Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Blood ; 138(15): 1331-1344, 2021 10 14.
Article in English | MEDLINE | ID: mdl-33971010

ABSTRACT

The mechanism underlying cell type-specific gene induction conferred by ubiquitous transcription factors as well as disruptions caused by their chimeric derivatives in leukemia is not well understood. Here, we investigate whether RNAs coordinate with transcription factors to drive myeloid gene transcription. In an integrated genome-wide approach surveying for gene loci exhibiting concurrent RNA and DNA interactions with the broadly expressed Runt-related transcription factor 1 (RUNX1), we identified the long noncoding RNA (lncRNA) originating from the upstream regulatory element of PU.1 (LOUP). This myeloid-specific and polyadenylated lncRNA induces myeloid differentiation and inhibits cell growth, acting as a transcriptional inducer of the myeloid master regulator PU.1. Mechanistically, LOUP recruits RUNX1 to both the PU.1 enhancer and the promoter, leading to the formation of an active chromatin loop. In t(8;21) acute myeloid leukemia (AML), wherein RUNX1 is fused to ETO, the resulting oncogenic fusion protein, RUNX1-ETO, limits chromatin accessibility at the LOUP locus, causing inhibition of LOUP and PU.1 expression. These findings highlight the important role of the interplay between cell-type-specific RNAs and transcription factors, as well as their oncogenic derivatives in modulating lineage-gene activation and raise the possibility that RNA regulators of transcription factors represent alternative targets for therapeutic development.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , RNA, Long Noncoding/genetics , RUNX1 Translocation Partner 1 Protein/genetics , Cell Line, Tumor , Gene Expression Regulation, Leukemic , Humans , Transcriptional Activation
2.
Nucleic Acids Res ; 47(14): 7460-7475, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31219573

ABSTRACT

DNMT3B is known as a de novo DNA methyltransferase. However, its preferential target sites for DNA methylation are largely unknown. Our analysis on ChIP-seq experiment in human embryonic stem cells (hESC) revealed that DNMT3B, mCA and H3K36me3 share the same genomic distribution profile. Deletion of DNMT3B or its histone-interacting domain (PWWP) demolished mCA in hESCs, suggesting that PWWP domain of DNMT3B directs the formation of mCA landscape. In contrast to the common presumption that PWWP guides DNMT3B-mediated mCG deposition, we found that deleting PWWP does not affect the mCG landscape. Nonetheless, DNMT3B knockout led to the formation of 2985 de novo hypomethylated regions at annotated promoter sites. Upon knockout, most of these promoters gain the bivalent marks, H3K4me3 and H3K27me3. We call them spurious bivalent promoters. Gene ontology analysis associated spurious bivalent promoters with development and cell differentiation. Overall, we found the importance of DNMT3B for shaping the mCA landscape and for maintaining the fidelity of the bivalent promoters in hESCs.


Subject(s)
CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Human Embryonic Stem Cells/metabolism , Promoter Regions, Genetic/genetics , Base Sequence , Cell Differentiation/genetics , Cell Line , DNA (Cytosine-5-)-Methyltransferases/metabolism , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Mutation , Protein Binding , DNA Methyltransferase 3B
3.
Glia ; 68(3): 543-560, 2020 03.
Article in English | MEDLINE | ID: mdl-31626368

ABSTRACT

Astrocytes perform important housekeeping functions in the nervous system including maintenance of adequate neuronal excitability, although the regulatory mechanisms are currently poorly understood. The astrocytic Ca2+ /calmodulin-activated phosphatase calcineurin (CaN) is implicated in the development of reactive gliosis and neuroinflammation, but its roles, including the control of neuronal excitability, in healthy brain is unknown. We have generated a mouse line with conditional knockout (KO) of CaN B1 (CaNB1) in glial fibrillary acidic protein-expressing astrocytes (astroglial calcineurin KO [ACN-KO]). Here, we report that postnatal and astrocyte-specific ablation of CaNB1 did not alter normal growth and development as well as adult neurogenesis. Yet, we found that specific deletion of astrocytic CaN selectively impairs intrinsic neuronal excitability in hippocampal CA1 pyramidal neurons and cerebellar granule cells (CGCs). This impairment was associated with a decrease in after hyperpolarization in CGC, while passive properties were unchanged, suggesting impairment of K+ homeostasis. Indeed, blockade of Na+ /K+ -ATPase (NKA) with ouabain phenocopied the electrophysiological alterations observed in ACN-KO CGCs. In addition, NKA activity was significantly lower in cerebellar and hippocampal lysates and in pure astrocytic cultures from ACN-KO mice. While no changes were found in protein levels, NKA activity was inhibited by the specific CaN inhibitor FK506 in both cerebellar lysates and primary astroglia from control mice, suggesting that CaN directly modulates NKA activity and in this manner controls neuronal excitability. In summary, our data provide formal evidence for the notion that astroglia is fundamental for controlling basic neuronal functions and place CaN center-stage as an astrocytic Ca2+ -sensitive switch.


Subject(s)
Astrocytes/metabolism , Calcineurin/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/cytology , Animals , Cells, Cultured , Cerebellum/metabolism , Gliosis/metabolism , Mice, Knockout , Neurons/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
4.
Nature ; 503(7476): 371-6, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24107992

ABSTRACT

DNA methylation was first described almost a century ago; however, the rules governing its establishment and maintenance remain elusive. Here we present data demonstrating that active transcription regulates levels of genomic methylation. We identify a novel RNA arising from the CEBPA gene locus that is critical in regulating the local DNA methylation profile. This RNA binds to DNMT1 and prevents CEBPA gene locus methylation. Deep sequencing of transcripts associated with DNMT1 combined with genome-scale methylation and expression profiling extend the generality of this finding to numerous gene loci. Collectively, these results delineate the nature of DNMT1-RNA interactions and suggest strategies for gene-selective demethylation of therapeutic targets in human diseases.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , Gene Expression Regulation/genetics , RNA, Untranslated/metabolism , Base Sequence , Cell Line , DNA/genetics , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , Gene Expression Profiling , Genome, Human/genetics , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Untranslated/genetics , RNA-Binding Proteins/metabolism , Substrate Specificity , Transcription, Genetic/genetics
5.
J Cell Physiol ; 231(3): 576-86, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26205888

ABSTRACT

INTRODUCTION: IL-6 influences several biological processes, including cardiac stem cell and cardiomyocyte physiology. Although JAK-STAT3 activation is the defining feature of IL-6 signaling, signaling molecules such as PI3K, PKCs, and ERK1/2 are also activated and elicit different responses. Moreover, most studies on the specific role of these signaling molecules focus on the adult heart, and few studies are available on the biological effects evoked by IL-6 in embryonic cardiomyocytes. AIM: The aim of this study was to clarify the biological response of embryonic heart derived cells to IL-6 by analyzing the morphological modifications and the signaling cascades evoked by the cytokine in H9c2 cells. RESULTS: IL-6 stimulation determined the terminal differentiation of H9c2 cells, as evidenced by the increased expression of cardiac transcription factors (NKX2.5 and GATA4), structural proteins (α-myosin heavy chain and cardiac Troponin T) and the gap junction protein Connexin 43. This process was mediated by the rapid modulation of PI3K, Akt, PTEN, and PKCζ phosphorylation levels. PI3K recruitment was an upstream event in the signaling cascade and when PI3K was inhibited, IL-6 failed to modify PKCζ, PTEN, and Akt phosphorylation. Blocking PKCζ activity affected only PTEN and Akt. Finally, the overexpression of a constitutively active form of PKCζ in H9c2 cells largely mimicked the morphological and molecular effects evoked by IL-6. CONCLUSIONS: This study demonstrated that IL-6 induces the cardiac differentiation of H9c2 embryonic cells though a signaling cascade that involves PI3K, PTEN, and PKCζ activities.


Subject(s)
Cell Differentiation/physiology , Interleukin-6/metabolism , Myocytes, Cardiac/cytology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Signal Transduction/physiology , Animals , Cell Line , Enzyme Activation , Myocytes, Cardiac/metabolism , Rats , Ventricular Myosins/metabolism
6.
Blood ; 124(15): 2391-9, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25185713

ABSTRACT

Runx transcription factors contribute to hematopoiesis and are frequently implicated in hematologic malignancies. All three Runx isoforms are expressed at the earliest stages of hematopoiesis; however, their function in hematopoietic stem cells (HSCs) is not fully elucidated. Here, we show that Runx factors are essential in HSCs by driving the expression of the hematopoietic transcription factor PU.1. Mechanistically, by using a knockin mouse model in which all three Runx binding sites in the -14kb enhancer of PU.1 are disrupted, we observed failure to form chromosomal interactions between the PU.1 enhancer and its proximal promoter. Consequently, decreased PU.1 levels resulted in diminished long-term HSC function through HSC exhaustion, which could be rescued by reintroducing a PU.1 transgene. Similarly, in a mouse model of AML/ETO9a leukemia, disrupting the Runx binding sites resulted in decreased PU.1 levels. Leukemia onset was delayed, and limiting dilution transplantation experiments demonstrated functional loss of leukemia-initiating cells. This is surprising, because low PU.1 levels have been considered a hallmark of AML/ETO leukemia, as indicated in mouse models and as shown here in samples from leukemic patients. Our data demonstrate that Runx-dependent PU.1 chromatin interaction and transcription of PU.1 are essential for both normal and leukemia stem cells.


Subject(s)
Core Binding Factor alpha Subunits/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Animals , Base Pairing/genetics , Binding Sites , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice, Inbred C57BL , Mutation/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Signal Transduction/genetics , Transcription, Genetic
7.
Blood ; 121(8): 1255-64, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23212524

ABSTRACT

Lineage-restricted cells can be reprogrammed to a pluripotent state known as induced pluripotent stem (iPS) cells through overexpression of 4 transcription factors. iPS cells are similar to human embryonic stem (hES) cells and have the same ability to generate all the cells of the human body, including blood cells. However, this process is extremely inefficient and to date has been unsuccessful at differentiating iPS into hematopoietic stem cells (HSCs). We hypothesized that iPS cells, injected into NOD.Cg-Prkdc(scid) Il2rg(tm1Wjl)/SzJ immunocompromised (NSG) mice could give rise to hematopoietic stem/progenitor cells (HSPCs) during teratoma formation. Here, we report a novel in vivo system in which human iPS cells differentiate within teratomas to derive functional myeloid and lymphoid cells. Similarly, HSPCs can be isolated from teratoma parenchyma and reconstitute a human immune system when transplanted into immunodeficient mice. Our data provide evidence that in vivo generation of patient customized cells is feasible, providing materials that could be useful for transplantation, human antibody generation, and drug screening applications.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Teratoma/pathology , Animals , B-Lymphocytes/cytology , Cell Differentiation/physiology , Hematopoietic Stem Cells/physiology , Humans , Induced Pluripotent Stem Cells/physiology , Keratinocytes/physiology , Lymphocytes/cytology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Myeloid Cells/cytology , Neoplasm Transplantation , Stromal Cells/cytology , Stromal Cells/physiology , Stromal Cells/transplantation , T-Lymphocytes/cytology , Teratoma/genetics , Transplantation, Heterologous , Tumor Cells, Cultured
8.
Nat Commun ; 14(1): 99, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36609400

ABSTRACT

DNA methylation is a fundamental epigenetic modification regulating gene expression. Aberrant DNA methylation is the most common molecular lesion in cancer cells. However, medical intervention has been limited to the use of broadly acting, small molecule-based demethylating drugs with significant side-effects and toxicities. To allow for targeted DNA demethylation, we integrated two nucleic acid-based approaches: DNMT1 interacting RNA (DiR) and RNA aptamer strategy. By combining the RNA inherent capabilities of inhibiting DNMT1 with an aptamer platform, we generated a first-in-class DNMT1-targeted approach - aptaDiR. Molecular modelling of RNA-DNMT1 complexes coupled with biochemical and cellular assays enabled the identification and characterization of aptaDiR. This RNA bio-drug is able to block DNA methylation, impair cancer cell viability and inhibit tumour growth in vivo. Collectively, we present an innovative RNA-based approach to modulate DNMT1 activity in cancer or diseases characterized by aberrant DNA methylation and suggest the first alternative strategy to overcome the limitations of currently approved non-specific hypomethylating protocols, which will greatly improve clinical intervention on DNA methylation.


Subject(s)
DNA Methylation , RNA , RNA/genetics , RNA/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Expression Regulation, Neoplastic , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Epigenesis, Genetic
9.
Mol Ther Nucleic Acids ; 29: 384-386, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-35991313

ABSTRACT

Aptamers are RNAs that can bind proteins directly and modulate protein-protein interactions. Given their therapeutic potential, aptamers would be expected to capture the interest of both scientists and investors. However, concerns regarding safety, efficacy, and delivery have delayed aptamer development and dampened investor support. Herein, we discuss the major hurdles stalling the translational application of aptamers over recent years and focus on approaches to overcome current barriers and attract the scientific community and investors to the aptamer field.

10.
Blood ; 114(27): 5415-25, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-19797526

ABSTRACT

Acute promyelocytic leukemia (APL) is characterized by a block in differentiation and accumulation of promyelocytes in the bone marrow and blood. The majority of APL patients harbor the t(15:17) translocation leading to expression of the fusion protein promyelocytic-retinoic acid receptor alpha. Treatment with retinoic acid leads to degradation of promyelocytic-retinoic acid receptor alpha protein and disappearance of leukemic cells; however, 30% of APL patients relapse after treatment. One potential mechanism for relapse is the persistence of cancer "stem" cells in hematopoietic organs after treatment. Using a novel sorting strategy we developed to isolate murine myeloid cells at distinct stages of differentiation, we identified a population of committed myeloid cells (CD34(+), c-kit(+), FcgammaRIII/II(+), Gr1(int)) that accumulates in the spleen and bone marrow in a murine model of APL. We observed that these cells are capable of efficiently generating leukemia in recipient mice, demonstrating that this population represents the APL cancer-initiating cell. These cells down-regulate the transcription factor CCAAT/enhancer binding protein alpha (C/EBPalpha) possibly through a methylation-dependent mechanism, indicating that C/EBPalpha deregulation contributes to transformation of APL cancer-initiating cells. Our findings provide further understanding of the biology of APL by demonstrating that a committed transformed progenitor can initiate and propagate the disease.


Subject(s)
Bone Marrow Cells/pathology , Leukemia, Promyelocytic, Acute/pathology , Myeloid Cells/pathology , Neoplastic Stem Cells/pathology , Animals , Antigens, CD34/metabolism , Bone Marrow Cells/metabolism , CCAAT-Enhancer-Binding Protein-alpha/genetics , Calgranulin A/genetics , Calgranulin A/metabolism , Cell Line, Tumor , DNA Methylation , Female , Flow Cytometry , Humans , Kaplan-Meier Estimate , Leukemia, Promyelocytic, Acute/blood , Leukemia, Promyelocytic, Acute/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/metabolism , Neoplastic Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-kit/metabolism , U937 Cells
11.
Genes (Basel) ; 12(3)2021 03 20.
Article in English | MEDLINE | ID: mdl-33804735

ABSTRACT

ADP-ribosylation, is a reversible post-translational modification implicated in major biological functions. Poly ADP-ribose polymerases (PARP) are specialized enzymes that catalyze the addition of ADP ribose units from "nicotinamide adenine dinucleotide-donor molecules" to their target substrates. This reaction known as PARylation modulates essential cellular processes including DNA damage response, chromatin remodeling, DNA methylation and gene expression. Herein, we discuss emerging roles of PARP1 in chromatin remodeling and epigenetic regulation, focusing on its therapeutic implications for cancer treatment and beyond.


Subject(s)
Epigenesis, Genetic , Neoplasms/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Chromatin Assembly and Disassembly , DNA Methylation , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Poly (ADP-Ribose) Polymerase-1/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Poly(ADP-ribose) Polymerases/metabolism , Protein Processing, Post-Translational
12.
Sci Adv ; 7(40): eabg1695, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34597139

ABSTRACT

Pseudogenes, noncoding homologs of protein-coding genes, once considered nonfunctional evolutionary relics, have recently been linked to patient prognoses and cancer subtypes. Despite this potential clinical importance, only a handful of >12,000 pseudogenes in humans have been characterized in cancers to date. Here, we describe a previously unrecognized role for pseudogenes as potent epigenetic regulators that can demethylate and activate oncogenes. We focused on SALL4, a known oncogene in hepatocellular carcinoma (HCC) with eight pseudogenes. Using a locus-specific demethylating technology, we identified the critical CpG region for SALL4 expression. We demonstrated that SALL4 pseudogene 5 hypomethylates this region through interaction with DNMT1, resulting in SALL4 up-regulation. Intriguingly, pseudogene 5 is significantly up-regulated in a hepatitis B virus model before SALL4 induction, and both are increased in patients with HBV-HCC. Our results suggest that pseudogene-mediated demethylation represents a novel mechanism of oncogene activation in cancer.

13.
Cells ; 10(11)2021 11 02.
Article in English | MEDLINE | ID: mdl-34831209

ABSTRACT

Nutritional intake impacts the human epigenome by directing epigenetic pathways in normal cell development via as yet unknown molecular mechanisms. Consequently, imbalance in the nutritional intake is able to dysregulate the epigenetic profile and drive cells towards malignant transformation. Here we present a novel epigenetic effect of the essential nutrient, NAD. We demonstrate that impairment of DNMT1 enzymatic activity by NAD-promoted ADP-ribosylation leads to demethylation and transcriptional activation of the CEBPA gene, suggesting the existence of an unknown NAD-controlled region within the locus. In addition to the molecular events, NAD- treated cells exhibit significant morphological and phenotypical changes that correspond to myeloid differentiation. Collectively, these results delineate a novel role for NAD in cell differentiation, and indicate novel nutri-epigenetic strategies to regulate and control gene expression in human cells.


Subject(s)
Cell Differentiation , DNA Methylation/genetics , NAD/pharmacology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/drug effects , Cell Line , DNA Demethylation/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Myeloid Cells/cytology , Myeloid Cells/drug effects , Neoplasms/genetics , Neoplasms/pathology , Oxidative Phosphorylation/drug effects , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects
14.
Sci Immunol ; 6(56)2021 02 05.
Article in English | MEDLINE | ID: mdl-33547048

ABSTRACT

E-cadherin is a calcium-dependent cell-cell adhesion molecule extensively studied for its involvement in tissue formation, epithelial cell behavior, and suppression of cancer. However, E-cadherin expression in the hematopoietic system has not been fully elucidated. Combining single-cell RNA-sequencing analyses and immunophenotyping, we revealed that progenitors expressing high levels of E-cadherin and contained within the granulocyte-monocyte progenitors (GMPs) fraction have an enriched capacity to differentiate into basophils and mast cells. We detected E-cadherin expression on committed progenitors before the expression of other reported markers of these lineages. We named such progenitors pro-BMPs (pro-basophil and mast cell progenitors). Using RNA sequencing, we observed transcriptional priming of pro-BMPs to the basophil and mast cell lineages. We also showed that GATA-2 directly regulates E-cadherin expression in the basophil and mast cell lineages, thus providing a mechanistic connection between the expression of this cell surface marker and the basophil and mast cell fate specification.


Subject(s)
Cadherins/genetics , GATA2 Transcription Factor/metabolism , Hematopoietic Stem Cells/physiology , Animals , Basophils/physiology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Cells, Cultured , Mast Cells/physiology , Mice , Primary Cell Culture , RNA-Seq , Single-Cell Analysis
15.
Blood Cells Mol Dis ; 45(3): 181-5, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20655775

ABSTRACT

DNA methylation is one of the major epigenetic changes in human cancers, leading to silencing of tumor suppressor genes, with a pathogenetic role in tumor development and progression in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Methylation of key promoter regions, induced by cytotoxic therapy together with complex genetic changes, is important in the biology of therapy-related myeloid neoplasms (t-MN). We were interested in the characterization of the methylation pattern of AML and MDS de novo and therapy-related. We studied 385 patients (179 females, 206 males), of a median age of 66 years (range 16-98 years). There were 105 MDS, 208 de novo AML and 72 t-MN (45 MDS and 27 AML). Using a methylation-specific PCR, we studied the promoter methylation status of E-cadherin (CDH1), TSP1 and DAP-Kinase 1. These genes have been shown to be involved in the malignant transformation, interfering with angiogenesis, interaction with micro-environment, apoptosis and xenobiotic detoxification. We found no associations between promoter hypermethylation and gender or age at the time of initial diagnosis. In patients with MDS, there were no associations between hypermethylation and clinical characteristics, including IPSS score, WHO classification and cytogenetics. DAPK1 was more frequently methylated in t-MDS/AML when compared to de novo MDS and AML (39% vs 15.3% and 24.4%, p=0.0001), while methylation of CDH1 was similar in t-MDS/AML and AML (51% and 53.4%), but less frequent in de novo MDS (29%) (p=0.003). In the t-MDS/AML group, we found that the methylation pattern appeared to be related to the primary tumor, with DAPK1 more frequently methylated in patients with a previous lymphoproliferative disease (75% vs 32%, p=0.006). On the other hand, methylation of CDH1 was associated to radiotherapy for the primary malignancy (84.5% vs 38%, p=0.003). TSP1 hypermethylation was rare and not characteristic of t-MDS/AML. In 177 patients studied for concurrent methylation of several promoters, t-MN and AML de novo were significantly more frequently hypermethylated in 2 or more promoter regions than de novo MDS (20% vs 12.4%, p<0.001). Chemotherapy and individual genetic predisposition have a role in t-MDS/AML development, the identification of specific epigenetic modifications may explain complexity and genomic instability of these diseases and give the basis for targeted-therapy. The significant association with previous malignancy subtypes may underlie a likely susceptibility to methylation of specific targets and a role for constitutional epimutations as predisposing factors for the development of therapy-related myeloid neoplasm.


Subject(s)
Apoptosis Regulatory Proteins , Cadherins , Calcium-Calmodulin-Dependent Protein Kinases , DNA Methylation , Leukemia, Myeloid, Acute/metabolism , Myelodysplastic Syndromes/metabolism , Neoplasms, Second Primary/metabolism , Promoter Regions, Genetic , Thrombospondin 1 , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, CD , Death-Associated Protein Kinases , Female , Humans , Male , Middle Aged
16.
Cells ; 9(5)2020 05 24.
Article in English | MEDLINE | ID: mdl-32456308

ABSTRACT

Human perinatal stem cells (SCs) can be isolated from fetal annexes without ethical or safety limitations. They are generally considered multipotent; nevertheless, their biological characteristics are still not fully understood. The aim of this study was to investigate the pluripotency potential of human perinatal SCs as compared to human induced pluripotent stem cells (hiPSCs). Despite the low expression of the pluripotent factors NANOG, OCT4, SOX2, and C-KIT in perinatal SC, we observed minor differences in the promoters DNA-methylation profile of these genes with respect to hiPSCs; we also demonstrated that in perinatal SCs miR-145-5p had an inverse trend in comparison to these stemness markers, suggesting that NANOG, OCT4, and SOX2 were regulated at the post-transcriptional level. The reduced expression of stemness markers was also associated with shorter telomere lengths and shift of the oxidative metabolism between hiPSCs and fetal annex-derived cells. Our findings indicate the differentiation ability of perinatal SCs might not be restricted to the mesenchymal lineage due to an epigenetic barrier, but other regulatory mechanisms such as telomere shortening or metabolic changes might impair their differentiation potential and challenge their clinical application.


Subject(s)
Epigenesis, Genetic , Stem Cells/cytology , Stem Cells/metabolism , Chromosomes, Human/metabolism , DNA Methylation/genetics , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Infant, Newborn , MicroRNAs/genetics , MicroRNAs/metabolism , Promoter Regions, Genetic , Telomere Homeostasis
17.
Cell Rep ; 32(12): 108181, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32966797

ABSTRACT

Hemopexin (Hx) is a scavenger of labile heme. Herein, we present data defining the role of tumor stroma-expressed Hx in suppressing cancer progression. Labile heme and Hx levels are inversely correlated in the plasma of patients with prostate cancer (PCa). Further, low expression of Hx in PCa biopsies characterizes poorly differentiated tumors and correlates with earlier time to relapse. Significantly, heme promotes tumor growth and metastases in an orthotopic murine model of PCa, with the most aggressive phenotype detected in mice lacking Hx. Mechanistically, labile heme accumulates in the nucleus and modulates specific gene expression via interacting with guanine quadruplex (G4) DNA structures to promote PCa growth. We identify c-MYC as a heme:G4-regulated gene and a major player in heme-driven cancer progression. Collectively, these results reveal that sequestration of labile heme by Hx may block heme-driven tumor growth and metastases, suggesting a potential strategy to prevent and/or arrest cancer dissemination.


Subject(s)
Heme/metabolism , Hemopexin/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , DNA/genetics , Disease Progression , G-Quadruplexes , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Metastasis , Phenotype , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/blood , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/metabolism , Treatment Outcome , Urokinase-Type Plasminogen Activator/metabolism
18.
Cells ; 8(10)2019 10 12.
Article in English | MEDLINE | ID: mdl-31614870

ABSTRACT

DNA methylation controls several cellular processes, from early development to old age, including biological responses to endogenous or exogenous stimuli contributing to disease transition. As a result, minimal DNA methylation changes during developmental stages drive severe phenotypes, as observed in germ-line imprinting disorders, while genome-wide alterations occurring in somatic cells are linked to cancer onset and progression. By summarizing the molecular events governing DNA methylation, we focus on the methods that have facilitated mapping and understanding of this epigenetic mark in healthy conditions and diseases. Overall, we review the bright (health-related) and dark (disease-related) side of DNA methylation changes, outlining how bulk and single-cell genomic analyses are moving toward the identification of new molecular targets and driving the development of more specific and less toxic demethylating agents.


Subject(s)
DNA Methylation/physiology , Animals , Epigenesis, Genetic , Epigenomics/methods , Genome , Genomic Imprinting , Humans , Neoplasms/metabolism
19.
Leuk Res ; 32(9): 1448-53, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18308386

ABSTRACT

Alterations of the transcription factors CCAAT/enhancer binding protein alpha (CEBPA) and PU.1 have been described in acute myeloid leukemia (AML). We studied CEBPA and PU.1 mRNA levels by real-time RT-PCR in 109 primary AML samples, compared with normal bone marrow and peripheral blood cells. Low PU.1 levels were observed in monoblastic leukemias, while low CEBPA levels were associated with leukopenia at diagnosis and lack of expression of differentiation antigens CD33 and CD11c. We conclude that down-regulation of CEBPA and PU.1 is not a general feature of primary AML, but appears to be restricted to distinct AML subtypes.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Adolescent , Adult , Aged , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Blotting, Western , CCAAT-Enhancer-Binding Proteins/metabolism , CD11c Antigen/genetics , CD11c Antigen/metabolism , Female , Flow Cytometry , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukopenia/genetics , Leukopenia/metabolism , Leukopenia/pathology , Male , Middle Aged , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sialic Acid Binding Ig-like Lectin 3 , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL