Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 19.340
Filter
1.
Nat Methods ; 21(8): 1411, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39122957

Subject(s)
RNA Splicing , Humans
2.
Nat Commun ; 15(1): 6964, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138195

ABSTRACT

Though RNAi and RNA-splicing machineries are involved in regulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication, their precise roles in coronavirus disease 2019 (COVID-19) pathogenesis remain unclear. Herein, we show that decreased RNAi component (Dicer and XPO5) and splicing factor (SRSF3 and hnRNPA3) expression correlate with increased COVID-19 severity. SARS-CoV-2 N protein induces the autophagic degradation of Dicer, XPO5, SRSF3, and hnRNPA3, inhibiting miRNA biogenesis and RNA splicing and triggering DNA damage, proteotoxic stress, and pneumonia. Dicer, XPO5, SRSF3, and hnRNPA3 knockdown increases, while their overexpression decreases, N protein-induced pneumonia's severity. Older mice show lower expression of Dicer, XPO5, SRSF3, and hnRNPA3 in their lung tissues and exhibit more severe N protein-induced pneumonia than younger mice. PJ34, a poly(ADP-ribose) polymerase inhibitor, or anastrozole, an aromatase inhibitor, ameliorates N protein- or SARS-CoV-2-induced pneumonia by restoring Dicer, XPO5, SRSF3, and hnRNPA3 expression. These findings will aid in developing improved treatments for SARS-CoV-2-associated pneumonia.


Subject(s)
COVID-19 , Karyopherins , Ribonuclease III , SARS-CoV-2 , Serine-Arginine Splicing Factors , Animals , Serine-Arginine Splicing Factors/metabolism , Serine-Arginine Splicing Factors/genetics , Humans , Ribonuclease III/metabolism , Ribonuclease III/genetics , SARS-CoV-2/genetics , COVID-19/metabolism , COVID-19/virology , COVID-19/genetics , Mice , Karyopherins/metabolism , Karyopherins/genetics , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Down-Regulation , Lung/metabolism , Lung/pathology , Lung/virology , Male , Female , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Splicing , Autophagy/genetics , DNA Damage , Heterogeneous-Nuclear Ribonucleoprotein Group A-B
3.
Sci Rep ; 14(1): 18982, 2024 08 16.
Article in English | MEDLINE | ID: mdl-39152135

ABSTRACT

Circular RNAs (circRNAs) as biomarkers for cancer detection have been extensively explored, however, the biogenesis mechanism is still elusive. In contrast to linear splicing (LS) involved in linear transcript formation, the so-called back splicing (BS) process has been proposed to explain circRNA formation. To investigate the potential mechanism of BS via the machine learning approach, we curated a high-quality BS and LS exon pairs dataset with evidence-based stringent filtering. Two convolutional neural networks (CNN) base models with different structures for processing splicing junction sequences including motif extraction were created and compared after extensive hyperparameter tuning. In contrast to the previous study, we are able to identify motifs corresponding to well-established BS-associated genes such as MBNL1, QKI, and ESPR2. Importantly, despite prevalent high false positive rates in existing circRNA detection pipelines and databases, our base models demonstrated a notable high specificity (greater than 90%). To further improve the model performance, a novo fast numerical method was proposed and implemented to calculate the reverse complementary matches (RCMs) crossing two flanking regions and within each flanking region of exon pairs. Our CircCNNs framework that incorporated RCM information into the optimal base models further reduced the false positive rates leading to 88% prediction accuracy.


Subject(s)
Exons , Neural Networks, Computer , RNA, Circular , RNA, Circular/genetics , Humans , Exons/genetics , RNA Splicing , Computational Biology/methods , Machine Learning
4.
Virol J ; 21(1): 175, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107824

ABSTRACT

BACKGROUND: Hemorrhagic enteritis, caused by Turkey Hemorrhagic Enteritis Virus (THEV), is a disease affecting turkey poults characterized by immunosuppression and bloody diarrhea. An avirulent THEV strain that retains the immunosuppressive ability is used as a live vaccine. Characterizing the splice map of THEV is an essential step that would allow studies of individual genes mediating its immunosuppressive functions. We used RNA sequencing to characterize the splice map of THEV for the first time, providing key insights into the THEV gene expression and mRNA structures. METHODS: After infecting a turkey B-cell line with the vaccine strain, samples in triplicates were collected at 4-, 12-, 24-, and 72-hours post-infection. Total RNA was extracted, and poly-A-tailed mRNA sequenced. Reads were mapped to the THEV genome after trimming and transcripts assembled with StringTie. We performed PCR of THEV cDNA, cloned the PCR products, and used Sanger sequencing to validate all identified splice junctions. RESULTS: Researchers previously annotated the THEV genome as encoding 23 open reading frames (ORFs). We identified 29 spliced transcripts from our RNA sequencing data, all containing novel exons although some exons matched some previously annotated ORFs. The three annotated splice junctions were also corroborated by our data. During validation we identified five additional unique transcripts, a subset of which were further validated by 3' rapid amplification of cDNA ends (3' RACE). Thus, we report that the genome of THEV contains 34 transcripts with the coding capacity for all annotated ORFs. However, we found six of the previously annotated ORFs to be truncated ORFs on the basis of the identification of an in-frame upstream start codon or the detection of additional coding exons. We also identified three of the annotated ORFs with longer or shorter isoforms, and seven novel unannotated ORFs that could potentially be translated; although it is beyond the scope of this manuscript to investigate whether they are translated. CONCLUSIONS: Similar to human adenoviruses, all THEV transcripts are spliced and organized into five transcription units under the control of their cognate promoters. The genes are expressed under temporal regulation and THEV also produces multiple distinctly spliced transcripts that code for the same protein. Studies of the newly identified potential proteins should be urgently performed as these proteins may have roles in THEV-induced immunosuppression. Also, knowing the splicing of THEV genes should be invaluable to future research focusing on studying THEV genes, as this will allow accurate cloning of the mRNAs.


Subject(s)
Open Reading Frames , Turkeys , Animals , Turkeys/virology , Coronavirus, Turkey/genetics , RNA, Messenger/genetics , RNA Splicing , Genome, Viral , Cell Line , RNA, Viral/genetics , Poultry Diseases/virology , Sequence Analysis, RNA
5.
Funct Integr Genomics ; 24(4): 135, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39117866

ABSTRACT

Gene co-expression networks may encode hitherto inadequately recognized vulnerabilities for adult gliomas. By identifying evolutionally conserved gene co-expression modules around EGFR (EM) or PDGFRA (PM), we recently proposed an EM/PM classification scheme, which assigns IDH-wildtype glioblastomas (GBM) into the EM subtype committed in neural stem cell compartment, IDH-mutant astrocytomas and oligodendrogliomas into the PM subtype committed in early oligodendrocyte lineage. Here, we report the identification of EM/PM subtype-specific gene co-expression networks and the characterization of hub gene polypyrimidine tract-binding protein 1 (PTBP1) as a genomic alteration-independent vulnerability in IDH-wildtype GBM. Supervised by the EM/PM classification scheme, we applied weighted gene co-expression network analysis to identify subtype-specific global gene co-expression modules. These gene co-expression modules were characterized for their clinical relevance, cellular origin and conserved expression pattern during brain development. Using lentiviral vector-mediated constitutive or inducible knockdown, we characterized the effects of PTBP1 on the survival of IDH-wildtype GBM cells, which was complemented with the analysis of PTBP1-depedent splicing pattern and overexpression of splicing target neuron-specific CDC42 (CDC42-N) isoform.  Transcriptomes of adult gliomas can be robustly assigned into 4 large gene co-expression modules that are prognostically relevant and are derived from either malignant cells of the EM/PM subtypes or tumor microenvironment. The EM subtype is associated with a malignant cell-intrinsic gene module involved in pre-mRNA splicing, DNA replication and damage response, and chromosome segregation, and a microenvironment-derived gene module predominantly involved in extracellular matrix organization and infiltrating immune cells. The PM subtype is associated with two malignant cell-intrinsic gene modules predominantly involved in transcriptional regulation and mRNA translation, respectively. Expression levels of these gene modules are independent prognostic factors and malignant cell-intrinsic gene modules are conserved during brain development. Focusing on the EM subtype, we identified PTBP1 as the most significant hub for the malignant cell-intrinsic gene module. PTBP1 is not altered in most glioma genomes. PTBP1 represses the conserved splicing of CDC42-N. PTBP1 knockdown or CDC42-N overexpression disrupts actin cytoskeleton dynamics, causing accumulation of reactive oxygen species and cell apoptosis. PTBP1-mediated repression of CDC42-N splicing represents a potential genomic alteration-independent, developmentally conserved vulnerability in IDH-wildtype GBM.


Subject(s)
Glioblastoma , Heterogeneous-Nuclear Ribonucleoproteins , Polypyrimidine Tract-Binding Protein , cdc42 GTP-Binding Protein , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Humans , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , Cell Line, Tumor , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Gene Regulatory Networks , Gene Expression Regulation, Neoplastic , RNA Splicing , Neurons/metabolism , Neurons/pathology
6.
Sci Rep ; 14(1): 18278, 2024 08 07.
Article in English | MEDLINE | ID: mdl-39107424

ABSTRACT

To decipher the molecular bases governing seed germination, this study presents the pivotal role of the cap-binding complex (CBC), comprising CBP20 and CBP80, in modulating the inhibitory effects of abscisic acid (ABA) in barley. Using both single and double barley mutants in genes encoding the CBC, we revealed that the double mutant hvcbp20.ab/hvcbp80.b displays ABA insensitivity, in stark contrast to the hypersensitivity observed in single mutants during germination. Our comprehensive transcriptome and metabolome analysis not only identified significant alterations in gene expression and splicing patterns but also underscored the regulatory nexus among CBC, ABA, and brassinosteroid (BR) signaling pathways.


Subject(s)
Abscisic Acid , Gene Expression Regulation, Plant , Germination , Hordeum , Plant Proteins , Hordeum/genetics , Hordeum/metabolism , Hordeum/growth & development , Germination/genetics , Abscisic Acid/metabolism , Abscisic Acid/pharmacology , Plant Proteins/genetics , Plant Proteins/metabolism , RNA Splicing , Mutation , Signal Transduction , Transcriptome , Gene Expression Profiling , RNA Cap-Binding Proteins/metabolism , RNA Cap-Binding Proteins/genetics , Seeds/growth & development , Seeds/genetics , Seeds/metabolism
7.
Nat Cell Biol ; 26(8): 1359-1372, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39095657

ABSTRACT

Circular RNA (circRNA) is covalently closed, single-stranded RNA produced by back-splicing. A few circRNAs have been implicated as functional; however, we lack understanding of pathways that are regulated by circRNAs. Here we generated a pooled short-hairpin RNA library targeting the back-splice junction of 3,354 human circRNAs that are expressed at different levels (ranging from low to high) in humans. We used this library for loss-of-function proliferation screens in a panel of 18 cancer cell lines from four tissue types harbouring mutations leading to constitutive activity of defined pathways. Both context-specific and non-specific circRNAs were identified. Some circRNAs were found to directly regulate their precursor, whereas some have a function unrelated to their precursor. We validated these observations with a secondary screen and uncovered a role for circRERE(4-10) and circHUWE1(22,23), two cell-essential circRNAs, circSMAD2(2-6), a WNT pathway regulator, and circMTO1(2,RI,3), a regulator of MAPK signalling. Our work sheds light on pathways regulated by circRNAs and provides a catalogue of circRNAs with a measurable function.


Subject(s)
Cell Proliferation , RNA, Circular , RNA, Circular/genetics , RNA, Circular/metabolism , Humans , Cell Proliferation/genetics , Cell Line, Tumor , Wnt Signaling Pathway/genetics , Signal Transduction , RNA/genetics , RNA/metabolism , RNA Splicing , Gene Expression Regulation, Neoplastic , Gene Library
8.
Mol Genet Genomic Med ; 12(8): e2507, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39132856

ABSTRACT

BACKGROUND: Mucopolysaccharidosis type I (MPS-I) is a rare autosomal recessive genetic lysosomal storage disorder that is caused by pathogenic variants of the α-L-iduronidase (IDUA) gene. This study aimed to identify the genetic causes of MPS-I in a Chinese patient and construct a minigene of IDUA to analyze its variants upon splicing. METHODS: Whole-exome sequencing (WES) and Sanger sequencing were used to confirm the potential causative variants. Single-nucleotide polymorphism (SNP) array was subsequently performed to confirm uniparental disomy (UPD). Minigene assay was performed to analyze the effect on splicing of mRNA. We meanwhile explored the conservative analysis and protein homology simulation. RESULTS: A novel homozygous splicing mutation of IDUA, c.159-9T>A, was identified in an individual presenting with overlapping features of MPS-I. Interestingly, only the father and sisters, but not the mother, carried the variant in a heterozygous state. WES and SNP array analyses validated paternal UPD on chromosome 4. Minigene splicing revealed two aberrant splicing events: exon 2 skipping and intron 1 retention. Moreover, the specific structure of the mutant protein obviously changed according to the results of the homologous model. CONCLUSIONS: This study describes a rare autosomal recessive disorder with paternal UPD of chromosome 4 leading to the homozygosity of the IDUA splicing variant in patients with MPS-I for the first time. This study expands the variant spectrum of IDUA and provides insights into the splicing system, facilitating its enhanced diagnosis and treatment.


Subject(s)
Chromosomes, Human, Pair 4 , Homozygote , Iduronidase , Mucopolysaccharidosis I , RNA Splicing , Uniparental Disomy , Humans , Uniparental Disomy/genetics , Uniparental Disomy/pathology , Iduronidase/genetics , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/pathology , Male , Chromosomes, Human, Pair 4/genetics , Female , Polymorphism, Single Nucleotide , Mutation , East Asian People
9.
RNA Biol ; 21(1): 32-45, 2024 Jan.
Article in English | MEDLINE | ID: mdl-39115224

ABSTRACT

Interactions between double-stranded RNA (dsRNA) and proteins play an important role in cellular homeostasis by regulating the editing, stability, and splicing of intracellular RNA. The identification of dsRNA-binding proteins (dsRBPs) is key; however, it has long been challenging to purify dsRBPs from cells. In this study, we developed a novel method, dsRBPC (dsRNA-binding protein capture), to purify cellular dsRBPs based on classic phase separation purification procedures. A global dsRNA-binding proteome of LLC-PK1 cells was obtained, and we identified 1326 dsRBPs, including 1303 putative novel dsRBPs. Functional analyses suggested that these enriched dsRBPs are mainly associated with rRNA processing, RNA splicing, transcriptional regulation, and nucleocytoplasmic transport. We also found that the ARM (armadillo/beta-catenin-like repeats) motif is a previously unknown dsRNA-binding domain, as demonstrated by biochemical experiments. Collectively, this study provides a useful approach for dsRBP identification and the discovery of a global dsRNA-binding proteome to comprehensively map the dsRNA - protein interaction network.


Subject(s)
RNA, Double-Stranded , RNA-Binding Proteins , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/chemistry , Animals , Proteome/metabolism , Protein Binding , Swine , Cell Line , RNA Splicing , Phase Separation
10.
Cells ; 13(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39120292

ABSTRACT

Biallelic variants in USH2A are associated with retinitis pigmentosa (RP) and Type 2 Usher Syndrome (USH2), leading to impaired vision and, additionally, hearing loss in the latter. Although the introduction of next-generation sequencing into clinical diagnostics has led to a significant uplift in molecular diagnostic rates, many patients remain molecularly unsolved. It is thought that non-coding variants or variants of uncertain significance contribute significantly to this diagnostic gap. This study aims to demonstrate the clinical utility of the reverse transcription-polymerase chain reaction (RT-PCR)-Oxford Nanopore Technology (ONT) sequencing of USH2A mRNA transcripts from nasal epithelial cells to determine the splice-altering effect of candidate variants. Five affected individuals with USH2 or non-syndromic RP who had undergone whole genome sequencing were recruited for further investigation. All individuals had uncertain genotypes in USH2A, including deep intronic rare variants, c.8682-654C>G, c.9055+389G>A, and c.9959-2971C>T; a synonymous variant of uncertain significance, c.2139C>T; p.(Gly713=); and a predicted loss of function duplication spanning an intron/exon boundary, c.3812-3_3837dup p.(Met1280Ter). In silico assessment using SpliceAI provided splice-altering predictions for all candidate variants which were investigated using ONT sequencing. All predictions were found to be accurate; however, in the case of c.3812-3_3837dup, the outcome was a complex cryptic splicing pattern with predominant in-frame exon 18 skipping and a low level of exon 18 inclusion leading to the predicted stop gain. This study detected and functionally characterised simple and complex mis-splicing patterns in USH2A arising from previously unknown deep intronic variants and previously reported variants of uncertain significance, confirming the pathogenicity of the variants.


Subject(s)
Extracellular Matrix Proteins , RNA Splicing , Usher Syndromes , Humans , Extracellular Matrix Proteins/genetics , Usher Syndromes/genetics , Female , Male , RNA Splicing/genetics , High-Throughput Nucleotide Sequencing/methods , Exons/genetics , Mutation/genetics , Retinitis Pigmentosa/genetics , Adult , RNA, Messenger/genetics , RNA, Messenger/metabolism , Introns/genetics , Middle Aged
11.
Mol Biol Rep ; 51(1): 899, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39115759

ABSTRACT

BACKGROUND: Global developmental delay with speech and behavioral abnormalities (OMIM: 619243) is an autosomal dominant disease caused by variants in TNRC6B gene. METHOD: We reviewed and summarized clinical manifestations and genotypes in patients previously reported with TNRC6B gene variants. We used several prediction tools to predict pathogenicity and performed minigene assays to verify the function of the synonymous variant affecting RNA splicing. RESULT: The patient presented with convulsive seizures and developmental delay. WES combined with functional studies diagnosed a child with a synonymous variant in TNRC6B gene. Through minigene assay and Sanger sequencing, we demonstrated that c.3141G > A variant induced exon 7 skipping and the synonymous variant was pathogenic. CONCLUSION: Synonymous variants do not change the amino acids encoded by the codon, so we usually consider synonymous variants to be benign and ignore their pathogenicity. Minigene assay is a valuable tool to identify the effect of variation on RNA splicing and identify synonymous variants' benign or pathogenic. We showed that the synonymous variant was pathogenic by minigene assay. WES combined with minigene assay establishes a robust basis for genetic counseling and diagnosing diseases.


Subject(s)
RNA Splicing , Humans , RNA Splicing/genetics , Developmental Disabilities/genetics , Exons/genetics , Male , Silent Mutation , Exome Sequencing/methods , Female , Genotype , Child , Child, Preschool
12.
Cell Mol Biol Lett ; 29(1): 106, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095708

ABSTRACT

BACKGROUND: The RNA N6-methyladenosine (m6A) modification has become an essential hotspot in epigenetic modulation. Serine-arginine protein kinase 1 (SRPK1) is associated with the pathogenesis of various cancers. However, the m6A modification of SRPK1 and its association with the mechanism of in lung adenocarcinoma (LUAD) remains unclear. METHODS: Western blotting and polymerase chain reaction (PCR) analyses were carried out to identify gene and protein expression. m6A epitranscriptomic microarray was utilized to the assess m6A profile. Loss and gain-of-function assays were carried out elucidate the impact of METTL3 and SRPK1 on LUAD glycolysis and tumorigenesis. RNA immunoprecipitation (RIP), m6A RNA immunoprecipitation (MeRIP), and RNA stability tests were employed to elucidate the SRPK1's METTL3-mediated m6A modification mechanism in LUAD. Metabolic quantification and co-immunoprecipitation assays were applied to investigate the molecular mechanism by which SRPK1 mediates LUAD metabolism. RESULTS: The epitranscriptomic microarray assay revealed that SRPK1 could be hypermethylated and upregulated in LUAD. The main transmethylase METTL3 was upregulated and induced the aberrant high m6A levels of SRPK1. Mechanistically, SRPK1's m6A sites were directly methylated by METTL3, which also stabilized SRPK1 in an IGF2BP2-dependent manner. Methylated SRPK1 subsequently promoted LUAD progression through enhancing glycolysis. Further metabolic quantification, co-immunoprecipitation and western blot assays revealed that SRPK1 interacts with hnRNPA1, an important modulator of PKM splicing, and thus facilitates glycolysis by upregulating PKM2 in LUAD. Nevertheless, METTL3 inhibitor STM2457 can reverse the above effects in vitro and in vivo by suppressing SRPK1 and glycolysis in LUAD. CONCLUSION: It was revealed that in LUAD, aberrantly expressed METTL3 upregulated SRPK1 levels via an m6A-IGF2BP2-dependent mechanism. METTL3-induced SRPK1 fostered LUAD cell proliferation by enhancing glycolysis, and the small-molecule inhibitor STM2457 of METTL3 could be an alternative novel therapeutic strategy for individuals with LUAD.


Subject(s)
Adenocarcinoma of Lung , Adenosine , Glycolysis , Lung Neoplasms , Methyltransferases , Protein Serine-Threonine Kinases , Humans , Adenosine/analogs & derivatives , Adenosine/metabolism , Glycolysis/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Methyltransferases/metabolism , Methyltransferases/genetics , Animals , Gene Expression Regulation, Neoplastic , Mice , Cell Line, Tumor , Mice, Nude , RNA Splicing/genetics , Thyroid Hormone-Binding Proteins , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cell Proliferation/genetics
14.
Proc Natl Acad Sci U S A ; 121(35): e2401781121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39159368

ABSTRACT

Recessive dystrophic epidermolysis bullosa (RDEB) is a rare and most often severe genetic disease characterized by recurrent blistering and erosions of the skin and mucous membranes after minor trauma, leading to major local and systemic complications. The disease is caused by loss-of-function variants in COL7A1 encoding type VII collagen (C7), the main component of anchoring fibrils, which form attachment structures stabilizing the cutaneous basement membrane zone. Alterations in C7 protein structure and/or expression lead to abnormal, rare or absent anchoring fibrils resulting in loss of dermal-epidermal adherence and skin blistering. To date, more than 1,200 distinct COL7A1 deleterious variants have been reported and 19% are splice variants. Here, we describe two RDEB patients for whom we identified two pathogenic deep intronic pathogenic variants in COL7A1. One of these variants (c.7795-97C > G) promotes the inclusion of a pseudoexon between exons 104 and 105 in the COL7A1 transcript, while the other causes partial or complete retention of intron 51. We used antisense oligonucleotide (ASO) mediated exon skipping to correct these aberrant splicing events in vitro. This led to increased normal mRNA splicing above 94% and restoration of C7 protein expression at a level (up to 56%) that should be sufficient to reverse the phenotype. This first report of exon skipping applied to counteract deep intronic variants in COL7A1 represents a promising therapeutic strategy for personalized medicine directed at patients with intronic variants at a distance of consensus splice sites.


Subject(s)
Collagen Type VII , Epidermolysis Bullosa Dystrophica , Introns , RNA Splicing , Collagen Type VII/genetics , Collagen Type VII/metabolism , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/pathology , Humans , Introns/genetics , Male , Female , Exons/genetics , Oligonucleotides, Antisense/genetics
15.
BMC Plant Biol ; 24(1): 768, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134938

ABSTRACT

BACKGROUND: In recent years, covalent modifications on RNA nucleotides have emerged as pivotal moieties influencing the structure, function, and regulatory processes of RNA Polymerase II transcripts such as mRNAs and lncRNAs. However, our understanding of their biological roles and whether these roles are conserved across eukaryotes remains limited. RESULTS: In this study, we leveraged standard polyadenylation-enriched RNA-sequencing data to identify and characterize RNA modifications that introduce base-pairing errors into cDNA reads. Our investigation incorporated data from three Poaceae (Zea mays, Sorghum bicolor, and Setaria italica), as well as publicly available data from a range of stress and genetic contexts in Sorghum and Arabidopsis thaliana. We uncovered a strong enrichment of RNA covalent modifications (RCMs) deposited on a conserved core set of nuclear mRNAs involved in photosynthesis and translation across these species. However, the cohort of modified transcripts changed based on environmental context and developmental program, a pattern that was also conserved across flowering plants. We determined that RCMs can partly explain accession-level differences in drought tolerance in Sorghum, with stress-associated genes receiving a higher level of RCMs in a drought tolerant accession. To address function, we determined that RCMs are significantly enriched near exon junctions within coding regions, suggesting an association with splicing. Intriguingly, we found that these base-pair disrupting RCMs are associated with stable mRNAs, are highly correlated with protein abundance, and thus likely associated with facilitating translation. CONCLUSIONS: Our data point to a conserved role for RCMs in mRNA stability and translation across the flowering plant lineage.


Subject(s)
Arabidopsis , RNA Splicing , Arabidopsis/genetics , Arabidopsis/metabolism , Sorghum/genetics , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Plant/genetics , Zea mays/genetics , Setaria Plant/genetics , Setaria Plant/metabolism , Gene Expression Regulation, Plant , Magnoliopsida/genetics , RNA Processing, Post-Transcriptional
16.
BMC Med Genomics ; 17(1): 206, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39135028

ABSTRACT

BACKGROUND: Hereditary spherocytosis (HS, MIM#612641) is one of the most common hereditary hemolytic disorders. This study aimed to confirm a novel variant's pathogenicity and reveal a patient's genetic etiology. METHODS: The clinical data of a patient with HS who underwent genetic sequencing at the Children's Hospital of Chongqing Medical University were reviewed retrospectively. In silico prediction and in vitro minigene splicing reporter system were then conducted on the detected variant to analyze its intramolecular impact. A summary of the literature related to HS due to SPTB gene variants was also presented. RESULTS: A novel variant (c.301-2 A > G) in the SPTB gene (NM_001024858.4) was identified in the proband. Using Sanger sequencing, we conclusively confirmed that the inheritance of the variant could not be traced to the biological parents. The in vitro minigene assay revealed three different transcripts derived from the c.301-2 A > G variant: r.301_474del, r.301_306delCCAAAG, and r.301-1_301-57ins. Through a literature review, patients with HS who had been genotypically validated were summarized and the SPTB gene variant profile was mapped. CONCLUSION: We identified a splicing variant of the SPTB gene, thus confirming its aberrant translation. The novel variant was the probable genetic etiology of the proband with HS. Our findings expanded the variant spectrum of the SPTB gene, thus improving the understanding of the associated hereditary hemolytic disorders from a clinical and molecular perspective and contributing to the foundation of genetic counseling and diagnosis.


Subject(s)
Spectrin , Spherocytosis, Hereditary , Humans , Spherocytosis, Hereditary/genetics , Spectrin/genetics , Male , Female , Pedigree , Mutation , RNA Splicing
17.
Methods Mol Biol ; 2824: 373-383, 2024.
Article in English | MEDLINE | ID: mdl-39039424

ABSTRACT

RNAseq is a valuable tool that can aid researchers in uncovering the transcriptional changes that occur when a viral pathogen infects a host cell. Viral infection will invariably cause differential expression of many genes, from transcription of mRNA to alternative splicing and degradation. This change in gene expression can be a result of immune activation or a direct activity of the virus to alter the host cell's environment to make it more favorable for viral replication. Studying the innate immune response to a pathogen can reveal which cellular pathways are active, indicating the steps that the host takes to halt viral infection, and detecting virus-mediated mRNA expression changes can help with identifying the pathways which may be exploited by the virus. Gene expression changes-both cell-caused and virus-caused-can be studied through RNAseq, helping to provide a clearer picture of the cellular changes that occur during viral infection. In this protocol, we outline methods to carry out mRNA sequencing in Rift Valley fever virus-infected cell cultures, from infection to library prep and analysis.


Subject(s)
Rift Valley Fever , Rift Valley fever virus , Rift Valley fever virus/genetics , Rift Valley fever virus/physiology , Humans , Rift Valley Fever/virology , Rift Valley Fever/genetics , Host-Pathogen Interactions/genetics , Sequence Analysis, RNA/methods , RNA, Messenger/genetics , RNA, Messenger/metabolism , Animals , Virus Replication/genetics , Alternative Splicing , RNA Splicing , Transcription, Genetic , Cell Line
18.
BMC Med Genomics ; 17(1): 189, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39020321

ABSTRACT

BACKGROUND: This study aims to analyze the pathogenic gene in a Chinese family with non-syndromic hearing loss and identify a novel mutation site in the TNC gene. METHODS: A five-generation Chinese family from Anhui Province, presenting with autosomal dominant non-syndromic hearing loss, was recruited for this study. By analyzing the family history, conducting clinical examinations, and performing genetic analysis, we have thoroughly investigated potential pathogenic factors in this family. The peripheral blood samples were obtained from 20 family members, and the pathogenic genes were identified through whole exome sequencing. Subsequently, the mutation of gene locus was confirmed using Sanger sequencing. The conservation of TNC mutation sites was assessed using Clustal Omega software. We utilized functional prediction software including dbscSNV_AdaBoost, dbscSNV_RandomForest, NNSplice, NetGene2, and Mutation Taster to accurately predict the pathogenicity of these mutations. Furthermore, exon deletions were validated through RT-PCR analysis. RESULTS: The family exhibited autosomal dominant, progressive, post-lingual, non-syndromic hearing loss. A novel synonymous variant (c.5247A > T, p.Gly1749Gly) in TNC was identified in affected members. This variant is situated at the exon-intron junction boundary towards the end of exon 18. Notably, glycine residue at position 1749 is highly conserved across various species. Bioinformatics analysis indicates that this synonymous mutation leads to the disruption of the 5' end donor splicing site in the 18th intron of the TNC gene. Meanwhile, verification experiments have demonstrated that this synonymous mutation disrupts the splicing process of exon 18, leading to complete exon 18 skipping and direct splicing between exons 17 and 19. CONCLUSION: This novel splice-altering variant (c.5247A > T, p.Gly1749Gly) in exon 18 of the TNC gene disrupts normal gene splicing and causes hearing loss among HBD families.


Subject(s)
Pedigree , Humans , Male , Female , Hearing Loss/genetics , Adult , Asian People/genetics , Genes, Dominant , Mutation , RNA Splicing , Middle Aged , China , Exons , East Asian People , Extracellular Matrix Proteins , GPI-Linked Proteins
19.
Mol Cell ; 84(14): 2618-2633.e10, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39025073

ABSTRACT

The twenty-three Fanconi anemia (FA) proteins cooperate in the FA/BRCA pathway to repair DNA interstrand cross-links (ICLs). The cell division cycle and apoptosis regulator 1 (CCAR1) protein is also a regulator of ICL repair, though its possible function in the FA/BRCA pathway remains unknown. Here, we demonstrate that CCAR1 plays a unique upstream role in the FA/BRCA pathway and is required for FANCA protein expression in human cells. Interestingly, CCAR1 co-immunoprecipitates with FANCA pre-mRNA and is required for FANCA mRNA processing. Loss of CCAR1 results in retention of a poison exon in the FANCA transcript, thereby leading to reduced FANCA protein expression. A unique domain of CCAR1, the EF hand domain, is required for interaction with the U2AF heterodimer of the spliceosome and for excision of the poison exon. Taken together, CCAR1 is a splicing modulator required for normal splicing of the FANCA mRNA and other mRNAs involved in various cellular pathways.


Subject(s)
Apoptosis Regulatory Proteins , Cell Cycle Proteins , Fanconi Anemia Complementation Group A Protein , Fanconi Anemia , RNA Splicing , Splicing Factor U2AF , Humans , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , DNA Repair , Endodeoxyribonucleases , Exons , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , HEK293 Cells , HeLa Cells , Protein Binding , RNA Precursors/metabolism , RNA Precursors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Spliceosomes/metabolism , Spliceosomes/genetics , Splicing Factor U2AF/metabolism , Splicing Factor U2AF/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism
20.
Nat Methods ; 21(8): 1466-1469, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39054391

ABSTRACT

Here we present biVI, which combines the variational autoencoder framework of scVI with biophysical models describing the transcription and splicing kinetics of RNA molecules. We demonstrate on simulated and experimental single-cell RNA sequencing data that biVI retains the variational autoencoder's ability to capture cell type structure in a low-dimensional space while further enabling genome-wide exploration of the biophysical mechanisms, such as system burst sizes and degradation rates, that underlie observations.


Subject(s)
Sequence Analysis, RNA , Single-Cell Analysis , Single-Cell Analysis/methods , Sequence Analysis, RNA/methods , Humans , RNA Splicing , Algorithms , RNA/genetics , RNA/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL