Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Cell ; 175(5): 1228-1243.e20, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30392959

ABSTRACT

Genetic drivers of cancer can be dysregulated through epigenetic modifications of DNA. Although the critical role of DNA 5-methylcytosine (5mC) inĀ the regulation of transcription is recognized, the functions of other non-canonical DNA modifications remain obscure. Here, we report the identification of novel N6-methyladenine (N6-mA) DNA modifications in human tissues and implicate this epigenetic mark in human disease, specifically theĀ highly malignant brain cancer glioblastoma. Glioblastoma markedly upregulated N6-mA levels, which co-localized with heterochromatic histone modifications, predominantly H3K9me3. N6-mA levels were dynamically regulated by the DNA demethylase ALKBH1, depletion of which led to transcriptional silencing of oncogenic pathways through decreasing chromatin accessibility. Targeting the N6-mA regulator ALKBH1 in patient-derived human glioblastoma models inhibited tumor cell proliferation and extended the survival of tumor-bearing mice, supporting this novel DNA modificationĀ as a potential therapeutic target for glioblastoma. Collectively, our results uncover a novel epigenetic node in cancer through the DNA modification N6-mA.


Subject(s)
Adenine/analogs & derivatives , Brain Neoplasms/pathology , DNA Methylation , Glioblastoma/pathology , Adenine/analysis , Adenine/chemistry , Adult , Aged , AlkB Homolog 1, Histone H2a Dioxygenase/antagonists & inhibitors , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Cell Hypoxia , Child , Epigenomics , Female , Glioblastoma/metabolism , Glioblastoma/mortality , Heterochromatin/metabolism , Histones/metabolism , Humans , Kaplan-Meier Estimate , Male , Mice , Middle Aged , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism
2.
Cell ; 167(3): 816-828.e16, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27745969

ABSTRACT

tRNA is a central component of protein synthesis andĀ the cell signaling network. One salient feature of tRNA is its heavily modified status, which can critically impact its function. Here, we show that mammalian ALKBH1 is a tRNA demethylase. It mediates the demethylation of N1-methyladenosine (m1A) in tRNAs. The ALKBH1-catalyzed demethylation of the target tRNAs results in attenuated translation initiation and decreased usage of tRNAs in protein synthesis. This process is dynamic and responds to glucose availability to affect translation. Our results uncover reversible methylation of tRNA as a new mechanism of post-transcriptional gene expression regulation.


Subject(s)
AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Gene Expression Regulation , Protein Biosynthesis/genetics , RNA, Transfer/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , Glucose/deficiency , HeLa Cells , Humans , Methylation , Polyribosomes/metabolism
3.
Mol Cell ; 71(2): 306-318.e7, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30017583

ABSTRACT

DNA N6-methyladenine (6mA) modification is the most prevalent DNA modification in prokaryotes, but whether it exists in human cells and whether it plays a role in human diseases remain enigmatic. Here, we showed that 6mA is extensively present in the human genome, and we cataloged 881,240 6mA sites accounting for Ć¢ĀˆĀ¼0.051% of the total adenines. [G/C]AGG[C/T] was the most significantly associated motif with 6mA modification. 6mA sites were enriched in the coding regions and mark actively transcribed genes in human cells. DNA 6mA and N6-demethyladenine modification in the human genome were mediated by methyltransferase N6AMT1 and demethylase ALKBH1, respectively. The abundance of 6mA was significantly lower in cancers, accompanied by decreased N6AMT1 and increased ALKBH1 levels, and downregulation of 6mA modification levels promoted tumorigenesis. Collectively, our results demonstrate that DNA 6mA modification is extensively present in human cells and the decrease of genomic DNA 6mA promotes human tumorigenesis.


Subject(s)
Adenine/analogs & derivatives , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Genome, Human , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism , Adenine/metabolism , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , Animals , Carcinogenesis/genetics , DNA/genetics , DNA Methylation , Heterografts , Humans , Mice , Mice, Nude , Site-Specific DNA-Methyltransferase (Adenine-Specific)/genetics
4.
Cell Mol Life Sci ; 81(1): 130, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38472355

ABSTRACT

ALKBH1 is a typical demethylase of nucleic acids, which is correlated with multiple types of biological processes and human diseases. Recent studies are focused on the demethylation of ALKBH1, but little is known about its non-demethylase function. Here, we demonstrate that ALKBH1 regulates the glycolysis process through HIF-1α signaling in a demethylase-independent manner. We observed that depletion of ALKBH1 inhibits glycolysis flux and extracellular acidification, which is attributable to reduced HIF-1α protein levels, and it can be rescued by reintroducing HIF-1α. Mechanistically, ALKBH1 knockdown enhances chaperone-mediated autophagy (CMA)-mediated HIF-1α degradation by facilitating the interaction between HIF-1α and LAMP2A. Furthermore, we identify that ALKBH1 competitively binds to the OST48, resulting in compromised structural integrity of oligosaccharyltransferase (OST) complex and subsequent defective N-glycosylation of LAMPs, particularly LAMP2A. Abnormal glycosylation of LAMP2A disrupts lysosomal homeostasis and hinders the efficient degradation of HIF-1α through CMA. Moreover, NGI-1, a small-molecule inhibitor that selectively targets the OST complex, could inhibit the glycosylation of LAMPs caused by ALKBH1 silencing, leading to impaired CMA activity and disruption of lysosomal homeostasis. In conclusion, we have revealed a non-demethylation role of ALKBH1 in regulating N-glycosylation of LAMPs by interacting with OST subunits and CMA-mediated degradation of HIF-1α.


Subject(s)
Autophagy , Signal Transduction , Humans , Lysosomal-Associated Membrane Protein 2/metabolism , Glycosylation , Glycolysis , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism
5.
Cell Commun Signal ; 22(1): 79, 2024 01 30.
Article in English | MEDLINE | ID: mdl-38291517

ABSTRACT

N1-methyladenosine (m1A) is a post-transcriptionally modified RNA molecule that plays a pivotal role in the regulation of various biological functions and activities. Especially in cancer cell invasion, proliferation and cell cycle regulation. Over recent years, there has been a burgeoning interest in investigating the m1A modification of RNA. Most studies have focused on the regulation of m1A in cancer enrichment areas and different regions. This review provides a comprehensive overview of the methodologies employed for the detection of m1A modification. Furthermore, this review delves into the key players in m1A modification, known as the "writers," "erasers," and "readers." m1A modification is modified by the m1A methyltransferases, or writers, such as TRMT6, TRMT61A, TRMT61B, TRMT10C, NML, and, removed by the demethylases, or erasers, including FTO and ALKBH1, ALKBH3. It is recognized by m1A-binding proteins YTHDF1, TYHDF2, TYHDF3, and TYHDC1, also known as "readers". Additionally, we explore the intricate relationship between m1A modification and its regulators and their implications for the development and progression of specific types of cancer, we discuss how m1A modification can potentially facilitate the discovery of novel approaches for cancer diagnosis, treatment, and prognosis. Our summary of m1A methylated adenosine modification detection methods and regulatory mechanisms in various cancers provides useful insights for cancer diagnosis, treatment, and prognosis. Video Abstract.


Subject(s)
Neoplasms , Humans , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/metabolism , RNA/genetics , RNA/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Methylation , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , AlkB Homolog 3, Alpha-Ketoglutarate-Dependent Dioxygenase/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism
6.
J Biol Chem ; 298(1): 101499, 2022 01.
Article in English | MEDLINE | ID: mdl-34922943

ABSTRACT

DNA N6-adenine methylation (6mA), as a novel adenine modification existing in eukaryotes, shows essential functions in embryogenesis and mitochondrial transcriptions. ALKBH1 is a demethylase of 6mA and plays critical roles in osteogenesis, tumorigenesis, and adaptation to stress. However, the integrated biological functions of ALKBH1 still require further exploration. Here, we demonstrate that knockdown of ALKBH1 inhibits adipogenic differentiation in both human mesenchymal stem cells (hMSCs) and 3T3-L1 preadipocytes, while overexpression of ALKBH1 leads to increased adipogenesis. Using a combination of RNA-seq and N6-mA-DNA-IP-seq analyses, we identify hypoxia-inducible factor-1 (HIF-1) signaling as a crucial downstream target of ALKBH1 activity. Depletion of ALKBH1 leads to hypermethylation of both HIF-1α and its downstream target GYS1. Simultaneous overexpression of HIF-1α and GYS1 restores the adipogenic commitment of ALKBH1-deficient cells. Taken together, our data indicate that ALKBH1 is indispensable for adipogenic differentiation, revealing a novel epigenetic mechanism that regulates adipogenesis.


Subject(s)
Adipogenesis , AlkB Homolog 1, Histone H2a Dioxygenase , Hypoxia-Inducible Factor 1 , Osteogenesis , 3T3-L1 Cells , Adenine/metabolism , Adipocytes/cytology , Adipocytes/metabolism , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Animals , Cell Differentiation , DNA/metabolism , DNA Methylation , Humans , Hypoxia-Inducible Factor 1/metabolism , Mice
7.
Eur J Clin Invest ; 53(7): e13986, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36920340

ABSTRACT

BACKGROUND: Renal cell carcinoma (RCC) accounts for approximately 4% of all adult malignancies with high mortality worldwide. Although conventional chemotherapy and radiotherapy treatment has been applied for RCC in clinic, the mortality rate of patients is increasing each year, and patients with metastatic RCC are still suffering from poor prognosis. Thus, further investigation of the molecular mechanisms responsible for the development and progression of RCC is of particular importance. METHODS: Total of 10 pairs of RCC tissues and adjacent nontumor tissues were collected for examination of ALKBH1 and GPR137 expression. The correlations between ALKBH1 and GPR137 expression in RCC patient were assessed by GEPIA online tool and were analyzed using auto best cutoff. The human RCC cell lines Caki-1, 786-O, ACHN, Osrc2, A498, and 769-P, were used for mechanistic investigation. RESULTS: Here, we report that the expression of AlkB homologue 1 (ALKBH1) is upregulated in RCC tissues, which is correlated with G-protein-coupled receptor 137 (GPR137) expression. The elevated expression of ALKBH1 is associated with RCC cell malignant characteristics, including cell proliferation and movement (migration and invasion). Mechanistic investigation further reveals that ALKBH1 reduces m6 A levels of GPR137 mRNA in RCC cells, which upregulates GPR137 mRNA levels, resulting in the increased GPR137 protein expression subsequently and the enhanced RCC cell biological actions consequently. In contrast, the suppression of GPR137 effectively alleviates the ALKBH1-induced malignancies of RCC cells. CONCLUSION: Our results indicate that ALKBH1-GPR137 axis might be used as a potential therapeutic target in RCC, contributing to finding new prognostic biomarkers for RCC at an early stage.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Adult , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Proliferation/genetics , RNA, Messenger , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism
8.
Exp Cell Res ; 400(2): 112492, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33529710

ABSTRACT

DNA N6-methyladenine (N6-mA) was recently recognized as a new epigenetic modification in mammalian genome, and ALKBH1 was discovered as its demethylase. Knock-out mice studies revealed that ALKBH1 was indispensable for normal embryonic development. However, the function of ALKBH1 in myogenesis is largely unknown. In this study, we found that N6-mA showed a steady increase, going along with a strong decrease of ALKBH1 during skeletal muscle development. Our results also showed that ALKBH1 enhanced proliferation and inhibited differentiation of C2C12Ā cells. Genome-wide transcriptome analysis and reporter assays further revealed that ALKBH1 accomplished the differentiation inhibiting function by regulating a core set of genes and multiple signaling pathways, including increasing chemokine (C-X-C motif) ligand 14 (CXCL14) and activating ERK signaling. Taken together, our results demonstrated that ALKBH1 is critical for the myogenic differentiation of C2C12Ā cells, and suggested that N6-mA might be a new epigenetic mechanism for the regulation of myogenesis.


Subject(s)
Adenine/analogs & derivatives , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Cell Differentiation , Epigenesis, Genetic , Muscle Development , Muscle, Skeletal/pathology , Myoblasts/pathology , Adenine/chemistry , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , Animals , DNA Methylation , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Myoblasts/metabolism
9.
J Cell Sci ; 132(19)2019 10 01.
Article in English | MEDLINE | ID: mdl-31434717

ABSTRACT

The Fe(II) and 2-oxoglutarate-dependent oxygenase Alkb homologue 1 (Alkbh1) has been shown to act on a wide range of substrates, like DNA, tRNA and histones. Thereby different enzymatic activities have been identified including, among others, demethylation of N3-methylcytosine (m3C) in RNA- and single-stranded DNA oligonucleotides, demethylation of N1-methyladenosine (m1A) in tRNA or formation of 5-formyl cytosine (f5C) in tRNA. In accordance with the different substrates, Alkbh1 has also been proposed to reside in distinct cellular compartments in human and mouse cells, including the nucleus, cytoplasm and mitochondria. Here, we describe further evidence for a role of human Alkbh1 in regulation of mitochondrial protein biogenesis, including visualizing localization of Alkbh1 into mitochondrial RNA granules with super-resolution 3D SIM microscopy. Electron microscopy and high-resolution respirometry analyses revealed an impact of Alkbh1 level on mitochondrial respiration, but not on mitochondrial structure. Downregulation of Alkbh1 impacts cell growth in HeLa cells and delays development in Caenorhabditis elegans, where the mitochondrial role of Alkbh1 seems to be conserved. Alkbh1 knockdown, but not Alkbh7 knockdown, triggers the mitochondrial unfolded protein response (UPRmt) in C. elegans.


Subject(s)
AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Mitochondria/metabolism , RNA, Mitochondrial/metabolism , A549 Cells , AlkB Enzymes/genetics , AlkB Enzymes/metabolism , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , Animals , Caenorhabditis elegans , Cell Nucleus/metabolism , Cytoplasm/metabolism , Electrophoresis, Polyacrylamide Gel , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , Mice , Microscopy, Electron , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxygen Consumption/physiology , Peptide Elongation Factor Tu/genetics , Peptide Elongation Factor Tu/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology
10.
Exp Mol Pathol ; 115: 104449, 2020 08.
Article in English | MEDLINE | ID: mdl-32380054

ABSTRACT

OBJECTIVE: To uncover the role of microRNA-339-5p (miRNA-339-5p) in the development of gastric cancer (GC) and its possible molecular mechanism. METHODS: Differential expressions of miRNA-339-5p in GC and adjacent normal tissues were detected. The relationship between miRNA-339-5p level and clinical features in GC patients was analyzed. Proliferative and migratory changes in BGC-823 and SGC-7901 cells overexpressing miRNA-339-5p were examined. Finally, luciferase assay and rescue experiments were conducted to explore the regulatory mechanism of miRNA-339-5p in its downstream gene ALKBH1, and their interaction in the development of GC. RESULTS: MiRNA-339-5p was downregulated in GC tissues. Lowly expressed miRNA-339-5p was unfavorable to prognosis in GC because of high rates of lymphatic metastasis and distant metastasis. Overexpression of miRNA-339-5p markedly reduced proliferative and migratory abilities in GC cells. ALKBH1 was identified to be the downstream gene of miRNA-339-5p. In GC tissues, ALKBH1 was upregulated and negatively correlated to miRNA-339-5p level. Overexpression of ALKBH1 was able to reverse the inhibitory effects of overexpressed miRNA-339-5p on proliferative and migratory abilities in GC. CONCLUSIONS: Lowly expressed miRNA-339-5p is closely related to metastasis and poor prognosis in GC patients. MiRNA-339-5p suppresses the malignant development of GC by negatively regulating ALKBH1.


Subject(s)
AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , MicroRNAs/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Middle Aged
11.
RNA Biol ; 17(8): 1092-1103, 2020 08.
Article in English | MEDLINE | ID: mdl-32521209

ABSTRACT

tiRNAs are small non-coding RNAs produced when tRNA is cleaved under stress. tRNA methylation modifications has emerged in recent years as important regulators for tRNA structural stability and sensitivity to cleavage and tiRNA generation during stress, however, the specificity and higher regulation of such a process is not fully understood. Alkbh1 is a m1A demethylase that leads to destabilization of tRNA and enhanced tRNA cleavage. We examined the impact of Alkbh1 targeting via gene knockdown or overexpression on B35 rat neuroblastoma cell line fate following stresses and on tRNA cleavage. We show that Alkbh1 impact on cell fate and tRNA cleavage is a stress specific process that is impacted by the demethylating capacity of the cellular stress in question. We also show that not all tRNAs are cleaved equally following Alkbh1 manipulation and stress, and that Alkbh1 KD fails to rescue tRNAs from cleavage following demethylating stresses. These findings shed a light on the specificity and higher regulation of tRNA cleavage and should act as a guide for future work exploring the utility of Alkbh1 as a therapeutic target for cancers or ischaemic insult.


Subject(s)
AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , RNA Cleavage , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA, Untranslated/genetics , Stress, Physiological/genetics , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , DNA Methylation , Gene Knockdown Techniques , Humans , Methylation , Oxidative Stress , RNA Processing, Post-Transcriptional , Rats
12.
Nucleic Acids Res ; 46(22): 11659-11670, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30412255

ABSTRACT

N6-methyldeoxyadenosine (6mA) is a well-characterized DNA modification in prokaryotes but reports on its presence and function in mammals have been controversial. To address this issue, we established the capacity of 6mA-Crosslinking-Exonuclease-sequencing (6mACE-seq) to detect genome-wide 6mA at single-nucleotide-resolution, demonstrating this by accurately mapping 6mA in synthesized DNA and bacterial genomes. Using 6mACE-seq, we generated a human-genome-wide 6mA map that accurately reproduced known 6mA enrichment at active retrotransposons and revealed mitochondrial chromosome-wide 6mA clusters asymmetrically enriched on the heavy-strand. We identified a novel putative 6mA-binding protein in single-stranded DNA-binding protein 1 (SSBP1), a mitochondrial DNA (mtDNA) replication factor known to coat the heavy-strand, linking 6mA with the regulation of mtDNA replication. Finally, we characterized AlkB homologue 1 (ALKBH1) as a mitochondrial protein with 6mA demethylase activity and showed that its loss decreases mitochondrial oxidative phosphorylation. Our results show that 6mA clusters play a previously unappreciated role in regulating human mitochondrial function, despite 6mA being an uncommon DNA modification in the human genome.


Subject(s)
DNA, Mitochondrial/genetics , DNA-Binding Proteins/genetics , DNA/genetics , Deoxyadenosines/genetics , Genome, Mitochondrial , Mitochondrial Proteins/genetics , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Bacteriophage lambda/genetics , Bacteriophage lambda/metabolism , Base Sequence , Chromosome Mapping , DNA/metabolism , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Deoxyadenosines/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Exodeoxyribonucleases , HEK293 Cells , Humans , Mitochondrial Proteins/metabolism , Oxidative Phosphorylation , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Sequence Analysis, DNA , Viral Proteins/chemistry , Viral Proteins/metabolism
13.
Dig Dis Sci ; 64(6): 1503-1513, 2019 06.
Article in English | MEDLINE | ID: mdl-30637548

ABSTRACT

BACKGROUND: Reversible N6-methyladenosine (m6A) modifications in messenger RNAs can be categorized under the field of "RNA epigenetics." However, the potential role of m6A-related genes in gastric cancer (GC) prognosis has not been systematically researched. AIMS: This study was aimed at providing insights into the prognostic role of m6A-related gene expression, at both mRNA and protein levels. METHODS: Kaplan-Meier (KM) plotter database and The Cancer Genome Atlas (TCGA) database were used to explore the prognostic significance of individual m6A-related genes in overall survival (OS) and progression-free survival at the mRNA level. For independent validation, the protein level of genes significantly associated with prognosis in both databases was further detected in 450 paired GC and corresponding adjacent non-tumor tissues using tissue microarray (TMA)-based immunohistochemistry (IHC). The relationship between the FTO and ALKBH1 expression and the clinicopathological characteristics was explored. RESULTS: Among nine m6A-related genes, aberrantly high mRNA expression of FTO and ALKBH1 was associated with poor OS in the KM and TCGA cohorts. However, the TMA-IHC indicated that protein expression of FTO and ALKBH1 was markedly downregulated in GC tissues. A lower protein level of ALKBH1 was closely correlated with larger tumor sizes (≥ 5Ā cm) and more advanced TNM stages, while lower FTO protein expression was associated with shorter OS in GC patients. CONCLUSIONS: Aberrant expression of demethylase genes, FTO and ALKBH1, has a distinct prognostic value in GC patients, indicating that FTO and ALKBH1 may play vital roles in GC progression and metastasis.


Subject(s)
AlkB Homolog 1, Histone H2a Dioxygenase/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Biomarkers, Tumor/genetics , RNA, Messenger/genetics , Stomach Neoplasms/enzymology , Adenosine/analogs & derivatives , Adenosine/metabolism , Aged , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Biomarkers, Tumor/metabolism , Databases, Genetic , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Progression-Free Survival , RNA Processing, Post-Transcriptional , RNA Stability , RNA, Messenger/metabolism , Risk Factors , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/therapy , Time Factors
14.
Biochem Biophys Res Commun ; 495(1): 98-103, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29097205

ABSTRACT

Alkbh1 is a mammalian homolog of the Escherichia coli DNA repair enzyme AlkB, an Fe(II) and 2-oxoglutarate dependent dioxygenase that removes alkyl lesions from DNA bases. The human homolog ALKBH1 has been associated with six different enzymatic activities including DNA, mRNA, or tRNA hydroxylation, cleavage at abasic (AP) sites in DNA, as well as demethylation of histones. The reported cellular roles of this protein reflect the diverse enzymatic activities and include direct DNA repair, tRNA modification, and histone modification. We demonstrate that ALKBH1 produced in mammalian cells (ALKBH1293) is similar to the protein produced in bacteria (ALKBH1Ec) with regard to its m6A demethylase and AP lyase activities. In addition, we find that ALKBH1293 forms a covalent adduct with the 5' product of the lyase product in a manner analogous to ALKBH1Ec. Localization and subcellular fractionation studies with the endogenous protein in two human cell strains confirm that ALKBH1 is primarily in the mitochondria. Two strains of CRISPR/Cas9-created ALKBH1-deficient HEK293Ā cells showed increases in mtDNA copy number and mitochondrial dysfunction as revealed by growth measurements and citrate synthase activity assays.


Subject(s)
AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Mitochondria/metabolism , AlkB Homolog 1, Histone H2a Dioxygenase/deficiency , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , Cell Proliferation , DNA Adducts/chemistry , DNA Adducts/genetics , DNA Adducts/metabolism , DNA Copy Number Variations , DNA, Mitochondrial/chemistry , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , HEK293 Cells , Humans , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
15.
Biochemistry ; 56(13): 1899-1910, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28290676

ABSTRACT

Alkbh1 is one of nine mammalian homologues of Escherichia coli AlkB, a 2-oxoglutarate-dependent dioxygenase that catalyzes direct DNA repair by removing alkyl lesions from DNA. Six distinct enzymatic activities have been reported for Alkbh1, including hydroxylation of variously methylated DNA, mRNA, tRNA, or histone substrates along with the cleavage of DNA at apurinic/apyrimidinic (AP) sites followed by covalent attachment to the 5'-product. The studies described here extend the biochemical characterization of two of these enzymatic activities using human ALKBH1: the AP lyase and 6-methyl adenine DNA demethylase activities. The steady-state and single-turnover kinetic parameters for ALKBH1 cleavage of AP sites in DNA were determined and shown to be comparable to those of other AP lyases. The α,Ɵ-unsaturated aldehyde of the 5'-product arising from DNA cleavage reacts predominantly with C129 of ALKBH1, but secondary sites also generate covalent adducts. The 6-methyl adenine demethylase activity was examined with a newly developed assay using a methylation-sensitive restriction endonuclease, and the enzymatic rate was found to be very low. Indeed, the demethylase activity was less than half that of the AP lyase activity when ALKBH1 samples were assayed using identical buffer conditions. The two enzymatic activities were examined using a series of site-directed variant proteins, revealing the presence of distinct but partially overlapping active sites for the two reactions. We postulate that the very low 6-methyl adenine oxygenase activity associated with ALKBH1 is unlikely to represent the major function of the enzyme in the cell, while the cellular role of the lyase activity (including its subsequent covalent attachment to DNA) remains uncertain.


Subject(s)
Adenine/chemistry , AlkB Homolog 1, Histone H2a Dioxygenase/chemistry , DNA/chemistry , Escherichia coli Proteins/chemistry , Mixed Function Oxygenases/chemistry , Oxidoreductases, O-Demethylating/chemistry , Adenine/metabolism , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Catalytic Domain , DNA/genetics , DNA/metabolism , DNA Adducts , Enzyme Assays , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression , Humans , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Models, Molecular , Oligonucleotides/chemistry , Oligonucleotides/metabolism , Oxidoreductases, O-Demethylating/genetics , Oxidoreductases, O-Demethylating/metabolism , Protein Structure, Secondary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Species Specificity
16.
Life Sci ; 338: 122372, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38135116

ABSTRACT

5-Methylcytosine (m5C) methylation is present in almost all types of RNA as an essential epigenetic modification. It is dynamically modulated by its associated enzymes, including m5C methyltransferases (NSUN, DNMT and TRDMT family members), demethylases (TET family and ALKBH1) and binding proteins (YTHDF2, ALYREF and YBX1). Among them, aberrant expression of the RNA-binding protein ALYREF can facilitate a variety of malignant phenotypes such as maintenance of proliferation, malignant heterogeneity, metastasis, and drug resistance to cell death through different regulatory mechanisms, including pre-mRNA processing, mRNA stability, and nuclear-cytoplasmic shuttling. The induction of these cellular processes by ALYREF results in treatment resistance and poor outcomes for patients. However, there are currently few reports of clinical applications or drug trials related to ALYREF. In addition, the looming observations on the role of ALYREF in the mechanisms of carcinogenesis and disease prognosis have triggered considerable interest, but critical evidence is not available. For example, animal experiments and ALYREF small molecule inhibitor trials. In this review, we, therefore, revisit the literature on ALYREF and highlight its importance as a prognostic biomarker for early prevention and as a therapeutic target.


Subject(s)
Neoplasms , Nuclear Proteins , Animals , Humans , Nuclear Proteins/metabolism , Transcription Factors/metabolism , RNA Processing, Post-Transcriptional , Neoplasms/drug therapy , Neoplasms/genetics , Biomarkers/metabolism , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , RNA-Binding Proteins/metabolism
17.
J Med Chem ; 67(17): 15456-15475, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39225755

ABSTRACT

DNA N6-methyladenine (6mA) demethylase ALKBH1 plays an important role in various cellular processes. Dysregulation of ALKBH1 is associated with the development of some cancer types, including gastric cancer, implicating a potential therapeutic target. However, there is still a lack of potent ALKBH1 inhibitors. Herein, we report the discovery of a highly potent ALKBH1 inhibitor, 1H-pyrazole-4-carboxylic acid derivative 29. The structure-activity relationship of this series of compounds was also discussed. Because of the poor cell membrane permeability of 29, we prepared a prodrug of 29 (29E), which showed excellent cellular activities. In gastric cancer cell lines HGC27 and AGS, 29E treatment significantly increased the abundance of 6mA, inhibited cell viability, and upregulated the AMP-activated protein kinase (AMPK) signaling pathway. In addition, the hydrolysis product 29 showed high exposure in mice after administration of 29E. Collectively, this research provides a new potent ALKBH1 inhibitor, which could serve as a lead compound for subsequent drug development.


Subject(s)
AlkB Homolog 1, Histone H2a Dioxygenase , Antineoplastic Agents , Enzyme Inhibitors , Pyrazoles , Stomach Neoplasms , Humans , Structure-Activity Relationship , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Animals , Pyrazoles/pharmacology , Pyrazoles/chemistry , Pyrazoles/chemical synthesis , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Cell Line, Tumor , Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , Carboxylic Acids/chemical synthesis , Cell Proliferation/drug effects , Molecular Structure , Molecular Docking Simulation , Mice, Nude , Mice, Inbred BALB C
18.
J Diabetes ; 15(4): 349-361, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36891946

ABSTRACT

OBJECTIVE: To clarify the expression of N6-methyladenosine (m6 A) modulators involved in the pathogenesis of type 2 diabetes mellitus (T2DM). We further explored the association of serum insulin-like growth factor 2 mRNA-binding proteins 3 (IGF2BP3) levels and odds of T2DM in a high-risk population. METHODS: The gene expression data set GSE25724 was obtained from the Gene Expression Omnibus, and a cluster heatmap was generated by using the R package ComplexHeatmap. Differential expression analysis for 13 m6 A RNA methylation regulators between nondiabetic controls and T2DM subjects was performed using an unpaired t test. A cross-sectional design, including 393 subjects (131 patients with newly diagnosed T2DM, 131 age- and sex-matched subjects with prediabetes, and 131 healthy controls), was carried out. The associations between serum IGF2BP3 concentrations and T2DM were modeled by restricted cubic spline and logistic regression models. RESULTS: Two upregulated (IGF2BP2 and IGF2BP3) and 5 downregulated (methyltransferase-like 3 [METTL3], alkylation repair homolog protein 1 [ALKBH1], YTH domain family 2 [YTHDF2], YTHDF3, and heterogeneous nuclear ribonucleoprotein [HNRNPC]) m6 A-related genes were found in islet samples of T2DM patients. A U-shaped association existed between serum IGF2BP3 levels and odds of T2DM according to cubic natural spline analysis models, after adjustment for body mass index, waist circumference, diastolic blood pressure, total cholesterol, and triglyeride. Multivariate logistic regression showed that progressively higher odds of T2DM were observed when serum IGF2BP3 levels were below 0.62 ng/mL (odds ratio 3.03 [95% confidence interval 1.23-7.47]) in model 4. CONCLUSION: Seven significantly altered m6 A RNA methylation genes were identified in T2DM. There was a U-shaped association between serum IGF2BP3 levels and odds of T2DM in the general Chinese adult population. This study provides important evidence for further examination of the role of m6 A RNA methylation, especially serum IGF2BP3 in T2DM risk assessment.


Subject(s)
Diabetes Mellitus, Type 2 , RNA-Binding Proteins , Adult , Humans , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Cross-Sectional Studies , East Asian People , Methyltransferases/genetics , Methyltransferases/metabolism , Risk Factors , RNA-Binding Proteins/blood
19.
Genome Biol ; 24(1): 198, 2023 08 30.
Article in English | MEDLINE | ID: mdl-37649077

ABSTRACT

BACKGROUND: The Fe (II)- and α-ketoglutarate-dependent AlkB family dioxygenases are implicated in nucleotide demethylation. AlkB homolog1 (ALKBH1) is shown to demethylate DNA adenine methylation (6mA) preferentially from single-stranded or unpaired DNA, while its demethylase activity and function in the chromatin context are unclear. RESULTS: Here, we find that loss-of-function of the rice ALKBH1 gene leads to increased 6mA in the R-loop regions of the genome but has a limited effect on the overall 6mA level. However, in the context of mixed tissues, rather than on individual loci, the ALKBH1 mutation or overexpression mainly affects the expression of genes with a specific combination of chromatin modifications in the body region marked with H3K4me3 and H3K27me3 but depleted of DNA CG methylation. In the similar context of mixed tissues, further analysis reveals that the ALKBH1 protein preferentially binds to genes marked by the chromatin signature and has a function to maintain a high H3K4me3/H3K27me3 ratio by impairing the binding of Polycomb repressive complex 2 (PRC2) to the targets, which is required for both the basal and stress-induced expression of the genes. CONCLUSION: Our findings unravel a function of ALKBH1 to control the balance between the antagonistic histone methylations for gene activity and provide insight into the regulatory mechanism of PRC2-mediated H3K27me3 deposition within the gene body region.


Subject(s)
Oryza , Protein Binding , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Oryza/enzymology , Oryza/genetics , Oryza/growth & development , Mutation , Histones/metabolism , Chromatin
20.
Mol Oncol ; 17(2): 344-364, 2023 02.
Article in English | MEDLINE | ID: mdl-36550779

ABSTRACT

Colorectal cancer (CRC) is one of the most common malignancies, and the main cause of death from CRC is tumor metastasis. m1 A RNA modification plays critical role in many biological processes. However, the role of m1 A modification in CRC remains unclear. Here, we find that the m1 A demethylase alkB homolog 1, histone H2A dioxygenase (ALKBH1) is overexpressed in CRC and is associated with metastasis and poor prognosis. Upregulation of ALKBH1 expression promotes CRC metastasis in vitro and in vivo. Mechanistically, knockdown of ALKBH1 results in a decrease in methyltransferase 3, N6-adenosine-methyltransferase complex catalytic subunit (METTL3) expression, probably due to m1 A modification of METTL3 mRNA, followed by m6 A demethylation of SMAD family member 7 (SMAD7) mRNA. In addition, downregulation of SMAD7 establishes an aggressive phenotype. More importantly, the cell migration and invasion defects caused by ALKBH1 depletion or METTL3 depletion are significantly reversed by SMAD7 silencing. Considering these results collectively, we propose that ALKBH1 promotes CRC metastasis by destabilizing SMAD7 through METTL3.


Subject(s)
Colorectal Neoplasms , Methyltransferases , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Up-Regulation , Demethylation , Colorectal Neoplasms/pathology , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Smad7 Protein/genetics , Smad7 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL