Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 22255, 2023 12 14.
Article in English | MEDLINE | ID: mdl-38097707

ABSTRACT

Cisplatin (CP) induces acute kidney injury (AKI) whereby proximal tubules undergo regulated necrosis. Repair is almost complete after a single dose. We now demonstrate a role for Apolipoprotein B mRNA editing enzyme, catalytic polypeptide 1 (Apobec-1) that is prominently expressed at the interface between acute and chronic kidney injury (CKD), in the recovery from AKI. Apobec-1 knockout (KO) mice exhibited greater mortality than in wild type (WT) and more severe AKI in both CP- and unilateral ischemia reperfusion (IR) with nephrectomy. Specifically, plasma creatinine (pCr) 2.6 ± 0.70 mg/dL for KO, n = 10 and 0.16 ± 0.02 for WT, n = 6, p < 0.0001 in CP model and 1.34 ± 0.22 mg/dL vs 0.75 ± 0.06, n = 5, p < 0.05 in IR model. The kidneys of Apobec-1 KO mice showed increased necrosis, increased expression of KIM-1, NGAL, RIPK1, ASCL4 and increased lipid accumulation compared to WT kidneys (p < 0.01). Neutrophils and activated T cells were both increased, while macrophages were reduced in kidneys of Apobec-1 KO animals. Overexpression of Apobec-1 in mouse proximal tubule cells protected against CP-induced cytotoxicity. These findings suggest that Apobec-1 mediates critical pro-survival responses to renal injury and increasing Apobec-1 expression could be an effective strategy to mitigate AKI.


Subject(s)
Acute Kidney Injury , Reperfusion Injury , Mice , Animals , APOBEC-1 Deaminase/metabolism , Cisplatin/adverse effects , Cisplatin/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Kidney/metabolism , Necrosis/metabolism , Mice, Knockout , Reperfusion Injury/metabolism , Mice, Inbred C57BL
2.
Sci Adv ; 9(35): eadj1568, 2023 09.
Article in English | MEDLINE | ID: mdl-37647411

ABSTRACT

CRISPR nucleases generate a broad spectrum of mutations that includes undesired editing outcomes. Here, we develop optimized C-to-T base editing systems for the generation of precise loss- or gain-of-function alleles in Drosophila and identify temperature as a crucial parameter for efficiency. We find that a variant of the widely used APOBEC1 deaminase has attenuated activity at 18° to 29°C and shows considerable dose-dependent toxicity. In contrast, the temperature-tolerant evoCDA1 domain mediates editing of typically more than 90% of alleles and is substantially better tolerated. Furthermore, formation of undesired mutations is exceptionally rare in Drosophila compared to other species. The predictable editing outcome, high efficiency, and product purity enables near homogeneous induction of STOP codons or alleles encoding protein variants in vivo. Last, we demonstrate how optimized expression enables conditional base editing in marked cell populations. This work substantially facilitates creation of precise alleles in Drosophila and provides key design parameters for developing efficient base editing systems in other ectothermic species.


Subject(s)
Drosophila , Gene Editing , Drosophila/genetics , Gene Editing/methods , Animals , APOBEC-1 Deaminase/genetics , APOBEC-1 Deaminase/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Silencing
3.
J Mol Biol ; 435(24)2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38708190

ABSTRACT

Cytidine (C) to Uridine (U) RNA editing is a post-transcription modification that is involved in diverse biological processes. APOBEC1 (A1) catalyzes the conversion of C-to-U in RNA, which is important in regulating cholesterol metabolism through its editing activity on ApoB mRNA. However, A1 requires a cofactor to form an "editosome" for RNA editing activity. A1CF and RBM47, both RNA-binding proteins, have been identified as cofactors that pair with A1 to form editosomes and edit ApoB mRNA and other cellular RNAs. SYNCRIP is another RNA-binding protein that has been reported as a potential regulator of A1, although it is not directly involved in A1 RNA editing activity. Here, we describe the identification and characterization of a novel cofactor, RBM46 (RNA-Binding-Motif-protein-46), that can facilitate A1 to perform C-to-U editing on ApoB mRNA. Additionally, using the low-error circular RNA sequencing technique, we identified novel cellular RNA targets for the A1/RBM46 editosome. Our findings provide further insight into the complex regulatory network of RNA editing and the potential new function of A1 with its cofactors.


Subject(s)
APOBEC-1 Deaminase , RNA Editing , RNA-Binding Proteins , Uridine , APOBEC-1 Deaminase/metabolism , APOBEC-1 Deaminase/genetics , Humans , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Uridine/metabolism , Uridine/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , HEK293 Cells , Apolipoproteins B/metabolism , Apolipoproteins B/genetics , Cytidine/metabolism , Cytidine/genetics
4.
BMC Bioinformatics ; 23(1): 312, 2022 Aug 02.
Article in English | MEDLINE | ID: mdl-35918642

ABSTRACT

Although there have been some recent cell and animal experiments indicating that expression of the gene encoding apolipoprotein B mRNA editing enzyme catalytic subunit 3B (APOBEC3B) is closely related to cancer, it still lacks pan-cancer analysis. Here we analyzed the potential carcinogenic role of APOBEC3B in 33 tumors based on The Cancer Genome Atlas (TCGA). APOBEC3B was highly expressed in most tumors and weakly expressed in a few. Differences in expression level were significantly correlated with the pathological tumor stage and prognosis of affected patients. The high-frequency APOBEC3B changes were principally mutations and amplifications in some tumors, such as uterine corpus endometrial carcinomas or cutaneous melanomas. In testicular germ cell tumors and invasive breast carcinomas, APOBEC3B expression and CD8+ T lymphocyte counts were correlated. In other cancers, such as human papilloma virus (HPV)-related head and neck squamous cell carcinomas or esophageal adenocarcinomas, there was also cancer-associated fibroblast infiltration. The APOBEC3B enzyme acts in the mitochondrial respiratory electron transport chain and in oxidative phosphorylation. This first pan-cancer study provides a comprehensive understanding of the multiple roles of APOBEC3B in different tumor types.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , APOBEC-1 Deaminase/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Catalytic Domain , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Esophageal Neoplasms/genetics , Humans , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism
5.
Mol Cell Biochem ; 476(12): 4493-4505, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34499322

ABSTRACT

RNA-binding proteins (RBPs) are critical players in the post-transcriptional regulation of gene expression and are associated with each event in RNA metabolism. The term 'RNA-binding motif' (RBM) is assigned to novel RBPs with one or more RNA recognition motif (RRM) domains that are mainly involved in the nuclear processing of RNAs. RBM47 is a novel RBP conserved in vertebrates with three RRM domains whose contributions to various aspects of cellular functions are as yet emerging. Loss of RBM47 function affects head morphogenesis in zebrafish embryos and leads to perinatal lethality in mouse embryos, thereby assigning it to be an essential gene in early development of vertebrates. Its function as an essential cofactor for APOBEC1 in C to U RNA editing of several targets through substitution for A1CF in the A1CF-APOBEC1 editosome, established a new paradigm in the field. Recent advances in the understanding of its involvement in cancer progression assigned RBM47 to be a tumor suppressor that acts by inhibiting EMT and Wnt/[Formula: see text]-catenin signaling through post-transcriptional regulation. RBM47 is also required to maintain immune homeostasis, which adds another facet to its regulatory role in cellular functions. Here, we review the emerging roles of RBM47 in various biological contexts and discuss the current gaps in our knowledge alongside future perspectives for the field.


Subject(s)
APOBEC-1 Deaminase/metabolism , Neoplasms/pathology , RNA Editing , RNA-Binding Proteins/metabolism , Vertebrates/growth & development , APOBEC-1 Deaminase/genetics , Animals , Humans , Neoplasms/genetics , Neoplasms/metabolism , RNA-Binding Proteins/genetics
6.
Viruses ; 13(8)2021 08 15.
Article in English | MEDLINE | ID: mdl-34452478

ABSTRACT

The apolipoprotein B mRNA editing enzyme, catalytic polypeptide (APOBEC) enzyme family in humans has 11 members with diverse functions in metabolism and immunity [...].


Subject(s)
APOBEC-1 Deaminase/genetics , DNA Viruses/immunology , Immunity, Innate , APOBEC-1 Deaminase/classification , APOBEC-1 Deaminase/metabolism , Animals , DNA Viruses/metabolism , Humans , Mice , RNA Editing
7.
Nucleic Acids Res ; 49(16): e95, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34197596

ABSTRACT

The SNAP-ADAR tool enables precise and efficient A-to-I RNA editing in a guideRNA-dependent manner by applying the self-labeling SNAP-tag enzyme to generate RNA-guided editases in cell culture. Here, we extend this platform by combining the SNAP-tagged tool with further effectors steered by the orthogonal HALO-tag. Due to their small size (ca. 2 kb), both effectors are readily integrated into one genomic locus. We demonstrate selective and concurrent recruitment of ADAR1 and ADAR2 deaminase activity for optimal editing with extended substrate scope and moderate global off-target effects. Furthermore, we combine the recruitment of ADAR1 and APOBEC1 deaminase activity to achieve selective and concurrent A-to-I and C-to-U RNA base editing of endogenous transcripts inside living cells, again with moderate global off-target effects. The platform should be readily transferable to further epitranscriptomic writers and erasers to manipulate epitranscriptomic marks in a programmable way with high molecular precision.


Subject(s)
Gene Editing/methods , RNA Editing , APOBEC-1 Deaminase/metabolism , Adenosine Deaminase/metabolism , Cell Line , Fluorescent Dyes/chemistry , Humans
8.
In Vitro Cell Dev Biol Anim ; 57(5): 501-509, 2021 May.
Article in English | MEDLINE | ID: mdl-34002283

ABSTRACT

Metastasis is inevitable in about 30% of patients with primary renal cell carcinoma after nephrectomy treatment. APOBEC1 complementation factor (A1CF), an RNA binding protein, participates in tumor progressions such as growth, apoptosis, differentiation, and invasion. Here, we explored biological functions of A1CF and provided a new insight into renal cell carcinoma metastasis. Wound healing assay was conducted to detect migration in A1CF overexpression and knockdown stable cell lines. Quantitative PCR and western blot assays were utilized to test transcriptional and translation levels of A1CF and SMAD3 in A1CF overexpression and knockdown renal carcinoma cells. Nuclear and cytoplasmic protein separation assays were conducted to evaluate the subcellular distribution of A1CF and SMAD3. Immunoprecipitation assay was conducted to detect the interaction between A1CF and SMAD3. Our study demonstrated A1CF overexpression facilitated cell migration in renal carcinoma cells. A1CF deficiency downregulated expression of SMAD3, Snail1, and N-cadherin. In addition, A1CF promoted nucleus translocation of SMAD3 and interacted with SMAD3. SMAD3 knockdown attenuated cell migration induced by A1CF overexpression. Our study suggested A1CF facilitated cell migration by promoting nucleus translocation of SMAD3 in renal cell carcinoma cells.


Subject(s)
APOBEC-1 Deaminase/metabolism , Active Transport, Cell Nucleus , Carcinoma, Renal Cell/pathology , Cell Movement , Kidney Neoplasms/pathology , Smad3 Protein/metabolism , Blotting, Western , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Immunoprecipitation , Kidney Neoplasms/metabolism , Polymerase Chain Reaction
9.
J Virol ; 95(13): e0017821, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33762419

ABSTRACT

As the hosts of lentiviruses, almost 40 species of felids (family Felidae) are distributed around the world, and more than 20 feline species test positive for feline immunodeficiency virus (FIV), a lineage of lentiviruses. These observations suggest that FIVs globally infected a variety of feline species through multiple cross-species transmission events during a million-year history. Cellular restriction factors potentially inhibit lentiviral replication and limit cross-species lentiviral transmission, and cellular APOBEC3 deaminases are known as a potent restriction factor. In contrast, lentiviruses have evolutionary-acquired viral infectivity factor (Vif) to neutralize the APOBEC3-mediated antiviral effect. Because the APOBEC3-Vif interaction is strictly specific for viruses and their hosts, a comprehensive investigation focusing on Vif-APOBEC3 interplay can provide clues that will elucidate the roles of this virus-host interplay on cross-species transmission of lentiviruses. Here, we performed a comprehensive investigation with 144 patterns of a round robin test using 18 feline APOBEC3Z3 genes, an antiviral APOBEC3 gene in felid, and 8 FIV Vifs and derived a matrix showing the interplay between feline APOBEC3Z3 and FIV Vif. We particularly focused on the interplay between the APOBEC3Z3 of three felids (domestic cat, ocelot, and Asian golden cat) and an FIV Vif (strain Petaluma), and revealed that residues 65 and 66 of the APOBEC3Z3 protein of multiple felids are responsible for the counteraction triggered by FIV Petaluma Vif. Altogether, our findings can be a clue to elucidate not only the scenarios of the cross-species transmissions of FIVs in felids but also the evolutionary interaction between mammals and lentiviruses. IMPORTANCE Most of the emergences of new virus infections originate from the cross-species transmission of viruses. The fact that some virus infections are strictly specific for the host species indicates that certain "species barriers" in the hosts restrict cross-species jump of viruses, while viruses have evolutionary acquired their own "arms" to overcome/antagonize/neutralize these hurdles. Therefore, understanding of the molecular mechanism leading to successful cross-species viral transmission is crucial for considering the menus of the emergence of novel pathogenic viruses. In the field of retrovirology, APOBEC3-Vif interaction is a well-studied example of the battles between hosts and viruses. Here, we determined the sequences of 11 novel feline APOBEC3Z3 genes and demonstrated that all 18 different feline APOBEC3Z3 proteins tested exhibit anti-feline immunodeficiency virus (FIV) activity. Our comprehensive investigation focusing on the interplay between feline APOBEC3 and FIV Vif can be a clue to elucidate the scenarios of the cross-species transmissions of FIVs in felids.


Subject(s)
APOBEC-1 Deaminase/metabolism , Gene Products, vif/metabolism , Immunodeficiency Virus, Feline/metabolism , Lentivirus Infections/transmission , Animals , Cats , Cell Line , HEK293 Cells , Host Specificity/physiology , Host-Pathogen Interactions/physiology , Humans , Lentivirus Infections/pathology , Panthera , Virus Replication/physiology
10.
Methods Mol Biol ; 2181: 69-81, 2021.
Article in English | MEDLINE | ID: mdl-32729075

ABSTRACT

APOBEC1 is a member of the AID/APOBECs, a group of deaminases responsible for the editing of C>U in both DNA and RNA. APOBEC1 is physiologically involved in C>U RNA editing: while hundreds of targets have been discovered in mice, in humans the only well-characterized target of APOBEC1 is the apolipoprotein B (ApoB) transcript. APOBEC1 edits a CAA codon into a stop codon, which causes the translation of a truncated form of ApoB. A number of assays have been developed to investigate this process. Early assays, poisoned primer extension and Sanger sequencing, have focused on accuracy and sensitivity but rely on extraction of the RNA from tissues and cells. More recently, the need to visualize the RNA editing process directly in live cells have led to the development of fluorescence-based tools. These assays detect RNA editing through reporters whose editing causes a change in cellular localization or a change in fluorescent properties. Here we review the available assays to quantify RNA editing, and we present the protocol for cytofluorimetric analysis using a double-fluorescent reporter.


Subject(s)
APOBEC-1 Deaminase/genetics , Computational Biology/methods , Cytidine/genetics , RNA Editing/genetics , RNA, Messenger/genetics , Subcellular Fractions/metabolism , Uridine/genetics , APOBEC-1 Deaminase/metabolism , Cytidine/chemistry , Genes, Reporter , HEK293 Cells , Hep G2 Cells , High-Throughput Nucleotide Sequencing , Humans , RNA, Messenger/metabolism , Uridine/chemistry
11.
Nat Biotechnol ; 39(1): 35-40, 2021 01.
Article in English | MEDLINE | ID: mdl-32690970

ABSTRACT

Current base editors (BEs) catalyze only base transitions (C to T and A to G) and cannot produce base transversions. Here we present BEs that cause C-to-A transversions in Escherichia coli and C-to-G transversions in mammalian cells. These glycosylase base editors (GBEs) consist of a Cas9 nickase, a cytidine deaminase and a uracil-DNA glycosylase (Ung). Ung excises the U base created by the deaminase, forming an apurinic/apyrimidinic (AP) site that initiates the DNA repair process. In E. coli, we used activation-induced cytidine deaminase (AID) to construct AID-nCas9-Ung and found that it converts C to A with an average editing specificity of 93.8% ± 4.8% and editing efficiency of 87.2% ± 6.9%. For use in mammalian cells, we replaced AID with rat APOBEC1 (APOBEC-nCas9-Ung). We tested APOBEC-nCas9-Ung at 30 endogenous sites, and we observed C-to-G conversions with a high editing specificity at the sixth position of the protospacer between 29.7% and 92.2% and an editing efficiency between 5.3% and 53.0%. APOBEC-nCas9-Ung supplements the current adenine and cytidine BEs (ABE and CBE, respectively) and could be used to target G/C disease-causing mutations.


Subject(s)
CRISPR-Cas Systems/genetics , Cytosine/metabolism , DNA Glycosylases , Gene Editing/methods , APOBEC-1 Deaminase/genetics , APOBEC-1 Deaminase/metabolism , Adenine/metabolism , Animals , Base Pairing/genetics , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Cytidine Deaminase , DNA Repair/genetics , Deoxyribonuclease I/genetics , Deoxyribonuclease I/metabolism , Escherichia coli/genetics , Guanine/metabolism , Rats , Uracil-DNA Glycosidase
12.
Nucleic Acids Res ; 49(1): 322-339, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33330905

ABSTRACT

Many APOBEC cytidine deaminase members are known to induce 'off-target' cytidine deaminations in 5'TC motifs in genomic DNA that contribute to cancer evolution. In this report, we characterized APOBEC1, which is a possible cancer related APOBEC since APOBEC1 mRNA is highly expressed in certain types of tumors, such as lung adenocarcinoma. We found a low level of APOBEC1-induced DNA damage, as measured by γH2AX foci, in genomic DNA of a lung cancer cell line that correlated to its inability to compete in vitro with replication protein A (RPA) for ssDNA. This suggests that RPA can act as a defense against off-target deamination for some APOBEC enzymes. Overall, the data support the model that the ability of an APOBEC to compete with RPA can better predict genomic damage than combined analysis of mRNA expression levels in tumors and analysis of mutation signatures.


Subject(s)
APOBEC-1 Deaminase/antagonists & inhibitors , DNA, Single-Stranded/metabolism , Neoplasm Proteins/metabolism , Replication Protein A/metabolism , APOBEC-1 Deaminase/metabolism , Binding, Competitive , Cell Line , Cell Line, Tumor , Cytidine/metabolism , DNA Damage , DNA Replication , DNA, Neoplasm/chemistry , DNA, Neoplasm/metabolism , DNA, Single-Stranded/chemistry , Deamination , Facilitated Diffusion , Histones/analysis , Humans , Lung/cytology , Lung/embryology , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/genetics , Neoplasms/pathology , Organ Specificity , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Recombinant Fusion Proteins/metabolism , Replication Protein A/genetics
13.
Nat Commun ; 11(1): 2052, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32345976

ABSTRACT

Cytosine base editors (CBEs) enable efficient, programmable reversion of T•A to C•G point mutations in the human genome. Recently, cytosine base editors with rAPOBEC1 were reported to induce unguided cytosine deamination in genomic DNA and cellular RNA. Here we report eight next-generation CBEs (BE4 with either RrA3F [wt, F130L], AmAPOBEC1, SsAPOBEC3B [wt, R54Q], or PpAPOBEC1 [wt, H122A, R33A]) that display comparable DNA on-target editing frequencies, whilst eliciting a 12- to 69-fold reduction in C-to-U edits in the transcriptome, and up to a 45-fold overall reduction in unguided off-target DNA deamination relative to BE4 containing rAPOBEC1. Further, no enrichment of genome-wide C•G to T•A edits are observed in mammalian cells following transfection of mRNA encoding five of these next-generation editors. Taken together, these next-generation CBEs represent a collection of base editing tools for applications in which minimized off-target and high on-target activity are required.


Subject(s)
Cytosine/metabolism , DNA/genetics , Gene Editing , RNA/genetics , APOBEC-1 Deaminase/metabolism , Cytosine Deaminase/metabolism , DNA Replication/genetics , Deamination , Genome , HEK293 Cells , Humans , Mutagenesis/genetics , Transcription, Genetic , Transcriptome/genetics
14.
Nat Commun ; 11(1): 1917, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32317634

ABSTRACT

The evolution and progression of multiple myeloma and its precursors over time is poorly understood. Here, we investigate the landscape and timing of mutational processes shaping multiple myeloma evolution in a large cohort of 89 whole genomes and 973 exomes. We identify eight processes, including a mutational signature caused by exposure to melphalan. Reconstructing the chronological activity of each mutational signature, we estimate that the initial transformation of a germinal center B-cell usually occurred during the first 2nd-3rd decades of life. We define four main patterns of activation-induced deaminase (AID) and apolipoprotein B mRNA editing catalytic polypeptide-like (APOBEC) mutagenesis over time, including a subset of patients with evidence of prolonged AID activity during the pre-malignant phase, indicating antigen-responsiveness and germinal center reentry. Our findings provide a framework to study the etiology of multiple myeloma and explore strategies for prevention and early detection.


Subject(s)
Gene Expression Regulation, Neoplastic , Multiple Myeloma/etiology , Multiple Myeloma/genetics , APOBEC-1 Deaminase/metabolism , Cytidine Deaminase/metabolism , DNA Mutational Analysis , Early Detection of Cancer , Exome , Genetics , Germinal Center/pathology , Humans , Linear Models , Minor Histocompatibility Antigens/metabolism , Mutation , Proteins/metabolism , RNA Editing , RNA, Messenger , Single-Cell Analysis
15.
J Med Virol ; 92(1): 53-61, 2020 01.
Article in English | MEDLINE | ID: mdl-31429946

ABSTRACT

Hepatitis B virus (HBV) DNA is vulnerable to editing by human apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like (APOBEC) cytidine deaminases. However, the distribution of APOBEC-induced mutations on HBV DNA is not well characterized. To this end, we obtained the HBV DNA sequence of HBV-infected individuals with and without hepatocellular carcinoma (HCC and non-HCC groups, respectively) from NCBI database and calculated the rapo values of APOBEC-induced TpCpW→TpKpW mutation prevalence in HBV DNA. The results showed that the APOBEC-induced mutations were mainly distributed in the minus strand of non-HCC-derived HBV DNA (rapo = 2.04), while the mutation on the plus-strand was weaker (rapo = 0.99). There were high APOBEC-induced mutation regions in the minus strand of HBV DNA 1 to 1000 nucleotides (nts) region and in the plus-strand of HBV DNA 1000 to 1500 nts region; the mutations in the 1 to 1000 nts region were mainly TpCpW→TpTpW mutation types (total T/G: 111/18) and a number of these were missense mutations (missense/synonymous: 35/94 in P gene, 17/15 in S gene, and 5/10 in X gene). The difference between minus to plus-strand rapo of HCC-derived HBV DNA (1.96) was greater than that of the non-HCC group (1.05). The minus-strand rapo of HCC-derived HBV DNA regions 1000 to1500nts and 1500 to 2000 nts (rapo = 4.2 and 4.2) was also higher than that of the same regions of non-HCC-derived HBV DNA (rapo = 1.2 and 1.1). Finally, the ratio of minus to plus-strand rapo was used to distinguish HCC-derived HBV DNA from non-HCC-derived HBV DNA. This study unraveled the distribution characteristics of APOBEC-induced mutations on double strands of HBV DNA from HCC and non-HCC samples. Our findings would help understand the mechanism of APOBECs on HBV DNA and may provide important insights for the screening of HCC.


Subject(s)
APOBEC-1 Deaminase/metabolism , DNA, Viral/genetics , Genotype , Hepatitis B virus/genetics , Mutation , Carcinoma, Hepatocellular/virology , Hepatitis B virus/classification , Humans , Liver Neoplasms/virology , Sequence Analysis, DNA
16.
BMC Genomics ; 20(1): 858, 2019 Nov 14.
Article in English | MEDLINE | ID: mdl-31726973

ABSTRACT

BACKGROUND: APOBEC1 (A1) enzymes are cytidine deaminases involved in RNA editing. In addition to this activity, a few A1 enzymes have been shown to be active on single stranded DNA. As two human ssDNA cytidine deaminases APOBEC3A (A3A), APOBEC3B (A3B) and related enzymes across the spectrum of placental mammals have been shown to introduce somatic mutations into nuclear DNA of cancer genomes, we explored the mutagenic threat of A1 cytidine deaminases to chromosomal DNA. RESULTS: Molecular cloning and expression of various A1 enzymes reveal that the cow, pig, dog, rabbit and mouse A1 have an intracellular ssDNA substrate specificity. However, among all the enzymes studied, mouse A1 appears to be singular, being able to introduce somatic mutations into nuclear DNA with a clear 5'TpC editing context, and to deaminate 5-methylcytidine substituted DNA which are characteristic features of the cancer related mammalian A3A and A3B enzymes. However, mouse A1 activity fails to elicit formation of double stranded DNA breaks, suggesting that mouse A1 possess an attenuated nuclear DNA mutator phenotype reminiscent of human A3B. CONCLUSIONS: At an experimental level mouse APOBEC1 is remarkable among 12 mammalian A1 enzymes in that it represents a source of somatic mutations in mouse genome, potentially fueling oncogenesis. While the order Rodentia is bereft of A3A and A3B like enzymes it seems that APOBEC1 may well substitute for it, albeit remaining much less active. This modifies the paradigm that APOBEC3 and AID enzymes are the sole endogenous mutator enzymes giving rise to off-target editing of mammalian genomes.


Subject(s)
APOBEC-1 Deaminase/metabolism , Chromosomes, Mammalian/genetics , Mutation , APOBEC-1 Deaminase/chemistry , APOBEC-1 Deaminase/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Breaks, Double-Stranded , DNA, Single-Stranded , Enzyme Activation , Gene Expression , Mice , Phylogeny , RNA Editing , Substrate Specificity
17.
Nat Commun ; 10(1): 3280, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31337756

ABSTRACT

Somatic mutations in the mitochondrial genome (mtDNA) have been linked to multiple disease conditions and to ageing itself. In Drosophila, knock-in of a proofreading deficient mtDNA polymerase (POLG) generates high levels of somatic point mutations and also small indels, but surprisingly limited impact on organismal longevity or fitness. Here we describe a new mtDNA mutator model based on a mitochondrially-targeted cytidine deaminase, APOBEC1. mito-APOBEC1 acts as a potent mutagen which exclusively induces C:G>T:A transitions with no indels or mtDNA depletion. In these flies, the presence of multiple non-synonymous substitutions, even at modest heteroplasmy, disrupts mitochondrial function and dramatically impacts organismal fitness. A detailed analysis of the mutation profile in the POLG and mito-APOBEC1 models reveals that mutation type (quality) rather than quantity is a critical factor in impacting organismal fitness. The specificity for transition mutations and the severe phenotypes make mito-APOBEC1 an excellent mtDNA mutator model for ageing research.


Subject(s)
APOBEC-1 Deaminase/physiology , DNA, Mitochondrial/chemistry , Drosophila/genetics , APOBEC-1 Deaminase/genetics , APOBEC-1 Deaminase/metabolism , Animals , Drosophila/physiology , Mitochondria/metabolism , Mitochondria/physiology , Models, Genetic , Mutation , Organisms, Genetically Modified
18.
Nature ; 569(7756): 433-437, 2019 05.
Article in English | MEDLINE | ID: mdl-30995674

ABSTRACT

CRISPR-Cas base-editor technology enables targeted nucleotide alterations, and is being increasingly used for research and potential therapeutic applications1,2. The most widely used cytosine base editors (CBEs) induce deamination of DNA cytosines using the rat APOBEC1 enzyme, which is targeted by a linked Cas protein-guide RNA complex3,4. Previous studies of the specificity of CBEs have identified off-target DNA edits in mammalian cells5,6. Here we show that a CBE with rat APOBEC1 can cause extensive transcriptome-wide deamination of RNA cytosines in human cells, inducing tens of thousands of C-to-U edits with frequencies ranging from 0.07% to 100% in 38-58% of expressed genes. CBE-induced RNA edits occur in both protein-coding and non-protein-coding sequences and generate missense, nonsense, splice site, and 5' and 3' untranslated region mutations. We engineered two CBE variants bearing mutations in rat APOBEC1 that substantially decreased the number of RNA edits (by more than 390-fold and more than 3,800-fold) in human cells. These variants also showed more precise on-target DNA editing than the wild-type CBE and, for most guide RNAs tested, no substantial reduction in editing efficiency. Finally, we show that an adenine base editor7 can also induce transcriptome-wide RNA edits. These results have implications for the use of base editors in both research and clinical settings, illustrate the feasibility of engineering improved variants with reduced RNA editing activities, and suggest the need to more fully define and characterize the RNA off-target effects of deaminase enzymes in base editor platforms.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing , RNA Editing , Substrate Specificity/genetics , Transcriptome/genetics , APOBEC-1 Deaminase/chemistry , APOBEC-1 Deaminase/genetics , APOBEC-1 Deaminase/metabolism , Animals , Base Sequence , Cytosine/metabolism , Deamination , HEK293 Cells , Hep G2 Cells , Humans , Mutation , RNA/chemistry , RNA/metabolism , Rats
19.
J Mol Biol ; 431(7): 1506-1517, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30844405

ABSTRACT

RNA editing is an important form of regulating gene expression and activity. APOBEC1 cytosine deaminase was initially characterized as pairing with a cofactor, A1CF, to form an active RNA editing complex that specifically targets APOB RNA in regulating lipid metabolism. Recent studies revealed that APOBEC1 may be involved in editing other potential RNA targets in a tissue-specific manner, and another protein, RBM47, appears to instead be the main cofactor of APOBEC1 for editing APOB RNA. In this report, by expressing APOBEC1 with either A1CF or RBM47 from human or mouse in an HEK293T cell line with no intrinsic APOBEC1/A1CF/RBM47 expression, we have compared direct RNA editing activity on several known cellular target RNAs. By using a sensitive cell-based fluorescence assay that enables comparative quantification of RNA editing through subcellular localization changes of eGFP, the two APOBEC1 cofactors, A1CF and RBM47, showed clear differences for editing activity on APOB and several other tested RNAs, and clear differences were observed when mouse versus human genes were tested. In addition, we have determined the minimal domain requirement of RBM47 needed for activity. These results provide useful functional characterization of RBM47 and direct biochemical evidence for the differential editing selectivity on a number of RNA targets.


Subject(s)
APOBEC-1 Deaminase/metabolism , RNA Editing , RNA-Binding Proteins/metabolism , APOBEC-1 Deaminase/chemistry , APOBEC-1 Deaminase/genetics , Animals , Gene Expression Regulation , HEK293 Cells , Humans , Mice , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics
20.
Biotechnol Bioeng ; 116(6): 1475-1483, 2019 06.
Article in English | MEDLINE | ID: mdl-30739328

ABSTRACT

Clostridium beijerinckii is a potentially important industrial microorganism as it can synthesize valuable chemicals and fuels from various carbon sources. The establishment of convenient to use, effective gene tools with which the organism can be rapidly modified is essential if its full potential is to be realized. Here, we developed a genomic editing tool (pCBEclos) for use in C. beijerinckii based on the fusion of cytidine deaminase (Apobec1), Cas9 D10A nickase and uracil DNA glycosylase inhibitor (UGI). Apobec1 and UGI are guided to the target site where they introduce specific base-pair substitutions through the conversion of C·G to T·A. By appropriate choice of target sequence, these nucleotide changes are capable of creating missense mutation or null mutations in a gene. Through optimization of pCBEclos, the system derived, pCBEclos-opt, has been used to rapidly generate four different mutants in C. beijerinckii, in pyrE, xylR, spo0A, and araR. The efficiency of the system was such that they could sometimes be directly obtained following transformation, otherwise only requiring one single restreaking step. Whilst CRISPR-Cas9 nickase systems, such as pNICKclos2.0, have previously been reported in C. beijerinckii, pCBEclos-opt does not rely on homologous recombination, a process that is intrinsically inefficient in clostridia such as C. beijerinckii. As a consequence, bulky editing templates do not need to be included in the knockout plasmids. This both reduces plasmid size and makes their construction simpler, for example, whereas the assembly of pNICKclos2.0 requires six primers for the assembly of a typical knockout plasmid, pCBEclos-opt requires just two primers. The pCBEclos-opt plasmid established here represents a powerful new tool for genome editing in C. beijerinckii, which should be readily applicable to other clostridial species.


Subject(s)
CRISPR-Associated Protein 9/metabolism , Clostridium beijerinckii/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Deoxyribonuclease I/metabolism , Gene Editing/methods , Recombinant Fusion Proteins/metabolism , APOBEC-1 Deaminase/genetics , APOBEC-1 Deaminase/metabolism , Base Pairing/genetics , CRISPR-Associated Protein 9/genetics , DNA/genetics , DNA/metabolism , Deoxyribonuclease I/genetics , Genetic Vectors , Plasmids , Recombinant Fusion Proteins/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...