Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mol Ther ; 28(7): 1673-1683, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32413280

ABSTRACT

Base editing technology efficiently generates nucleotide conversions without inducing excessive double-strand breaks (DSBs), which makes it a promising approach for genetic disease therapy. In this study, we generated a novel hereditary tyrosinemia type 1 (HT1) mouse model, which contains a start codon mutation in the fumarylacetoacetate hydrolase (Fah) gene by using an adenine base editor (ABE7.10). To investigate the feasibility of base editing for recombinant adeno-associated virus (rAAV)-mediated gene therapy, an intein-split cytosine base editor (BE4max) was developed. BE4max efficiently induced C-to-T conversion and restored the start codon to ameliorate HT1 in mice, but an undesired bystander mutation abolished the effect of on-target editing. To solve this problem, an upstream sequence was targeted to generate a de novo in-frame start codon to initiate the translation of FAH. After treatment, almost all C-to-T conversions created a start codon and restored Fah expression, which efficiently ameliorated the disease without inducing off-target mutations. Our study demonstrated that base editing-mediated creation of de novo functional elements would be an applicable new strategy for genetic disease therapy.


Subject(s)
Codon, Initiator , Gene Editing/methods , Hydrolases/genetics , Tyrosinemias/therapy , Animals , Cytidine/genetics , Dependovirus/genetics , Disease Models, Animal , Feasibility Studies , Genetic Therapy , Genetic Vectors/administration & dosage , HEK293 Cells , Humans , Inteins , Mice , Tyrosinemias/genetics
2.
J Pathol ; 243(2): 165-175, 2017 10.
Article in English | MEDLINE | ID: mdl-28671728

ABSTRACT

Gastro-oesophageal junction (GEJ) carcinoma and distal gastric cancer (GC) have distinct epidemiology and clinical features and their relationship is uncertain. Synchronous multiple gastric cancers located mostly at proximal and distal sites provide rare specimens for investigating the comprehensive genomic relationships among these cancers in the context of identical genetic circumstances. Formalin-fixed, paraffin-embedded (FFPE) samples from 12 patients with synchronous GEJ carcinoma and distal GC were collected in this study. Whole-exome sequencing (WES) was performed using normal tissues as a control. Mutational profiling, clonality analysis, a detailed clinico-pathological review, determination of MSI status, EBER in situ hybridization (ISH), and programmed cell death 1 (PD-1)/programmed death-ligand 1 (PD-L1) immunohistochemical staining were performed. Twenty-three of the 24 samples were microsatellite-stable (MSS). Subclonal analysis revealed that nine pairs of GEJ and distal GC tumours in neoadjuvant chemotherapy naïve patients developed independently from different origins. Two patients who received neoadjuvant chemotherapy shared clonal origins with highly similar somatic alterations. The remaining one patient who shared a rare mutation died within 6.2 months at the N3 stage. However, the enriched pathway identified from the overall mutation spectra in distal GC and GEJ carcinoma showed the close relationship of these cancers. Thus, although these cancers may have similar characteristics, histopathological and genetic profiling from single tumour specimens may still underestimate the mutational burden and somatic heterogeneity of multiple GCs. In addition, this series of cases also showed a PD-L1 expression rate of 58.3% and 66.7% in distal GC and GEJ carcinoma, respectively, with all the cases expressing PD-1. This result suggests the potential benefit of immunotherapeutic treatments. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Esophageal Neoplasms/genetics , Esophagogastric Junction , Stomach Neoplasms/genetics , Aged , Clone Cells/physiology , DNA Mutational Analysis/methods , Esophageal Neoplasms/pathology , Exome/genetics , Female , Humans , Male , Microsatellite Repeats/genetics , Middle Aged , Mutation/genetics , Neoplasm Proteins/genetics , Stomach Neoplasms/pathology , Tumor Burden
3.
Genome Res ; 24(10): 1613-23, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25170028

ABSTRACT

Cytosine methylation is critical in mammalian development and plays a role in diverse biologic processes such as genomic imprinting, X chromosome inactivation, and silencing of repeat elements. Several factors regulate DNA methylation in early embryogenesis, but their precise role in the establishment of DNA methylation at a given site remains unclear. We have generated a comprehensive methylation map in fibroblasts derived from the murine DNA methylation mutant Hells(-/-) (helicase, lymphoid specific, also known as LSH). It has been previously shown that HELLS can influence de novo methylation of retroviral sequences and endogenous genes. Here, we describe that HELLS controls cytosine methylation in a nuclear compartment that is in part defined by lamin B1 attachment regions. Despite widespread loss of cytosine methylation at regulatory sequences, including promoter regions of protein-coding genes and noncoding RNA genes, overall relative transcript abundance levels in the absence of HELLS are similar to those in wild-type cells. A subset of promoter regions shows increases of the histone modification H3K27me3, suggesting redundancy of epigenetic silencing mechanisms. Furthermore, HELLS modulates CG methylation at all classes of repeat elements and is critical for repression of a subset of repeat elements. Overall, we provide a detailed analysis of gene expression changes in relation to DNA methylation alterations, which contributes to our understanding of the biological role of cytosine methylation.


Subject(s)
Cytosine/metabolism , DNA Helicases/genetics , DNA Methylation , DNA/metabolism , Gene Expression Regulation, Developmental , Animals , Cell Nucleus/genetics , Embryonic Stem Cells , Epigenesis, Genetic , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Knockout Techniques , Histones/metabolism , Lamin Type B/metabolism , Mice , Molecular Sequence Data , Mutation , Regulatory Sequences, Nucleic Acid , Repetitive Sequences, Nucleic Acid
4.
Nucleic Acids Res ; 43(3): 1444-55, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25578963

ABSTRACT

Lsh, a chromatin remodeling protein of the SNF2 family, is critical for normal heterochromatin structure. In particular, DNA methylation at repeat elements, a hallmark of heterochromatin, is greatly reduced in Lsh(-/-) (KO) cells. Here, we examined the presumed nucleosome remodeling activity of Lsh on chromatin in the context of DNA methylation. We found that dynamic CG methylation was dependent on Lsh in embryonic stem cells. Moreover, we demonstrate that ATP function is critical for de novo methylation at repeat sequences. The ATP binding site of Lsh is in part required to promote stable association of the DNA methyltransferase 3b with the repeat locus. By performing nucleosome occupancy assays, we found distinct nucleosome occupancy in KO ES cells compared to WT ES cells after differentiation. Nucleosome density was restored to wild-type level by re-expressing wild-type Lsh but not the ATP mutant in KO ES cells. Our results suggest that ATP-dependent nucleosome remodeling is the primary molecular function of Lsh, which may promote de novo methylation in differentiating ES cells.


Subject(s)
Adenosine Triphosphate/metabolism , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , DNA Methylation , Nucleosomes/metabolism , Repetitive Sequences, Nucleic Acid , Binding Sites , Cells, Cultured , Humans
5.
Proc Natl Acad Sci U S A ; 111(16): 5890-5, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24711395

ABSTRACT

DNA methylation patterns are established in early embryogenesis and are critical for cellular differentiation. To investigate the role of CG methylation in potential enhancer formation, we assessed H3K4me1 modification in murine embryonic fibroblasts (MEFs) derived from the DNA methylation mutant Lsh(-/-) mice. We report here de novo formation of putative enhancer elements at CG hypomethylated sites that can be dynamically altered. We found a subset of differentially enriched H3K4me1 regions clustered at neuronal lineage genes and overlapping with known cis-regulatory elements present in brain tissue. Reprogramming of Lsh(-/-) MEFs into induced pluripotent stem (iPS) cells leads to increased neuronal lineage gene expression of premarked genes and enhanced differentiation potential of Lsh(-/-) iPS cells toward the neuronal lineage pathway compared with WT iPS cells in vitro and in vivo. The state of CG hypomethylation and H3K4me1 enrichment is partially maintained in Lsh(-/-) iPS cells. The acquisition of H3K27ac and activity of subcloned fragments in an enhancer reporter assay indicate functional activity of several of de novo H3K4me1-marked sequences. Our results suggest a functional link of H3K4me1 enrichment at CG hypomethylated sites, enhancer formation, and cellular plasticity.


Subject(s)
CpG Islands/genetics , DNA Helicases/deficiency , DNA Methylation/genetics , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Histones/metabolism , Lysine/metabolism , Animals , Biomarkers/metabolism , Cell Lineage , DNA Helicases/metabolism , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Fibroblasts/cytology , HeLa Cells , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Knockout , Neurons/cytology , Protein Binding , Signal Transduction , Transcription Factors/metabolism
6.
Proc Natl Acad Sci U S A ; 111(4): E511-20, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24474800

ABSTRACT

Malaria infection triggers vigorous host immune responses; however, the parasite ligands, host receptors, and the signaling pathways responsible for these reactions remain unknown or controversial. Malaria parasites primarily reside within RBCs, thereby hiding themselves from direct contact and recognition by host immune cells. Host responses to malaria infection are very different from those elicited by bacterial and viral infections and the host receptors recognizing parasite ligands have been elusive. Here we investigated mouse genome-wide transcriptional responses to infections with two strains of Plasmodium yoelii (N67 and N67C) and discovered differences in innate response pathways corresponding to strain-specific disease phenotypes. Using in vitro RNAi-based gene knockdown and KO mice, we demonstrated that a strong type I IFN (IFN-I) response triggered by RNA polymerase III and melanoma differentiation-associated protein 5, not Toll-like receptors (TLRs), binding of parasite DNA/RNA contributed to a decline of parasitemia in N67-infected mice. We showed that conventional dendritic cells were the major sources of early IFN-I, and that surface expression of phosphatidylserine on infected RBCs might promote their phagocytic uptake, leading to the release of parasite ligands and the IFN-I response in N67 infection. In contrast, an elevated inflammatory response mediated by CD14/TLR and p38 signaling played a role in disease severity and early host death in N67C-infected mice. In addition to identifying cytosolic DNA/RNA sensors and signaling pathways previously unrecognized in malaria infection, our study demonstrates the importance of parasite genetic backgrounds in malaria pathology and provides important information for studying human malaria pathogenesis.


Subject(s)
Host-Parasite Interactions , Immunity, Innate , Malaria/immunology , Parasitemia/immunology , Plasmodium yoelii/physiology , Signal Transduction , Aged , Animals , Humans , Inflammation/immunology , Inflammation/metabolism , Interferon Type I/metabolism , Malaria/mortality , Malaria/parasitology , Mice , Mice, Knockout , Parasitemia/parasitology , Phagocytosis , Plasmodium yoelii/immunology
7.
Nucleic Acids Res ; 39(6): 2234-48, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21097889

ABSTRACT

Cell growth and proliferation are tightly controlled via the regulation of the p53-MDM2 feedback loop in response to various cellular stresses. In this study, we identified a nucleolar protein called PAK1IP1 as another regulator of this loop. PAK1IP1 was induced when cells were treated with chemicals that disturb ribosome biogenesis. Overexpression of PAK1IP1 inhibited cell proliferation by inducing p53-dependent G1 cell-cycle arrest. PAK1IP1 bound to MDM2 and inhibited its ability to ubiquitinate and to degrade p53, consequently leading to the accumulation of p53 levels. Interestingly, knockdown of PAK1IP1 in cells also inhibited cell proliferation and induced p53-dependent G1 arrest. Deficiency of PAK1IP1 increased free ribosomal protein L5 and L11 which were required for PAK1IP1 depletion-induced p53 activation. Taken together, our results reveal that PAK1IP1 is a new nucleolar protein that is crucial for rRNA processing and plays a regulatory role in cell proliferation via the p53-MDM2 loop.


Subject(s)
Cell Proliferation , Intracellular Signaling Peptides and Proteins/physiology , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Cell Cycle , Cell Line , Cell Nucleolus/chemistry , DNA/biosynthesis , G1 Phase , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Sequence Data , Nuclear Localization Signals , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/metabolism , Ubiquitination
8.
Biochem J ; 440(2): 273-82, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21831044

ABSTRACT

Heparanase is involved in the cleavage of the HS (heparan sulfate) chain of HSPGs (HS proteoglycans) and hence participates in remodelling of the ECM (extracellular matrix) and BM (basement membrane). In the present study we have shown that NGF (nerve growth factor) promoted nuclear enrichment of EGR1 (early growth response 1), a transcription factor for heparanase, and markedly induced heparanase expression in rat adrenal pheochromocytoma (PC12) cells. K252a, an antagonist of the NGF receptor TrkA (tyrosine kinase receptor A), decreased heparanase protein expression induced by NGF in PC12 cells. Suramin, a heparanase inhibitor, decreased heparanase in PC12 cells and blocked NGF-induced PC12 neuritogenesis. Stable overexpression of heparanase activated p38 MAPK (mitogen-activated protein kinase) by phosphorylation and enhanced the neurite outgrowth induced by NGF, whereas knock down of heparanase impaired this process. However, overexpression of latent pro-heparanase with a Y156A mutation still led to enhanced NGF-induced neurite outgrowth and increased p38 MAPK phosphorylation. Inhibition of p38 MAPK by SB203580 suppressed the promotion of NGF-induced neuritogenesis by the wild-type and mutant heparanase. The impaired differentiation by knock down of heparanase could be restored by transfection of wild-type or mutant heparanase in PC12 cells. The results of the present study suggest that heparanase, at least in the non-enzymatic form, may promote NGF-induced neuritogenesis via the p38 MAPK pathway.


Subject(s)
Nerve Growth Factor/pharmacology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Carbazoles/pharmacology , Gene Knockdown Techniques , Glucuronidase/antagonists & inhibitors , Indole Alkaloids/pharmacology , Neurites/physiology , PC12 Cells , Rats , Receptor, trkA/antagonists & inhibitors , Suramin/pharmacology
9.
Sci China Life Sci ; 65(4): 718-730, 2022 04.
Article in English | MEDLINE | ID: mdl-32815069

ABSTRACT

Genome editing through adeno-associated viral (AAV) vectors is a promising gene therapy strategy for various diseases, especially genetic disorders. However, homologous recombination (HR) efficiency is extremely low in adult animal models. We assumed that increasing AAV transduction efficiency could increase genome editing activity, especially HR efficiency, for in vivo gene therapy. Firstly, a mouse phenylketonuria (PKU) model carrying a pathogenic R408W mutation in phenylalanine hydroxylase (Pah) was generated. Through co-delivery of the general AAV receptor (AAVR), we found that AAVR could dramatically increase AAV transduction efficiency in vitro and in vivo. Furthermore, co-delivery of SaCas9/sgRNA/donor templates with AAVR via AAV8 vectors increased indel rate over 2-fold and HR rate over 15-fold for the correction of the single mutation in PahR408W mice. Moreover, AAVR co-injection successfully increased the site-specific insertion rate of a 1.4 kb Pah cDNA by 11-fold, bringing the HR rate up to 7.3% without detectable global off-target effects. Insertion of Pah cDNA significantly decreased the Phe level and ameliorated PKU symptoms. This study demonstrates a novel strategy to dramatically increase AAV transduction which substantially enhanced in vivo genome editing efficiency in adult animal models, showing clinical potential for both conventional and genome editing-based gene therapy.


Subject(s)
Liver Diseases , Phenylalanine Hydroxylase , Phenylketonurias , Animals , DNA, Complementary , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Gene Editing , Genetic Vectors/genetics , Mice , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Phenylketonurias/genetics , Phenylketonurias/therapy
10.
Nucleic Acids Res ; 37(21): 7059-71, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19783823

ABSTRACT

SMYD1 is a heart and muscle specific SET-MYND domain containing protein, which functions as a histone methyltransferase and regulates downstream gene transcription. We demonstrated that the expression of SMYD1 is restricted in the heart and skeletal muscle tissues in human. To reveal the regulatory mechanisms of SMYD1 expression during myogenesis and cardiogenesis, we cloned and characterized the human SMYD1 promoter, which contains highly conserved serum response factor (SRF) and myogenin binding sites. Overexpression of SRF and myogenin significantly increased the endogenous expression level of Smyd1 in C2C12 cells, respectively. Deletion of Srf in the heart of mouse embryos dramatically decreased the expression level of Smyd1 mRNA and the expression of Smyd1 can be rescued by exogenous SRF introduction in SRF null ES cells during differentiation. Furthermore, we demonstrated that SRF binds to the CArG site and myogenin binds to the E-box element on Smyd1 promoter region using EMSA and ChIP assays. Moreover, forced expression of SMYD1 accelerates myoblast differentiation and myotube formation in C2C12 cells. Taken together, these studies demonstrated that SMYD1 is a key regulator of myogenic differentiation and acts as a downstream target of muscle regulatory factors, SRF and myogenin.


Subject(s)
DNA-Binding Proteins/genetics , Muscle Proteins/genetics , Myogenin/metabolism , Serum Response Factor/metabolism , Transcription Factors/genetics , Animals , Binding Sites , Cell Differentiation , Cell Line , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , E-Box Elements , Embryo, Mammalian/metabolism , Humans , Mice , Mice, Knockout , Muscle Development , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Myocardium/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , Serum Response Factor/genetics , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcriptional Activation
11.
Article in English | MEDLINE | ID: mdl-33568421

ABSTRACT

Infantile spasms are a potentially catastrophic form of epilepsy syndrome that are usually associated with substantial developmental delay and commonly occur in children younger than 1 yr. Recent reports on four cases revealed that variants harbored in a novel gene CDK19 were causative for the syndrome. We report a fifth affected individual, a 10-mo-old male patient who presented with a neurodevelopmental syndrome characterized by infantile spasms. We identified a novel de novo missense variant c.92C > A (p.Thr31Asn) in CDK19 that was classified as a likely pathogenic disease-causing variant. The characterized clinical phenotypes of the proband were similar to the previously reported four patients, but he had few variable features including earlier seizure onset age and earlier occurring developmental abnormality. Protein structure modeling analysis revealed that CDK19 variants may disable its kinase activity, which would further impede the transcriptional regulation, thus leading to detrimental pathologies. Our report expanded CDK19 genotype spectrum and further demonstrated that a CDK19 missense variant was causative of neurodevelopmental disorder clinically marked by infantile spasms.


Subject(s)
Cyclin-Dependent Kinases/genetics , Genetic Variation , Neurodevelopmental Disorders/genetics , Spasms, Infantile/genetics , Cyclin-Dependent Kinases/chemistry , Epileptic Syndromes , Humans , Infant , Male , Mutation, Missense , Neurodevelopmental Disorders/diagnostic imaging , Neurodevelopmental Disorders/pathology , Pedigree , Phenotype , Seizures/genetics , Exome Sequencing
12.
BMC Med Genomics ; 14(1): 152, 2021 06 09.
Article in English | MEDLINE | ID: mdl-34107977

ABSTRACT

BACKGROUND: Variants identified through parent-child trio-WES yield up to 28-55% positive diagnostic rate across a variety of Mendelian disorders, there remain numerous patients who do not receive a genetic diagnosis. Studies showed that some aberrant splicing variants, which are either not readily detectable by WES or could be miss-interpreted by regular detecting pipelines, are highly relevant to human diseases. METHODS: We retrospectively investigated the negative molecular diagnostics through trio-WES for 15 genetically undiagnosed patients whose clinical manifestations were highly suspected to be genetic disorders with well-established genotype-phenotype relationships. We scrutinized the synonymous variants from WES data and Sanger sequenced the suspected intronic region for deep intronic variants. The functional consequences of variants were analyzed by in vitro minigene experiments. RESULTS: Here, we report two abnormal splicing events, one of which caused exon truncating due to the activation of cryptic splicing site by a synonymous variant; the other caused partial intron retention due to the generation of splicing sites by a deep intronic variant. CONCLUSIONS: We suggest that, despite initial negative genetic test results in clinically highly suspected genetic diseases, the combination of predictive bioinformatics and functional analysis should be considered to unveil the genetic etiology of undiagnosed rare diseases.


Subject(s)
RNA Splicing
13.
Nat Metab ; 3(1): 90-106, 2021 01.
Article in English | MEDLINE | ID: mdl-33398195

ABSTRACT

Organelles use specialized molecules to regulate their essential cellular processes. However, systematically elucidating the subcellular distribution and function of molecules such as long non-coding RNAs (lncRNAs) in cellular homeostasis and diseases has not been fully achieved. Here, we reveal the diverse and abundant subcellular distribution of organelle-associated lncRNAs from mitochondria, lysosomes and endoplasmic reticulum. Among them, we identify the mitochondrially localized lncRNA growth-arrest-specific 5 (GAS5) as a tumour suppressor in maintaining cellular energy homeostasis. Mechanistically, energy-stress-induced GAS5 modulates mitochondrial tricarboxylic acid flux by disrupting metabolic enzyme tandem association of fumarate hydratase, malate dehydrogenase and citrate synthase, the canonical members of the tricarboxylic acid cycle. GAS5 negatively correlates with levels of its associated mitochondrial metabolic enzymes in tumours and benefits overall survival in individuals with breast cancer. Together, our detailed annotation of subcellular lncRNA distribution identifies a functional role for lncRNAs in regulating cellular metabolic homeostasis, highlighting organelle-associated lncRNAs as potential clinical targets to manipulate cellular metabolism and diseases.


Subject(s)
Citric Acid Cycle/physiology , Mitochondria/metabolism , RNA, Long Noncoding/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Female , Homeostasis , Humans , Lysosomes/metabolism , Mice , Mice, Inbred BALB C , Nutrients , Organelles/metabolism , RNA, Neoplasm/genetics
14.
Nat Cell Biol ; 22(6): 740-750, 2020 06.
Article in English | MEDLINE | ID: mdl-32393889

ABSTRACT

Cytidine base editors are powerful genetic tools that catalyse cytidine to thymidine conversion at specific genomic loci, and further improvement of the editing range and efficiency is critical for their broader applications. Through insertion of a non-sequence-specific single-stranded DNA-binding domain from Rad51 protein between Cas9 nickase and the deaminases, serial hyper cytidine base editors were generated with substantially increased activity and an expanded editing window towards the protospacer adjacent motif in both cell lines and mouse embryos. Additionally, hyeA3A-BE4max selectively catalysed cytidine conversion in TC motifs with a broader editing range and much higher activity (up to 257-fold) compared with eA3A-BE4max. Moreover, hyeA3A-BE4max specifically generated a C-to-T conversion without inducing bystander mutations in the haemoglobin gamma gene promoter to mimic a naturally occurring genetic variant for amelioration of ß-haemoglobinopathy, suggesting the therapeutic potential of the improved base editors.


Subject(s)
CRISPR-Cas Systems , Cytidine/genetics , DNA-Binding Proteins/metabolism , Gene Editing , Mutation , Rad51 Recombinase/metabolism , Animals , Cell Differentiation , Cytidine/chemistry , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Protein Domains , Rad51 Recombinase/genetics
15.
Nat Biotechnol ; 38(7): 856-860, 2020 07.
Article in English | MEDLINE | ID: mdl-32483363

ABSTRACT

Although base editors are useful tools for precise genome editing, current base editors can only convert either adenines or cytosines. We developed a dual adenine and cytosine base editor (A&C-BEmax) by fusing both deaminases with a Cas9 nickase to achieve C-to-T and A-to-G conversions at the same target site. Compared to single base editors, A&C-BEmax's activity on adenines is slightly reduced, whereas activity on cytosines is higher and RNA off-target activity is substantially decreased.


Subject(s)
Adenine , CRISPR-Cas Systems/genetics , Cytosine , Gene Editing/methods , CRISPR-Associated Protein 9/genetics , Deoxyribonuclease I/genetics , Humans , RNA/genetics
16.
Peptides ; 30(1): 130-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18996159

ABSTRACT

Kisspeptins are the protein products encoded by KiSS1 gene, an important tumor metastatic suppressor and pivotal master hormone of puberty. Although KiSS1 gene is expressed in both central and peripheral tissues, the molecular mechanisms that determine the temporal and spatial expression of KiSS1 gene are not well understood. This review provides an update on the latest studies and ideas about the expression of KiSS1 gene as a puberty gatekeeper and a metastasis suppressor, with special emphasis on the molecular mechanisms for the transcriptional regulation of KiSS1 gene expression.


Subject(s)
Gene Expression Regulation , Tumor Suppressor Proteins/genetics , Cell Line, Tumor , Gonads/physiology , Humans , Hypothalamo-Hypophyseal System/physiology , Kisspeptins , Neoplasms/genetics , Neoplasms/metabolism , Tissue Distribution , Transcription, Genetic , Tumor Suppressor Proteins/metabolism
17.
Mol Cells ; 26(5): 443-53, 2008 Nov 30.
Article in English | MEDLINE | ID: mdl-18719355

ABSTRACT

The Bric-a-brac, Tramtrack, Broad-complex (BTB) domain is a protein-protein interaction domain that is found in many zinc finger transcription factors. BTB containing proteins play important roles in a variety of cellular functions including regulation of transcription, regulation of the cytoskeleton, protein ubiquitination, angiogenesis, and apoptosis. Here, we report the cloning and characterization of a novel human gene, KLHL31, from a human embryonic heart cDNA library. The cDNA of KLHL31 is 5743 bp long, encoding a protein product of 634 amino acids containing a BTB domain. The protein is highly conserved across different species. Western blot analysis indicates that the KLHL31 protein is abundantly expressed in both embryonic skeletal and heart tissue. In COS-7 cells, KLHL31 proteins are localized to both the nucleus and the cytoplasm. In primary cultures of nascent mouse cardiomyocytes, the majority of endogenous KLHL31 proteins are localized to the cytoplasm. KLHL31 acts as a transcription repressor when fused to GAL4 DNA-binding domain and deletion analysis indicates that the BTB domain is the main region responsible for this repression. Overexpression of KLHL31 in COS-7 cells inhibits the transcriptional activities of both the TPA-response element (TRE) and serum response element (SRE). KLHL31 also significantly reduces JNK activation leading to decreased phosphorylation and protein levels of the JNK target c-Jun in both COS-7 and Hela cells. These results suggest that KLHL31 protein may act as a new transcriptional repressor in MAPK/JNK signaling pathway to regulate cellular functions.


Subject(s)
Muscles/metabolism , Myocardium/metabolism , Proteins/metabolism , Serum Response Element/genetics , Transcription, Genetic , Amino Acid Sequence , Animals , Antibodies/metabolism , Cell Nucleus/metabolism , Conserved Sequence , Embryo, Mammalian/metabolism , Enzyme Activation , Evolution, Molecular , Gene Expression Profiling , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Organ Specificity , Phosphorylation , Proteins/chemistry , Proteins/genetics , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Transcriptional Activation
18.
Mutat Res ; 637(1-2): 182-9, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17915261

ABSTRACT

p63 is a transcription factor homologous to p53 and p73; mutations in this gene have been identified in individuals with several types of developmental abnormalities, including EEC (ectrodactyly, ectodermal dysplasia, facial clefts) syndrome and split-hand/split-foot malformation (SHFM). Several mutations in the p63 gene have previously been shown to be related to SHFM. In this study, we report on a Chinese family with intrafamilial clinical variability of SHFM that have a novel heterozygous mutation in all four affected individuals. The mutation is in exon 8 of p63, 1046G --> A, which predicts an amino acid substitution G310E. SSCP analysis of the segregation pattern of the mutation strongly suggests a causal relationship to the SHFM phenotype in p63. This mutation has not been observed in other countries in the world.


Subject(s)
Foot Deformities, Congenital/genetics , Genes, p53 , Hand Deformities, Congenital/genetics , Mutation , Asian People , Base Sequence , Humans , Male , Molecular Sequence Data , Pedigree , Polymorphism, Single-Stranded Conformational , Syndactyly/genetics
19.
Cancer Res ; 78(13): 3484-3496, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29588351

ABSTRACT

Long noncoding RNAs (lncRNA) have been associated with various types of cancer; however, the precise role of many lncRNAs in tumorigenesis remains elusive. Here we demonstrate that the cytosolic lncRNA P53RRA is downregulated in cancers and functions as a tumor suppressor by inhibiting cancer progression. Chromatin remodeling proteins LSH and Cfp1 silenced or increased P53RRA expression, respectively. P53RRA bound Ras GTPase-activating protein-binding protein 1 (G3BP1) using nucleotides 1 and 871 of P53RRA and the RRM interaction domain of G3BP1 (aa 177-466). The cytosolic P53RRA-G3BP1 interaction displaced p53 from a G3BP1 complex, resulting in greater p53 retention in the nucleus, which led to cell-cycle arrest, apoptosis, and ferroptosis. P53RRA promoted ferroptosis and apoptosis by affecting transcription of several metabolic genes. Low P53RRA expression significantly correlated with poor survival in patients with breast and lung cancers harboring wild-type p53. These data show that lncRNAs can directly interact with the functional domain of signaling proteins in the cytoplasm, thus regulating p53 modulators to suppress cancer progression.Significance: A cytosolic lncRNA functions as a tumor suppressor by activating the p53 pathway. Cancer Res; 78(13); 3484-96. ©2018 AACR.


Subject(s)
Breast Neoplasms/pathology , Cell Nucleus/metabolism , DNA Helicases/metabolism , Lung Neoplasms/pathology , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , RNA, Long Noncoding/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Cycle Checkpoints/genetics , Cytoplasm/pathology , DNA Helicases/genetics , Datasets as Topic , Disease Progression , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Male , Mice , Mice, Nude , Mice, SCID , Poly-ADP-Ribose Binding Proteins/genetics , Protein Binding , RNA Helicases/genetics , RNA Recognition Motif/genetics , RNA Recognition Motif Proteins/genetics , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
20.
Cell Death Dis ; 9(5): 490, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29706625

ABSTRACT

Most cancer patients receive radiotherapy in the course of their disease and the occurrence of radioresistance is associated with poor prognosis. The molecular pathways that drive enhanced tumorigenic potential during the development of radioresistance are poorly understood. Here, we demonstrate that aryl hydrocarbon receptor (AhR) plays a vital role in the maintenance of cancer stem-like properties. AhR promotes the cancer stem-like phenotype and drives metastasis by directly targeting the promoters of 'stemness' genes, such as the ATP-binding cassette sub-family G member 2 (ABCG2) gene. Moreover, the radioresistant sublines display high levels of oncometabolites including α-ketoglutarate, and treatment of cancer cells with α-ketoglutarate enhances their stem-like properties in an AhR activation-dependent manner. IKKα directly activates stemness-related genes through an interaction with AhR as a bone fide chromatin modifier. Thus, AhR is functionally linked with cancer stem-like properties, and it drives tumorigenesis in the occurrence of radioresistance.


Subject(s)
Adenocarcinoma of Lung/radiotherapy , Basic Helix-Loop-Helix Transcription Factors/metabolism , I-kappa B Kinase/metabolism , Lung Neoplasms/radiotherapy , Neoplastic Stem Cells/radiation effects , Radiation Tolerance , Receptors, Aryl Hydrocarbon/metabolism , A549 Cells , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Adenocarcinoma of Lung/enzymology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Animals , Binding Sites , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Nude , Mice, SCID , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Phenotype , Promoter Regions, Genetic , Signal Transduction , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL