Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Cell ; 172(1-2): 90-105.e23, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29249359

ABSTRACT

R-2-hydroxyglutarate (R-2HG), produced at high levels by mutant isocitrate dehydrogenase 1/2 (IDH1/2) enzymes, was reported as an oncometabolite. We show here that R-2HG also exerts a broad anti-leukemic activity in vitro and in vivo by inhibiting leukemia cell proliferation/viability and by promoting cell-cycle arrest and apoptosis. Mechanistically, R-2HG inhibits fat mass and obesity-associated protein (FTO) activity, thereby increasing global N6-methyladenosine (m6A) RNA modification in R-2HG-sensitive leukemia cells, which in turn decreases the stability of MYC/CEBPA transcripts, leading to the suppression of relevant pathways. Ectopically expressed mutant IDH1 and S-2HG recapitulate the effects of R-2HG. High levels of FTO sensitize leukemic cells to R-2HG, whereas hyperactivation of MYC signaling confers resistance that can be reversed by the inhibition of MYC signaling. R-2HG also displays anti-tumor activity in glioma. Collectively, while R-2HG accumulated in IDH1/2 mutant cancers contributes to cancer initiation, our work demonstrates anti-tumor effects of 2HG in inhibiting proliferation/survival of FTO-high cancer cells via targeting FTO/m6A/MYC/CEBPA signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Glutarates/pharmacology , Leukemia/drug therapy , Signal Transduction/drug effects , Adenosine/analogs & derivatives , Adenosine/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Animals , Antineoplastic Agents/therapeutic use , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line, Tumor , Glutarates/therapeutic use , HEK293 Cells , Humans , Jurkat Cells , Mice , Proto-Oncogene Proteins c-myc/metabolism , RNA Processing, Post-Transcriptional
2.
Nature ; 567(7748): 414-419, 2019 03.
Article in English | MEDLINE | ID: mdl-30867593

ABSTRACT

DNA and histone modifications have notable effects on gene expression1. Being the most prevalent internal modification in mRNA, the N6-methyladenosine (m6A) mRNA modification is as an important post-transcriptional mechanism of gene regulation2-4 and has crucial roles in various normal and pathological processes5-12. However, it is unclear how m6A is specifically and dynamically deposited in the transcriptome. Here we report that histone H3 trimethylation at Lys36 (H3K36me3), a marker for transcription elongation, guides m6A deposition globally. We show that m6A modifications are enriched in the vicinity of H3K36me3 peaks, and are reduced globally when cellular H3K36me3 is depleted. Mechanistically, H3K36me3 is recognized and bound directly by METTL14, a crucial component of the m6A methyltransferase complex (MTC), which in turn facilitates the binding of the m6A MTC to adjacent RNA polymerase II, thereby delivering the m6A MTC to actively transcribed nascent RNAs to deposit m6A co-transcriptionally. In mouse embryonic stem cells, phenocopying METTL14 knockdown, H3K36me3 depletion also markedly reduces m6A abundance transcriptome-wide and in pluripotency transcripts, resulting in increased cell stemness. Collectively, our studies reveal the important roles of H3K36me3 and METTL14 in determining specific and dynamic deposition of m6A in mRNA, and uncover another layer of gene expression regulation that involves crosstalk between histone modification and RNA methylation.


Subject(s)
Adenosine/analogs & derivatives , Histones/chemistry , Histones/metabolism , Lysine/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Transcription, Genetic , Adenosine/metabolism , Animals , Cell Differentiation , Cell Line , Embryonic Stem Cells/metabolism , Humans , Lysine/chemistry , Methylation , Methyltransferases/deficiency , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , RNA Polymerase II/metabolism , Transcription Elongation, Genetic , Transcriptome/genetics
3.
Trends Genet ; 36(1): 44-52, 2020 01.
Article in English | MEDLINE | ID: mdl-31810533

ABSTRACT

N6-Methyladenosine (m6A) is the most prevalent internal RNA modification in mRNA, and has been found to be highly conserved and hard-coded in mammals and other eukaryotic species. The importance of m6A for gene expression regulation and cell fate decisions has been well acknowledged in the past few years. However, it was only until recently that the mechanisms underlying the biogenesis and specificity of m6A modification in cells were uncovered. We review up-to-date knowledge on the biogenesis of the RNA m6A modification, including the cis-regulatory elements and trans-acting factors that determine general de novo m6A deposition and modulate cell type-specific m6A patterns, and we discuss the biological significance of such regulation.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/genetics , Methylation , RNA/genetics , Adenosine/metabolism , Animals , Cell Differentiation/genetics , Eukaryotic Cells , Gene Expression Regulation/genetics , Humans , Protein Processing, Post-Translational/genetics , RNA/metabolism , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Adv Exp Med Biol ; 1442: 105-123, 2023.
Article in English | MEDLINE | ID: mdl-38228961

ABSTRACT

Over 170 nucleotide variants have been discovered in messenger RNAs (mRNAs) and non-coding RNAs so far. However, only a few of them, including N6-methyladenosine (m6A), 5-methylcytidine (m5C), and N1-methyladenosine (m1A), could be mapped in the transcriptome. These RNA modifications appear to be dynamically regulated, with writer, eraser, and reader proteins being identified for each modification. As a result, there is a growing interest in studying their biological impacts on normal bioprocesses and tumorigenesis over the past few years. As the most abundant internal modification in eukaryotic mRNAs, m6A plays a vital role in the post-transcriptional regulation of mRNA fate via regulating almost all aspects of mRNA metabolism, including RNA splicing, nuclear export, RNA stability, and translation. Studies on mRNA m6A modification serve as a great example for exploring other modifications on mRNA. In this chapter, we will review recent advances in the study of biological functions and regulation of mRNA modifications, specifically m6A, in both normal hematopoiesis and malignant hematopoiesis. We will also discuss the potential of targeting mRNA modifications as a treatment for hematopoietic disorders.


Subject(s)
Adenosine/analogs & derivatives , Gene Expression Regulation , Hematopoiesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Methylation , Hematopoiesis/genetics , RNA/genetics , RNA/metabolism
5.
Haematologica ; 105(1): 148-160, 2020 01.
Article in English | MEDLINE | ID: mdl-30975912

ABSTRACT

Homoharringtonine, a plant alkaloid, has been reported to suppress protein synthesis and has been approved by the US Food and Drug Administration for the treatment of chronic myeloid leukemia. Here we show that in acute myeloid leukemia (AML), homoharringtonine potently inhibits cell growth/viability and induces cell cycle arrest and apoptosis, significantly inhibits disease progression in vivo, and substantially prolongs survival of mice bearing murine or human AML. Strikingly, homoharringtonine treatment dramatically decreases global DNA 5-hydroxymethylcytosine abundance through targeting the SP1/TET1 axis, and TET1 depletion mimics homoharringtonine's therapeutic effects in AML. Our further 5hmC-seq and RNA-seq analyses, followed by a series of validation and functional studies, suggest that FLT3 is a critical down-stream target of homoharringtonine/SP1/TET1/5hmC signaling, and suppression of FLT3 and its downstream targets (e.g. MYC) contributes to the high sensitivity of FLT3-mutated AML cells to homoharringtonine. Collectively, our studies uncover a previously unappreciated DNA epigenome-related mechanism underlying the potent antileukemic effect of homoharringtonine, which involves suppression of the SP1/TET1/5hmC/FLT3/MYC signaling pathways in AML. Our work also highlights the particular promise of clinical application of homoharringtonine to treat human AML with FLT3 mutations, which accounts for more than 30% of total cases of AML.


Subject(s)
Epigenome , Leukemia, Myeloid, Acute , Animals , Cell Line, Tumor , DNA , DNA-Binding Proteins , Homoharringtonine , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mice , Proto-Oncogene Proteins/genetics , fms-Like Tyrosine Kinase 3
6.
Adv Exp Med Biol ; 1143: 75-93, 2019.
Article in English | MEDLINE | ID: mdl-31338816

ABSTRACT

As the most abundant internal modification in eukaryotic messenger RNAs (mRNAs), N 6-methyladenosine (m6A) modification has been shown recently to posttranscriptionally regulate expression of thousands of messenger RNA (mRNA) transcripts in each mammalian cell type in a dynamic and reversible manner. This epigenetic mark is deposited by the m6A methyltransferase complex (i.e., the METTL3/METTL14/WTAP complex and other cofactor proteins) and erased by m6A demethylases such as FTO and ALKBH5. Specific recognition of these m6A-modified mRNAs by m6A-binding proteins (i.e., m6A readers) determines the fate of target mRNAs through affecting splicing, nuclear export, RNA stability, and/or translation. During the past few years, m6A modification has been demonstrated to play a critical role in many major normal bioprocesses including self-renewal and differentiation of embryonic stem cells and hematopoietic stem cells, tissue development, circadian rhythm, heat shock or DNA damage response, and sex determination. Thus, it is not surprising that dysregulation of the m6A machinery is also closely associated with pathogenesis and drug response of both solid tumors and hematologic malignancies. In this chapter, we summarize and discuss recent findings regarding the biological functions and underlying mechanisms of m6A modification and the associated machinery in normal hematopoiesis and the initiation, progression, and drug response of acute myeloid leukemia (AML), a major subtype of leukemia usually associated with unfavorable prognosis.


Subject(s)
Adenosine , Hematopoiesis , Leukemia, Myeloid, Acute , Methyltransferases , RNA, Messenger , Adenosine/metabolism , Animals , Cell Differentiation , Drug Resistance, Neoplasm/genetics , Hematopoiesis/genetics , Humans , Leukemia, Myeloid, Acute/physiopathology , Methyltransferases/metabolism , RNA, Messenger/metabolism
7.
Blood ; 126(17): 2005-15, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26361793

ABSTRACT

It is generally assumed that gain- and loss-of-function manipulations of a functionally important gene should lead to the opposite phenotypes. We show in this study that both overexpression and knockout of microRNA (miR)-126 surprisingly result in enhanced leukemogenesis in cooperation with the t(8;21) fusion genes AML1-ETO/RUNX1-RUNX1T1 and AML1-ETO9a (a potent oncogenic isoform of AML1-ETO). In accordance with our observation that increased expression of miR-126 is associated with unfavorable survival in patients with t(8;21) acute myeloid leukemia (AML), we show that miR-126 overexpression exhibits a stronger effect on long-term survival and progression of AML1-ETO9a-mediated leukemia stem cells/leukemia initiating cells (LSCs/LICs) in mice than does miR-126 knockout. Furthermore, miR-126 knockout substantially enhances responsiveness of leukemia cells to standard chemotherapy. Mechanistically, miR-126 overexpression activates genes that are highly expressed in LSCs/LICs and/or primitive hematopoietic stem/progenitor cells, likely through targeting ERRFI1 and SPRED1, whereas miR-126 knockout activates genes that are highly expressed in committed, more differentiated hematopoietic progenitor cells, presumably through inducing FZD7 expression. Our data demonstrate that miR-126 plays a critical but 2-faceted role in leukemia and thereby uncover a new layer of miRNA regulation in cancer. Moreover, because miR-126 depletion can sensitize AML cells to standard chemotherapy, our data also suggest that miR-126 represents a promising therapeutic target.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Transformation, Neoplastic/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , MicroRNAs/genetics , Animals , Cell Differentiation/genetics , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 8/genetics , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Staging , Oncogene Proteins, Fusion/genetics , Prognosis , Survival Rate , Translocation, Genetic/genetics
8.
Curr Opin Genet Dev ; 86: 102205, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38776766

ABSTRACT

N6-methyladenosine (m6A) is the most prevalent internal RNA modification in eukaryotic messenger RNAs (mRNAs), regulating gene expression at the transcription and post-transcription levels. Complex interplay between m6A and other well-studied epigenetic modifications, including histone modifications and DNA modification, has been extensively reported in recent years. The crosstalk between RNA m6A modification and histone/DNA modifications plays a critical role in establishing the chromatin state for the precise and specific fine-tuning of gene expression and undoubtedly has profound impacts on both physiological and pathological processes. In this review, we discuss the crosstalk between RNA m6A modification and histone/DNA modifications, emphasizing their sophisticated communications and the mechanisms underlying to gain a comprehensive view of the biological relevance of m6A-based epigenetic network.


Subject(s)
Adenosine , Epigenesis, Genetic , Histones , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/genetics , Chromatin/genetics , Chromatin/metabolism , DNA/genetics , DNA/metabolism , DNA Methylation/genetics , Histones/metabolism , Histones/genetics , RNA/metabolism , RNA/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Genes Dis ; 11(1): 382-396, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37588203

ABSTRACT

As the most common internal modification of mRNA, N6-methyladenosine (m6A) and its regulators modulate gene expression and play critical roles in various biological and pathological processes including tumorigenesis. It was reported previously that m6A methyltransferase (writer), methyltransferase-like 3 (METTL3) adds m6A in primary microRNAs (pri-miRNAs) and facilitates its processing into precursor miRNAs (pre-miRNAs). However, it is unknown whether m6A modification also plays a role in the maturation process of pre-miRNAs and (if so) whether such a function contributes to tumorigenesis. Here, we found that YTHDF2 is aberrantly overexpressed in acute myeloid leukemia (AML) patients, especially in relapsed patients, and plays an oncogenic role in AML. Moreover, YTHDF2 promotes expression of miR-126-3p (also known as miR-126, as it is the main product of precursor miR-126 (pre-miR-126)), a miRNA that was reported as an oncomiRNA in AML, through facilitating the processing of pre-miR-126 into mature miR-126. Mechanistically, YTHDF2 recognizes m6A modification in pre-miR-126 and recruits AGO2, a regulator of pre-miRNA processing, to promote the maturation of pre-miR-126. YTHDF2 positively and negatively correlates with miR-126 and miR-126's downstream target genes, respectively, in AML patients, and forced expression of miR-126 could largely rescue YTHDF2/Ythdf2 depletion-mediated suppression on AML cell growth/proliferation and leukemogenesis, indicating that miR-126 is a functionally important target of YTHDF2 in AML. Overall, our studies not only reveal a previously unappreciated YTHDF2/miR-126 axis in AML and highlight the therapeutic potential of targeting this axis for AML treatment, but also suggest that m6A plays a role in pre-miRNA processing that contributes to tumorigenesis.

10.
Sci Transl Med ; 15(689): eabq8513, 2023 03 29.
Article in English | MEDLINE | ID: mdl-36989375

ABSTRACT

Although the overall survival rate of B cell acute lymphoblastic leukemia (B-ALL) in childhood is more than 80%, it is merely 30% in refractory/relapsed and adult patients with B-ALL. This demonstrates a need for improved therapy targeting this subgroup of B-ALL. Here, we show that the ten-eleven translocation 1 (TET1) protein, a dioxygenase involved in DNA demethylation, is overexpressed and plays a crucial oncogenic role independent of its catalytic activity in B-ALL. Consistent with its oncogenic role in B-ALL, overexpression of TET1 alone in normal precursor B cells is sufficient to transform the cells and cause B-ALL in mice within 3 to 4 months. We found that TET1 protein is stabilized and overexpressed because of its phosphorylation mediated by protein kinase C epsilon (PRKCE) and ATM serine/threonine kinase (ATM), which are also overexpressed in B-ALL. Mechanistically, TET1 recruits STAT5B to the promoters of CD72 and JCHAIN and promotes their transcription, which in turn promotes B-ALL development. Destabilization of TET1 protein by treatment with PKC or ATM inhibitors (staurosporine or AZD0156; both tested in clinical trials), or by pharmacological targeting of STAT5B, greatly decreases B-ALL cell viability and inhibits B-ALL progression in vitro and in vivo. The combination of AZD0156 with staurosporine or vincristine exhibits a synergistic effect on inhibition of refractory/relapsed B-ALL cell survival and leukemia progression in PDX models. Collectively, our study reveals an oncogenic role of the phosphorylated TET1 protein in B-ALL independent of its catalytic activity and highlights the therapeutic potential of targeting TET1 signaling for the treatment of refractory/relapsed B-ALL.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Proto-Oncogene Proteins , Animals , Mice , Proto-Oncogene Proteins/metabolism , Phosphorylation , Staurosporine , Signal Transduction , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , DNA-Binding Proteins/metabolism
11.
RNA Biol ; 9(8): 1099-109, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22858673

ABSTRACT

cyclin D3 (CCND3) is one of the three D-type cyclins that regulate the G1/S phase transition of the cell cycle. Expression of CCND3 is observed in nearly all proliferating cells; however, the presence of high levels of CCND3 has been linked to a poor prognosis for several types of cancer. Therefore, further mechanistic studies on the regulation of CCND3 expression are urgently needed to provide therapeutic implications. In this study, we report that a conserved RNA G-quadruplex-forming sequence (hereafter CRQ), located in the 5' UTR of mammalian CCND3 mRNA, is able to fold into an extremely stable, intramolecular, parallel G-quadruplex in vitro. The CRQ G-quadruplex dramatically reduces the activity of a reporter gene in human cell lines, but it has little impact on its mRNA level, indicating a translational repression. Moreover, the CRQ sequence in its natural context inhibits translation of CCND3. Disruption of the G-quadruplex structure by G/U-mutation or deletion results in an elevated expression of CCND3 and an increased phosphorylation of Rb, a downstream target of CCND3, which promotes progression of cells through the G1 phase. Our results add to the growing understanding of the regulation of CCND3 expression and provide a potential therapeutic target for cancer treatment.


Subject(s)
5' Untranslated Regions , Cell Cycle , Cyclin D3/genetics , G-Quadruplexes , Gene Expression Regulation , Protein Biosynthesis , Base Sequence , Cell Proliferation , Humans , Molecular Sequence Data , RNA, Messenger/chemistry , Ribonuclease T1/metabolism
12.
Cancer Cell ; 40(12): 1566-1582.e10, 2022 12 12.
Article in English | MEDLINE | ID: mdl-36306790

ABSTRACT

N6-Methyladenosine (m6A) modification and its modulators play critical roles and show promise as therapeutic targets in human cancers, including acute myeloid leukemia (AML). IGF2BP2 was recently reported as an m6A binding protein that enhances mRNA stability and translation. However, its function in AML remains largely elusive. Here we report the oncogenic role and the therapeutic targeting of IGF2BP2 in AML. High expression of IGF2BP2 is observed in AML and associates with unfavorable prognosis. IGF2BP2 promotes AML development and self-renewal of leukemia stem/initiation cells by regulating expression of critical targets (e.g., MYC, GPT2, and SLC1A5) in the glutamine metabolism pathways in an m6A-dependent manner. Inhibiting IGF2BP2 with our recently identified small-molecule compound (CWI1-2) shows promising anti-leukemia effects in vitro and in vivo. Collectively, our results reveal a role of IGF2BP2 and m6A modification in amino acid metabolism and highlight the potential of targeting IGF2BP2 as a promising therapeutic strategy in AML.


Subject(s)
Glutamine , Leukemia, Myeloid, Acute , Humans , Glutamine/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , RNA Stability , Prognosis , Minor Histocompatibility Antigens , Amino Acid Transport System ASC/genetics , Amino Acid Transport System ASC/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
13.
Cancer Commun (Lond) ; 41(7): 538-559, 2021 07.
Article in English | MEDLINE | ID: mdl-33955720

ABSTRACT

As one of the most studied ribonucleic acid (RNA) modifications in eukaryotes, N6 -methyladenosine (m6 A) has been shown to play a predominant role in controlling gene expression and influence physiological and pathological processes such as oncogenesis and tumor progression. Writer and eraser proteins, acting opposite to deposit and remove m6 A epigenetic marks, respectively, shape the cellular m6 A landscape, while reader proteins preferentially recognize m6 A modifications and mediate fate decision of the methylated RNAs, including RNA synthesis, splicing, exportation, translation, and stability. Therefore, RNA metabolism in cells is greatly influenced by these three classes of m6 A regulators. Aberrant expression of m6 A regulators has been widely reported in various types of cancer, leading to cancer initiation, progression, and drug resistance. The close links between m6 A and cancer shed light on the potential use of m6 A methylation and its regulators as prognostic biomarkers and drug targets for cancer therapy. Given the notable effects of m6 A in reversing chemoresistance and enhancing immune therapy, it is a promising target for combined therapy. Herein, we summarize the recent discoveries on m6 A and its regulators, emphasizing their influences on RNA metabolism, their dysregulation and impacts in diverse malignancies, and discuss the clinical implications of m6 A modification in cancer.


Subject(s)
Adenosine , Neoplasms , Adenosine/metabolism , Humans , Methylation , Neoplasms/drug therapy , Neoplasms/genetics , RNA , RNA, Messenger/metabolism
14.
J Hematol Oncol ; 13(1): 117, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32854717

ABSTRACT

N6-methyladenosine (m6A) is the most abundant mRNA modification and is catalyzed by the methyltransferase complex, in which methyltransferase-like 3 (METTL3) is the sole catalytic subunit. Accumulating evidence in recent years reveals that METTL3 plays key roles in a variety of cancer types, either dependent or independent on its m6A RNA methyltransferase activity. While the roles of m6A modifications in cancer have been extensively reviewed elsewhere, the critical functions of METTL3 in various types of cancer, as well as the potential targeting of METTL3 as cancer treatment, have not yet been highlighted. Here we summarize our current understanding both on the oncogenic and tumor-suppressive functions of METTL3, as well as the underlying molecular mechanisms. The well-documented protein structure of the METTL3/METTL14 heterodimer provides the basis for potential therapeutic targeting, which is also discussed in this review.


Subject(s)
Adenosine/analogs & derivatives , Methyltransferases/physiology , Molecular Targeted Therapy , Neoplasm Proteins/physiology , Neoplasms/metabolism , RNA Processing, Post-Transcriptional , RNA, Neoplasm/metabolism , Adenosine/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Binding, Competitive , Carcinogens , Drug Design , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic , Humans , Methylation , Methyltransferases/antagonists & inhibitors , Methyltransferases/chemistry , Methyltransferases/genetics , Models, Molecular , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/therapy , Oncogenes , Protein Conformation , Protein Domains , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Substrate Specificity , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
15.
Cancer Cell ; 37(3): 270-288, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32183948

ABSTRACT

N6-Methyladenosine (m6A) RNA modification has emerged in recent years as a new layer of regulatory mechanism controlling gene expression in eukaryotes. As a reversible epigenetic modification found not only in messenger RNAs but also in non-coding RNAs, m6A affects the fate of the modified RNA molecules and plays important roles in almost all vital bioprocesses, including cancer development. Here we review the up-to-date knowledge of the pathological roles and underlying molecular mechanism of m6A modifications (in both coding and non-coding RNAs) in cancer pathogenesis and drug response/resistance, and discuss the therapeutic potential of targeting m6A regulators for cancer therapy.


Subject(s)
Adenosine/analogs & derivatives , Immunotherapy/methods , Neoplasms/genetics , RNA, Messenger , RNA, Untranslated , Adenosine/genetics , Adenosine/metabolism , Epigenesis, Genetic , Female , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Humans , Male , Mutation , Neoplasms/pathology , Neoplasms/therapy , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Int J Biol Sci ; 16(15): 2853-2867, 2020.
Article in English | MEDLINE | ID: mdl-33061801

ABSTRACT

MicroRNAs (miRNAs) and N6-methyladenosine (m6A) are known to serve as key regulators of acute myeloid leukemia (AML). Our previous microarray analysis indicated miR-550-1 was significantly downregulated in AML. The specific biological roles of miR-550-1 and its indirect interactions and regulation of m6A in AML, however, remain poorly understood. At the present study, we found that miR-550-1 was significantly down-regulated in primary AML samples from human patients, likely owing to hypermethylation of the associated CpG islands. When miR-550-1 expression was induced, it impaired AML cell proliferation both in vitro and in vivo, thus suppressing tumor development. When ectopically expressed, miR-550-1 drove the G0/1 cell cycle phase arrest, differentiation, and apoptotic death of affected cells. We confirmed mechanistically that WW-domain containing transcription regulator-1 (WWTR1) gene was a downstream target of miR-550-1. Moreover, we also identified Wilms tumor 1-associated protein (WTAP), a vital component of the m6A methyltransferase complex, as a target of miR-550-1. These data indicated that miR-550-1 might mediate a decrease in m6A levels via targeting WTAP, which led to a further reduction in WWTR1 stability. Using gain- and loss-of-function approaches, we were able to determine that miR-550-1 disrupted the proliferation and tumorigenesis of AML cells at least in part via the direct targeting of WWTR1. Taken together, our results provide direct evidence that miR-550-1 acts as a tumor suppressor in the context of AML pathogenesis, suggesting that efforts to bolster miR-550-1 expression in AML patients may thus be a viable clinical strategy to improve patient outcomes.


Subject(s)
Leukemia, Myeloid, Acute , MicroRNAs , Cell Line, Tumor , Cell Proliferation/genetics , Genes, Tumor Suppressor , Hippo Signaling Pathway , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , MicroRNAs/metabolism
17.
Cell Stem Cell ; 27(1): 64-80.e9, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32402250

ABSTRACT

N6-methyladenosine (m6A), the most abundant internal modification in mRNA, has been implicated in tumorigenesis. As an m6A demethylase, ALKBH5 has been shown to promote the development of breast cancer and brain tumors. However, in acute myeloid leukemia (AML), ALKBH5 was reported to be frequently deleted, implying a tumor-suppressor role. Here, we show that ALKBH5 deletion is rare in human AML; instead, ALKBH5 is aberrantly overexpressed in AML. Moreover, its increased expression correlates with poor prognosis in AML patients. We demonstrate that ALKBH5 is required for the development and maintenance of AML and self-renewal of leukemia stem/initiating cells (LSCs/LICs) but not essential for normal hematopoiesis. Mechanistically, ALKBH5 exerts tumor-promoting effects in AML by post-transcriptional regulation of its critical targets such as TACC3, a prognosis-associated oncogene in various cancers. Collectively, our findings reveal crucial functions of ALKBH5 in leukemogenesis and LSC/LIC self-renewal/maintenance and highlight the therapeutic potential of targeting the ALKBH5/m6A axis.


Subject(s)
Cell Self Renewal , Leukemia, Myeloid, Acute , AlkB Homolog 5, RNA Demethylase/genetics , Carcinogenesis/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Microtubule-Associated Proteins , Neoplastic Stem Cells
19.
Cancer Cell ; 38(1): 79-96.e11, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32531268

ABSTRACT

Fat mass and obesity-associated protein (FTO), an RNA N6-methyladenosine (m6A) demethylase, plays oncogenic roles in various cancers, presenting an opportunity for the development of effective targeted therapeutics. Here, we report two potent small-molecule FTO inhibitors that exhibit strong anti-tumor effects in multiple types of cancers. We show that genetic depletion and pharmacological inhibition of FTO dramatically attenuate leukemia stem/initiating cell self-renewal and reprogram immune response by suppressing expression of immune checkpoint genes, especially LILRB4. FTO inhibition sensitizes leukemia cells to T cell cytotoxicity and overcomes hypomethylating agent-induced immune evasion. Our study demonstrates that FTO plays critical roles in cancer stem cell self-renewal and immune evasion and highlights the broad potential of targeting FTO for cancer therapy.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/antagonists & inhibitors , Cell Self Renewal/drug effects , Enzyme Inhibitors/pharmacology , Immune Evasion/drug effects , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/chemistry , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Anthracenes/chemistry , Anthracenes/pharmacology , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immune Evasion/genetics , Leukemia/genetics , Leukemia/pathology , Leukemia/prevention & control , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Molecular Structure , Protein Binding/drug effects , Protein Domains , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL