA-seq studies revealed novel p53Y220C-dependent upregulation of TRAIL apoptotic genes and downregulation of DNA damage response pathways upon acetylation induced by MS78. Altogether, the AceTAC strategy could provide a generalizable platform for targeting proteins, such as tumor suppressors, via acetylation.


Subject(s)
Tumor Suppressor Protein p53 , Acetylation , Humans , Cell Line, Tumor , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Mutation , Models, Molecular , Protein Processing, Post-Translational , Protein Structure, Tertiary
5.
Pigment Cell Melanoma Res ; 36(5): 407-415, 2023 09.
Article in English | MEDLINE | ID: mdl-37086018

ABSTRACT

In melanoma, immune cell infiltration into the tumor is associated with better patient outcomes and response to immunotherapy. T-cell non-inflamed tumors (cold tumors) are associated with tumor cell-intrinsic Wnt/ß-catenin activation, and are typically resistant to anti-PD-1 alone or in combination with anti-CTLA-4 therapy. Reversal of the 'cold tumor' phenotype and identifying new effective immunotherapies are challenges. We sought to investigate the role of a newer immunotherapy agent, B7-H3, in this setting. RNA sequencing was used to identify co-targeting strategies upon B7-H3 inhibition in a well-defined preclinical melanoma model driven by ß-catenin. We found that immune checkpoint molecule B7-H3 confers a suppressive tumor microenvironment by modulating antiviral signals and innate immunity. B7-H3 inhibition led to an inflamed microenvironment, up-regulation of CD47/SIRPa signaling, and together with blockade of the macrophage checkpoint CD47 resulted in additive antitumor responses. We found that the antitumor effects of the B7-H3/CD47 antibody combination were dependent on cytokine signaling pathways (CCR5/CCL5 and IL4).


Subject(s)
Melanoma , Humans , B7-H1 Antigen , beta Catenin , CD47 Antigen , Immunosuppression Therapy , Immunotherapy/methods , Melanoma/therapy , Tumor Microenvironment
6.
Exp Hematol ; 122: 41-54, 2023 06.
Article in English | MEDLINE | ID: mdl-37001723

ABSTRACT

The regenerative potential of human hematopoietic stem cells (HSCs) is functionally defined by their ability to provide life-long blood cell production and to repopulate myeloablated allogeneic transplant recipients. The expansion of HSC numbers is dependent not only on HSC divisions but also on a coordinated adaptation of HSCs to metabolic stress. These variables are especially critical during the ex vivo culture of HSCs with cytokine combinations, which frequently results in HSC exhaustion. We have previously reported that human CD34+ hematopoietic stem and progenitor cells (HSPCs) can be efficiently reprogrammed ex vivo and that the number of phenotypic HSCs with long-term repopulation capacity is expanded in the presence of a combination of cytokines and an epigenetic modifier. Here, we present evidence that ex vivo HSC reprogramming and maintenance is accompanied by increased transcripts of genes regulating metabolic integrity, including SIRT1 and SIRT3.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Humans , Hematopoietic Stem Cells/metabolism , Cytokines/metabolism , Antigens, CD34/metabolism , Fetal Blood , Cells, Cultured
7.
Thyroid ; 33(3): 301-311, 2023 03.
Article in English | MEDLINE | ID: mdl-36719767

ABSTRACT

Background: Thyroid hormones play a key role in differentiation and metabolism and are known regulators of gene expression through both genomic and epigenetic processes including DNA methylation. The aim of this study was to examine associations between thyroid hormones and DNA methylation. Methods: We carried out a fixed-effect meta-analysis of epigenome-wide association study (EWAS) of blood DNA methylation sites from 8 cohorts from the ThyroidOmics Consortium, incorporating up to 7073 participants of both European and African ancestry, implementing a discovery and replication stage. Statistical analyses were conducted using normalized beta CpG values as dependent and log-transformed thyrotropin (TSH), free thyroxine, and free triiodothyronine levels, respectively, as independent variable in a linear model. The replicated findings were correlated with gene expression levels in whole blood and tested for causal influence of TSH and free thyroxine by two-sample Mendelian randomization (MR). Results: Epigenome-wide significant associations (p-value <1.1E-7) of three CpGs for free thyroxine, five for free triiodothyronine, and two for TSH concentrations were discovered and replicated (combined p-values = 1.5E-9 to 4.3E-28). The associations included CpG sites annotated to KLF9 (cg00049440) and DOT1L (cg04173586) that overlap with all three traits, consistent with hypothalamic-pituitary-thyroid axis physiology. Significant associations were also found for CpGs in FKBP5 for free thyroxine, and at CSNK1D/LINCO1970 and LRRC8D for free triiodothyronine. MR analyses supported a causal effect of thyroid status on DNA methylation of KLF9. DNA methylation of cg00049440 in KLF9 was inversely correlated with KLF9 gene expression in blood. The CpG at CSNK1D/LINC01970 overlapped with thyroid hormone receptor alpha binding peaks in liver cells. The total additive heritability of the methylation levels of the six significant CpG sites was between 25% and 57%. Significant methylation QTLs were identified for CpGs at KLF9, FKBP5, LRRC8D, and CSNK1D/LINC01970. Conclusions: We report novel associations between TSH, thyroid hormones, and blood-based DNA methylation. This study advances our understanding of thyroid hormone action particularly related to KLF9 and serves as a proof-of-concept that integrations of EWAS with other -omics data can provide a valuable tool for unraveling thyroid hormone signaling in humans by complementing and feeding classical in vitro and animal studies.


Subject(s)
Epigenome , Triiodothyronine , Humans , Thyroid Gland , Thyroxine/genetics , CpG Islands , Genome-Wide Association Study , Kruppel-Like Transcription Factors/genetics
8.
Nat Commun ; 13(1): 6041, 2022 10 17.
Article in English | MEDLINE | ID: mdl-36253360

ABSTRACT

Tumors exhibit enhancer reprogramming compared to normal tissue. The etiology is largely attributed to cell-intrinsic genomic alterations. Here, using freshly resected primary CRC tumors and patient-matched adjacent normal colon, we find divergent epigenetic landscapes between CRC tumors and cell lines. Intriguingly, this phenomenon extends to highly recurrent aberrant super-enhancers gained in CRC over normal. We find one such super-enhancer activated in epithelial cancer cells due to surrounding inflammation in the tumor microenvironment. We restore this super-enhancer and its expressed gene, PDZK1IP1, following treatment with cytokines or xenotransplantation into nude mice, thus demonstrating cell-extrinsic etiology. We demonstrate mechanistically that PDZK1IP1 enhances the reductive capacity CRC cancer cells via the pentose phosphate pathway. We show this activation enables efficient growth under oxidative conditions, challenging the previous notion that PDZK1IP1 acts as a tumor suppressor in CRC. Collectively, these observations highlight the significance of epigenomic profiling on primary specimens.


Subject(s)
Colorectal Neoplasms , Tumor Microenvironment , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Cytokines/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic , Mice , Mice, Nude , Tumor Microenvironment/genetics
9.
Cell Rep ; 39(1): 110637, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35385731

ABSTRACT

ARID2 is the most recurrently mutated SWI/SNF complex member in melanoma; however, its tumor-suppressive mechanisms in the context of the chromatin landscape remain to be elucidated. Here, we model ARID2 deficiency in melanoma cells, which results in defective PBAF complex assembly with a concomitant genomic redistribution of the BAF complex. Upon ARID2 depletion, a subset of PBAF and shared BAF-PBAF-occupied regions displays diminished chromatin accessibility and associated gene expression, while BAF-occupied enhancers gain chromatin accessibility and expression of genes linked to the process of invasion. As a function of altered accessibility, the genomic occupancy of melanoma-relevant transcription factors is affected and significantly correlates with the observed transcriptional changes. We further demonstrate that ARID2-deficient cells acquire the ability to colonize distal organs in multiple animal models. Taken together, our results reveal a role for ARID2 in mediating BAF and PBAF subcomplex chromatin dynamics with consequences for melanoma metastasis.


Subject(s)
Chromosomal Proteins, Non-Histone , Melanoma , Transcription Factors , Animals , Chromatin , Chromatin Assembly and Disassembly , Gene Expression Regulation , Humans , Melanoma/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Stud Health Technol Inform ; 289: 65-68, 2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35062093

ABSTRACT

The goal of this study was to build a machine learning model for early prostate cancer prediction based on healthcare utilization patterns. We examined the frequency and pattern changes of healthcare utilization in 2916 prostate cancer patients 3 years prior to their prostate cancer diagnoses and explored several supervised machine learning techniques to predict possible prostate cancer diagnosis. Analysis of patients' medical activities between 1 year and 2 years prior to their prostate cancer diagnoses using XGBoost model provided the best prediction accuracy with high F1 score (0.9) and AUC score (0.73). These pilot results indicated that application of machine learning to healthcare utilization patterns may result in early identification of prostate cancer diagnosis.


Subject(s)
Machine Learning , Prostatic Neoplasms , Humans , Male , Patient Acceptance of Health Care , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/therapy , Supervised Machine Learning
11.
Cancer Discov ; 11(12): 3064-3089, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34301793

ABSTRACT

Using a panel of cancer cell lines, we characterized a novel degrader of AKT, MS21. In mutant PI3K-PTEN pathway cell lines, AKT degradation was superior to AKT kinase inhibition for reducing cell growth and sustaining lower signaling over many days. AKT degradation, but not kinase inhibition, profoundly lowered Aurora kinase B (AURKB) protein, which is known to be essential for cell division, and induced G2-M arrest and hyperploidy. PI3K activated AKT phosphorylation of AURKB on threonine 73, which protected it from proteasome degradation. A mutant of AURKB (T73E) that mimics phosphorylation and blocks degradation rescued cells from growth inhibition. Degrader-resistant lines were associated with low AKT phosphorylation, wild-type PI3K/PTEN status, and mutation of KRAS/BRAF. Pan-cancer analysis identified that 19% of cases have PI3K-PTEN pathway mutation without RAS pathway mutation, suggesting that these patients with cancer could benefit from AKT degrader therapy that leads to loss of AURKB. SIGNIFICANCE: MS21 depletes cells of phosphorylated AKT (pAKT) and a newly identified AKT substrate, AURKB, to inhibit tumor growth in mice. MS21 is superior to prior agents that target PI3K and AKT due to its ability to selectively target active, pAKT and sustain repression of signaling to deplete AURKB. This article is highlighted in the In This Issue feature, p. 2945.


Subject(s)
Neoplasms , Proto-Oncogene Proteins c-akt , Animals , Apoptosis/genetics , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints , Humans , Mice , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
12.
Commun Biol ; 4(1): 312, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33750924

ABSTRACT

Downregulation of the PTEN tumor suppressor transcript is frequent in breast cancer and associates with poor prognosis and triple-negative breast cancer (TNBC) when comparing breast cancers to one another. Here we show that in almost all cases, when comparing breast tumors to adjacent normal ducts, PTEN expression is decreased and the PRC2-associated methyltransferase EZH2 is increased. We further find that when comparing breast cancer cases in large cohorts, EZH2 inversely correlates with PTEN expression. Within the highest EZH2 expressing group, NOTCH alterations are frequent, and also associate with decreased PTEN expression. We show that repression of PTEN occurs through the combined action of NOTCH (NOTCH1 or NOTCH2) and EZH2 alterations in a subset of breast cancers. In fact, in cases harboring NOTCH1 mutation or a NOTCH2 fusion gene, NOTCH drives EZH2, HES-1, and HEY-1 expression to repress PTEN transcription at the promoter, which may contribute to poor prognosis in this subgroup. Restoration of PTEN expression can be achieved with an EZH2 inhibitor (UNC1999), a γ-secretase inhibitor (Compound E), or knockdown of EZH2 or NOTCH. These findings elucidate a mechanism of transcriptional repression of PTEN induced by NOTCH1 or NOTCH2 alterations, and identifies actionable signaling pathways responsible for driving a large subset of poor-prognosis breast cancers.


Subject(s)
Breast Neoplasms/enzymology , Enhancer of Zeste Homolog 2 Protein/metabolism , PTEN Phosphohydrolase/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Fusion , Humans , Mutation , PTEN Phosphohydrolase/genetics , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Transcription, Genetic
13.
Gastroenterology ; 159(6): 2203-2220.e14, 2020 12.
Article in English | MEDLINE | ID: mdl-32814112

ABSTRACT

BACKGROUND AND AIMS: The pattern of genetic alterations in cancer driver genes in patients with hepatocellular carcinoma (HCC) is highly diverse, which partially explains the low efficacy of available therapies. In spite of this, the existing mouse models only recapitulate a small portion of HCC inter-tumor heterogeneity, limiting the understanding of the disease and the nomination of personalized therapies. Here, we aimed at establishing a novel collection of HCC mouse models that captured human HCC diversity. METHODS: By performing hydrodynamic tail-vein injections, we tested the impact of altering a well-established HCC oncogene (either MYC or ß-catenin) in combination with an additional alteration in one of eleven other genes frequently mutated in HCC. Of the 23 unique pairs of genetic alterations that we interrogated, 9 were able to induce HCC. The established HCC mouse models were characterized at histopathological, immune, and transcriptomic level to identify the unique features of each model. Murine HCC cell lines were generated from each tumor model, characterized transcriptionally, and used to identify specific therapies that were validated in vivo. RESULTS: Cooperation between pairs of driver genes produced HCCs with diverse histopathology, immune microenvironments, transcriptomes, and drug responses. Interestingly, MYC expression levels strongly influenced ß-catenin activity, indicating that inter-tumor heterogeneity emerges not only from specific combinations of genetic alterations but also from the acquisition of expression-dependent phenotypes. CONCLUSIONS: This novel collection of murine HCC models and corresponding cell lines establishes the role of driver genes in diverse contexts and enables mechanistic and translational studies.


Subject(s)
Carcinoma, Hepatocellular/genetics , Genetic Heterogeneity , Proto-Oncogenes/genetics , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Computational Biology , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Male , Mice , Mice, Transgenic , Tumor Escape/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
14.
Cells ; 9(3)2020 03 09.
Article in English | MEDLINE | ID: mdl-32182948

ABSTRACT

The pathogenesis of autoimmune thyroid diseases (AITD) is poorly understood and the association between different immune features and the germline variants involved in AITD are yet unclear. We previously observed systemic depletion of IgG core fucosylation and antennary α1,2 fucosylation in peripheral blood mononuclear cells in AITD, correlated with anti-thyroid peroxidase antibody (TPOAb) levels. Fucose depletion is known to potentiate strong antibody-mediated NK cell activation and enhanced target antigen-expressing cell killing. In autoimmunity, this may translate to autoantibody-mediated immune cell recruitment and attack of self-antigen expressing normal tissues. Hence, we investigated the crosstalk between immune cell traits, secreted proteins, genetic variants and the glycosylation patterns of serum IgG, in a multi-omic and cross-sectional study of 622 individuals from the TwinsUK cohort, 172 of whom were diagnosed with AITD. We observed associations between two genetic variants (rs505922 and rs687621), AITD status, the secretion of Desmoglein-2 protein, and the profile of two IgG N-glycan traits in AITD, but further studies need to be performed to better understand their crosstalk in AITD. On the other side, enhanced afucosylated IgG was positively associated with activatory CD335- CD314+ CD158b+ NK cell subsets. Increased levels of the apoptosis and inflammation markers Caspase-2 and Interleukin-1α positively associated with AITD. Two genetic variants associated with AITD, rs1521 and rs3094228, were also associated with altered expression of the thyrocyte-expressed ligands known to recognize the NK cell immunoreceptors CD314 and CD158b. Our analyses reveal a combination of heightened Fc-active IgG antibodies, effector cells, cytokines and apoptotic signals in AITD, and AITD genetic variants associated with altered expression of thyrocyte-expressed ligands to NK cell immunoreceptors. Together, TPOAb responses, dysregulated immune features, germline variants associated with immunoactivity profiles, are consistent with a positive autoreactive antibody-dependent NK cell-mediated immune response likely drawn to the thyroid gland in AITD.


Subject(s)
Autoantibodies/metabolism , Iodide Peroxidase/metabolism , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/metabolism , Thyroid Diseases/metabolism , Autoantibodies/immunology , Cross-Sectional Studies , Fucose/immunology , Fucose/metabolism , Humans , Iodide Peroxidase/genetics , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/immunology , Thyroid Diseases/immunology
15.
Mol Cell Proteomics ; 19(5): 774-792, 2020 05.
Article in English | MEDLINE | ID: mdl-32024769

ABSTRACT

Autoimmune thyroid diseases (AITD) are the most common group of autoimmune diseases, associated with lymphocyte infiltration and the production of thyroid autoantibodies, like thyroid peroxidase antibodies (TPOAb), in the thyroid gland. Immunoglobulins and cell-surface receptors are glycoproteins with distinctive glycosylation patterns that play a structural role in maintaining and modulating their functions. We investigated associations of total circulating IgG and peripheral blood mononuclear cells glycosylation with AITD and the influence of genetic background in a case-control study with several independent cohorts and over 3,000 individuals in total. The study revealed an inverse association of IgG core fucosylation with TPOAb and AITD, as well as decreased peripheral blood mononuclear cells antennary α1,2 fucosylation in AITD, but no shared genetic variance between AITD and glycosylation. These data suggest that the decreased level of IgG core fucosylation is a risk factor for AITD that promotes antibody-dependent cell-mediated cytotoxicity previously associated with TPOAb levels.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Autoimmune Diseases/immunology , Fucose/metabolism , Immunoglobulin G/metabolism , Thyroid Diseases/immunology , Adult , Blood Cells/metabolism , Cohort Studies , Gene Expression Regulation , Glycomics , Glycosylation , Humans , Immunoglobulin G/genetics , Iodide Peroxidase/immunology , Linkage Disequilibrium/genetics , Models, Biological , Polymorphism, Single Nucleotide/genetics , Polysaccharides/metabolism
16.
Front Cell Dev Biol ; 8: 592348, 2020.
Article in English | MEDLINE | ID: mdl-33384995

ABSTRACT

Ex vivo expansion strategies of human hematopoietic stem cell (HSC) grafts with suboptimal stem cell dose have emerged as promising strategies for improving outcomes of HSC transplantation in patients with hematological malignancies. While exposure of HSCs to ex vivo cultures expands the number of phenotypically identifiable HSCs, it frequently alters the transcriptomic and metabolic profiles, therefore, compromising their long-term (LT) hematopoietic reconstitution capacity. Within the heterogeneous pool of expanded HSCs, the precise phenotypic, transcriptomic and metabolic profile and thus, the identity of HSCs that confer LT repopulation potential remains poorly described. Utilizing valproic acid (VPA) in ex vivo cultures of umbilical cord blood (UCB)-CD34+ cells, we demonstrate that expanded HSCs phenotypically marked by expression of the stem cell markers CD34, CD90 and EPCR (CD201) are highly enriched for LT-HSCs. Furthermore, we report that low mitochondrial membrane potential, and, hence, mitochondrial activity distinguishes LT-HSCs within the expanded pool of phenotypically defined HSCs. Remarkably, such reduced mitochondrial activity is restricted to cells with the highest expression levels of CD34, CD90 and EPCR phenotypic markers. Together, our findings reveal that high expression of CD34, CD90 and EPCR in conjunction with low mitochondrial activity is critical for identification of functional LT-HSCs generated within ex vivo expansion cultures.

17.
Nat Chem Biol ; 16(2): 214-222, 2020 02.
Article in English | MEDLINE | ID: mdl-31819273

ABSTRACT

The enhancer of zeste homolog 2 (EZH2) is the main enzymatic subunit of the PRC2 complex, which catalyzes trimethylation of histone H3 lysine 27 (H3K27me3) to promote transcriptional silencing. EZH2 is overexpressed in multiple types of cancer including triple-negative breast cancer (TNBC), and high expression levels correlate with poor prognosis. Several EZH2 inhibitors, which inhibit the methyltransferase activity of EZH2, have shown promise in treating sarcoma and follicular lymphoma in clinics. However, EZH2 inhibitors are ineffective at blocking proliferation of TNBC cells, even though they effectively reduce the H3K27me3 mark. Using a hydrophobic tagging approach, we generated MS1943, a first-in-class EZH2 selective degrader that effectively reduces EZH2 levels in cells. Importantly, MS1943 has a profound cytotoxic effect in multiple TNBC cells, while sparing normal cells, and is efficacious in vivo, suggesting that pharmacologic degradation of EZH2 can be advantageous for treating the cancers that are dependent on EZH2.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Enhancer of Zeste Homolog 2 Protein/metabolism , Piperazines/pharmacology , Pyridines/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Cell Death/drug effects , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Gene Knockout Techniques , Humans , Hydrophobic and Hydrophilic Interactions , Male , Mice , Mice, Inbred BALB C , Molecular Targeted Therapy , Proteolysis/drug effects , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Unfolded Protein Response/drug effects , Xenograft Model Antitumor Assays
18.
Nat Commun ; 10(1): 4505, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31582752

ABSTRACT

The human gut is inhabited by a complex and metabolically active microbial ecosystem. While many studies focused on the effect of individual microbial taxa on human health, their overall metabolic potential has been under-explored. Using whole-metagenome shotgun sequencing data in 1,004 twins, we first observed that unrelated subjects share, on average, almost double the number of metabolic pathways (82%) than species (43%). Then, using 673 blood and 713 faecal metabolites, we found metabolic pathways to be associated with 34% of blood and 95% of faecal metabolites, with over 18,000 significant associations, while species showed less than 3,000 associations. Finally, we estimated that the microbiome was involved in a dialogue between 71% of faecal, and 15% of blood, metabolites. This study underlines the importance of studying the microbial metabolic potential rather than focusing purely on taxonomy to find therapeutic and diagnostic targets, and provides a unique resource describing the interplay between the microbiome and the systemic and faecal metabolic environments.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome/physiology , Host Microbial Interactions/physiology , Metabolic Networks and Pathways/physiology , Metabolome/physiology , Age Distribution , Age Factors , Aged , Aged, 80 and over , Bacteria/isolation & purification , Biomarkers/blood , Biomarkers/metabolism , Datasets as Topic , Feces/microbiology , Female , Humans , Male , Metabolomics/methods , Metagenome , Middle Aged , Whole Genome Sequencing
19.
Nat Commun ; 10(1): 2581, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31197173

ABSTRACT

Despite existing reports on differential DNA methylation in type 2 diabetes (T2D) and obesity, our understanding of its functional relevance remains limited. Here we show the effect of differential methylation in the early phases of T2D pathology by a blood-based epigenome-wide association study of 4808 non-diabetic Europeans in the discovery phase and 11,750 individuals in the replication. We identify CpGs in LETM1, RBM20, IRS2, MAN2A2 and the 1q25.3 region associated with fasting insulin, and in FCRL6, SLAMF1, APOBEC3H and the 15q26.1 region with fasting glucose. In silico cross-omics analyses highlight the role of differential methylation in the crosstalk between the adaptive immune system and glucose homeostasis. The differential methylation explains at least 16.9% of the association between obesity and insulin. Our study sheds light on the biological interactions between genetic variants driving differential methylation and gene expression in the early pathogenesis of T2D.


Subject(s)
DNA Methylation/physiology , Diabetes Mellitus, Type 2/genetics , Glucose/metabolism , Insulin/metabolism , Obesity/genetics , Adult , Aged , Aged, 80 and over , Computer Simulation , CpG Islands/genetics , Diabetes Mellitus, Type 2/metabolism , Epigenesis, Genetic/physiology , Epigenomics/methods , Female , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Genome-Wide Association Study/methods , Homeostasis/genetics , Humans , Male , Metabolic Networks and Pathways/genetics , Middle Aged , Obesity/metabolism , Polymorphism, Single Nucleotide/physiology , Young Adult
20.
Clin Epigenetics ; 10(1): 126, 2018 10 20.
Article in English | MEDLINE | ID: mdl-30342560

ABSTRACT

BACKGROUND: Tobacco smoking is a risk factor for multiple diseases, including cardiovascular disease and diabetes. Many smoking-associated signals have been detected in the blood methylome, but the extent to which these changes are widespread to metabolically relevant tissues, and impact gene expression or metabolic health, remains unclear. METHODS: We investigated smoking-associated DNA methylation and gene expression variation in adipose tissue biopsies from 542 healthy female twins. Replication, tissue specificity, and longitudinal stability of the smoking-associated effects were explored in additional adipose, blood, skin, and lung samples. We characterized the impact of adipose tissue smoking methylation and expression signals on metabolic disease risk phenotypes, including visceral fat. RESULTS: We identified 42 smoking-methylation and 42 smoking-expression signals, where five genes (AHRR, CYP1A1, CYP1B1, CYTL1, F2RL3) were both hypo-methylated and upregulated in current smokers. CYP1A1 gene expression achieved 95% prediction performance of current smoking status. We validated and replicated a proportion of the signals in additional primary tissue samples, identifying tissue-shared effects. Smoking leaves systemic imprints on DNA methylation after smoking cessation, with stronger but shorter-lived effects on gene expression. Metabolic disease risk traits such as visceral fat and android-to-gynoid ratio showed association with methylation at smoking markers with functional impacts on expression, such as CYP1A1, and at tissue-shared smoking signals, such as NOTCH1. At smoking-signals, BHLHE40 and AHRR DNA methylation and gene expression levels in current smokers were predictive of future gain in visceral fat upon smoking cessation. CONCLUSIONS: Our results provide the first comprehensive characterization of coordinated DNA methylation and gene expression markers of smoking in adipose tissue. The findings relate to human metabolic health and give insights into understanding the widespread health consequence of smoking outside of the lung.


Subject(s)
Adipose Tissue/chemistry , DNA Methylation , Gene Expression Profiling/methods , Smoking/genetics , Twins/genetics , Up-Regulation , Adult , Aged , Blood Proteins/genetics , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1B1/genetics , Cytokines/genetics , Epigenesis, Genetic , Female , Gene Expression Regulation , Healthy Volunteers , Humans , Male , Middle Aged , Receptor, Notch1/genetics , Receptors, Thrombin
SELECTION OF CITATIONS
SEARCH DETAIL
...