Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.731
Filter
1.
Nat Commun ; 15(1): 7067, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152130

ABSTRACT

RNA-binding proteins (RBPs) have pivotal functions in RNA metabolism, but current methods are limited in retrieving RBP-RNA interactions within endogenous biological contexts. Here, we develop INSCRIBE (IN situ Sensitive Capture of RNA-protein Interactions in Biological Environments), circumventing the challenges through in situ RNA labeling by precisely directing a purified APOBEC1-nanobody fusion to the RBP of interest. This method enables highly specific RNA-binding site identification across a diverse range of fixed biological samples such as HEK293T cells and mouse brain tissue and accurately identifies the canonical binding motifs of RBFOX2 (UGCAUG) and TDP-43 (UGUGUG) in native cellular environments. Applicable to any RBP with available primary antibodies, INSCRIBE enables sensitive capture of RBP-RNA interactions from ultra-low input equivalent to ~5 cells. The robust, versatile, and sensitive INSCRIBE workflow is particularly beneficial for precious tissues such as clinical samples, empowering the exploration of genuine RBP-RNA interactions in RNA-related disease contexts.


Subject(s)
DNA-Binding Proteins , RNA Splicing Factors , RNA-Binding Proteins , RNA , Humans , Animals , HEK293 Cells , Mice , RNA-Binding Proteins/metabolism , RNA/metabolism , RNA Splicing Factors/metabolism , DNA-Binding Proteins/metabolism , Binding Sites , Protein Binding , Brain/metabolism , Repressor Proteins
3.
J Exp Clin Cancer Res ; 43(1): 217, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39098911

ABSTRACT

Aberrant alternative splicing events play a critical role in cancer biology, contributing to tumor invasion, metastasis, epithelial-mesenchymal transition, and drug resistance. Recent studies have shown that alternative splicing is a key feature for transcriptomic variations in colorectal cancer, which ranks third among malignant tumors worldwide in both incidence and mortality. Long non-coding RNAs can modulate this process by acting as trans-regulatory agents, recruiting splicing factors, or driving them to specific targeted genes. LncH19 is a lncRNA dis-regulated in several tumor types and, in colorectal cancer, it plays a critical role in tumor onset, progression, and metastasis. In this paper, we found, that in colorectal cancer cells, the long non-coding RNA H19 can bind immature RNAs and splicing factors as hnRNPM and RBFOX2. Through bioinformatic analysis, we identified 57 transcripts associated with lncH19 and containing binding sites for both splicing factors, hnRNPM, and RBFOX2. Among these transcripts, we identified the mRNA of the GTPase-RAC1, whose alternatively spliced isoform, RAC1B, has been ascribed several roles in the malignant transformation. We confirmed, in vitro, the binding of the splicing factors to both the transcripts RAC1 and lncH19. Loss and gain of expression experiments in two colorectal cancer cell lines (SW620 and HCT116) demonstrated that lncH19 is required for RAC1B expression and, through RAC1B, it induces c-Myc and Cyclin-D increase. In vivo, investigation from biopsies of colorectal cancer patients showed higher levels of all the explored genes (lncH19, RAC1B, c-Myc and Cyclin-D) concerning the healthy counterpart, thus supporting our in vitro model. In addition, we identified a positive correlation between lncH19 and RAC1B in colorectal cancer patients. Finally, we demonstrated that lncH19, as a shuttle, drives the splicing factors RBFOX2 and hnRNPM to RAC1 allowing exon retention and RAC1B expression. The data shown in this paper represent the first evidence of a new mechanism of action by which lncH19 carries out its functions as an oncogene by prompting colorectal cancer through the modulation of alternative splicing.


Subject(s)
Alternative Splicing , Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , RNA Splicing Factors , RNA, Long Noncoding , rac1 GTP-Binding Protein , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , RNA, Long Noncoding/genetics , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Cell Line, Tumor , Heterogeneous-Nuclear Ribonucleoprotein Group M/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group M/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
4.
Nat Commun ; 15(1): 6925, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138201

ABSTRACT

Sex chromosomes underlie the development of male or female sex organs across species. While systemic signals derived from sex organs prominently contribute to sex-linked differences, it is unclear whether the intrinsic presence of sex chromosomes in somatic tissues has a specific function. Here, we use genetic tools to show that cellular sex is crucial for sexual differentiation throughout the body in Drosophila melanogaster. We reveal that every somatic cell converts the intrinsic presence of sex chromosomes into the active production of a sex determinant, a female specific serine- and arginine-rich (SR) splicing factor. This discovery dismisses the mosaic model which posits that only a subset of cells has the potential to sexually differentiate. Using cell-specific sex reversals, we show that this prevalence of cellular sex drives sex differences in organ size and body weight and is essential for fecundity. These findings demonstrate that cellular sex drives differentiation programs at an organismal scale and highlight the importance of cellular sex pathways in sex trait evolution.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Sex Chromosomes , Sex Differentiation , Animals , Male , Female , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Sex Differentiation/genetics , Sex Differentiation/physiology , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Sex Chromosomes/genetics , Fertility/genetics , Sex Characteristics , Organ Size , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , Body Weight , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics
5.
Clin Transl Med ; 14(8): e1784, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39135292

ABSTRACT

BACKGROUND: The modification of N6-methyladenosine (m6A) plays a pivotal role in tumor by altering both innate and adaptive immune systems through various pathways, including the regulation of messenger RNA. The YTH domain protein family, acting as "readers" of m6A modifications, affects RNA splicing, stability, and immunogenicity, thereby playing essential roles in immune regulation and antitumor immunity. Despite their significance, the impact of the YTH domain protein family on tumor initiation and progression, as well as their involvement in tumor immune regulation and therapy, remains underexplored and lacks comprehensive review. CONCLUSION: This review introduces the molecular characteristics of the YTH domain protein family and their physiological and pathological roles in biological behavior, emphasizing their mechanisms in regulating immune responses and antitumor immunity. Additionally, the review discusses the roles of the YTH domain protein family in immune-related diseases and tumor resistance, highlighting that abnormal expression or dysfunction of YTH proteins is closely linked to tumor resistance. KEY POINTS: This review provides an in-depth understanding of the YTH domain protein family in immune regulation and antitumor immunity, suggesting new strategies and directions for immunotherapy of related diseases. These insights not only deepen our comprehension of m6A modifications and YTH protein functions but also pave the way for future research and clinical applications.


Subject(s)
Immunomodulation , Immunotherapy , Neoplasms , Humans , Neoplasms/immunology , Neoplasms/therapy , Immunotherapy/methods , RNA-Binding Proteins/immunology , RNA-Binding Proteins/genetics , RNA Splicing Factors/genetics , RNA Splicing Factors/immunology
6.
Retina ; 44(9): 1580-1589, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39167579

ABSTRACT

PURPOSE: To determine the association between gene-expression profiling (GEP), next-generation sequencing (NGS), preferentially expressed antigen in melanoma (PRAME) features, and metastatic risk in patients with uveal melanoma (UM). METHODS: A retrospective analysis of patients with UM treated by brachytherapy or enucleation by a single ocular oncologist was conducted from November 2020 and July 2022. Clinicopathologic features, patient outcomes, GEP classification, NGS, and PRAME results were recorded. RESULTS: Comprehensive GEP, PRAME, and NGS testing was performed on 135 UMs. The presence of eukaryotic translation initiation factor 1A, X-chromosomal and splicing factor 3B subunit 1 mutations was significantly associated with GEP class 1A and GEP class 1B, respectively. The presence of BRCA- associated protein-1 mutation was significantly associated with GEP class 2. The average largest basal diameter for tumors with eukaryotic translation initiation factor 1A, X-chromosomal mutations was significantly smaller than those with splicing factor 3B subunit 1 mutations and BRCA1-associated protein-1 mutations. Class 2 tumors metastasized sooner than GEP class 1 tumors. Tumors with splicing factor 3B subunit 1 and/or BRCA1-associated protein-1 mutations metastasized sooner compared with tumors that had either no driver mutation or no mutations at all. Tumors with splicing factor 3B subunit 1 did not have a significantly different time to metastasis compared with tumors with BRCA1-associated protein-1 (P value = 0.97). Forty tumors (30%) were PRAME positive, and the remaining 95 tumors (70%) were PRAME negative. Tumors with PRAME-positive status did not have a significantly different time to metastasis compared with tumors without PRAME-positive status (P value = 0.11). CONCLUSION: GEP, NGS, and PRAME expression analysis help determine different levels of metastatic risk in UM. Although other prognostic tests exist, the following study reports on the use of NGS for metastatic prognostication in UM. However, limitations of NGS exist, especially with small lesions that are technically difficult to biopsy.


Subject(s)
Antigens, Neoplasm , Biomarkers, Tumor , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Melanoma , Uveal Neoplasms , Humans , Uveal Neoplasms/genetics , Uveal Neoplasms/diagnosis , Melanoma/genetics , Retrospective Studies , Male , Female , Middle Aged , Antigens, Neoplasm/genetics , Gene Expression Profiling/methods , Aged , Biomarkers, Tumor/genetics , Mutation , Adult , Gene Expression Regulation, Neoplastic , Aged, 80 and over , Eukaryotic Initiation Factor-1/genetics , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Brachytherapy , Phosphoproteins , Tumor Suppressor Proteins , Ubiquitin Thiolesterase
7.
Cell Death Dis ; 15(8): 554, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090090

ABSTRACT

Glioma is the most common primary brain tumor, and targeting glioma stem cells (GSCs) has become a key aspect of glioma treatment. In this study, we discovered a molecular network in which circRNA forms an R-loop structure with its parental gene to regulate the biological behavior of GSCs. Genes with abnormal expression in GSCs were screened using RNA-seq and circRNA microarray analyses. The study results showed that high expression of YTHDC1 in GSCs promoted the transportation of N6-methyladenosine (m6A)-modified circPOLR2B from the nucleus to the cytoplasm. Decreased circPOLR2B levels in the nucleus resulted in fewer R-loop structures formed with its parental gene POLR2B. This reduction in R-loop structures relieved the inhibitory effect on POLR2B transcription and upregulated PBX1 expression through alternative polyadenylation (APA) action, thereby promoting the malignant biological behavior of GSCs. Knockdown of YTHDC1, POLR2B, and PBX1 reduced xenograft tumor volume and prolonged the survival of nude mice. The YTHDC1/circPOLR2B/POLR2B/PBX1 axis plays a regulatory role in the biological behavior of GSCs, offering potential targets and novel strategies for the treatment of glioma.


Subject(s)
Glioma , Neoplastic Stem Cells , RNA, Circular , Animals , Humans , Mice , Adenosine/analogs & derivatives , Adenosine/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Cell Line, Tumor , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Methylation , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , RNA, Circular/genetics , RNA, Circular/metabolism
8.
Biol Direct ; 19(1): 54, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38978074

ABSTRACT

OBJECTIVE: Retinal vascular endothelial cell (RVECs) injury is a major cause of morbidity and mortality among the patients with diabetes. RVECs dysfunction is the predominant pathological manifestation of vascular complication in diabetic retinopathy. N6-methyladenosine (m6A) serves as the most prevalent modification in eukaryotic mRNAs. However, the role of m6A RNA modification in RVECs dysfunction is still unclear. METHODS: RT-qPCR analysis and western blot were conducted to detect the change of m6A RNA modification in diabetic retinopathy. CCK-8 assay, transwell experiment, wound healing assay, tube formation experiment, m6A-IP-qPCR were performed to determine the role of YTHDC1 in RVECs. Retinal trypsin digestion test and H&E staining were used to evaluate histopathological changes. RESULTS: The levels of m6A RNA methylation were significantly up-regulated in HG-induced RVECs, which were caused by increased expression of YTHDC1. YTHDC1 regulated the viability, proliferation, migration and tube formation ability in vitro. YTHDC1 overexpression impaired RVECs function by repressing CDK6 expression, which was mediated by YTHDC1-dependent mRNA decay. Moreover, it showed sh-YTHDC1 inhibited CDK6 nuclear export. Sh-YTHDC1 promotes the mRNA degradation of CDK6 in the nucleus but does not affect the cytoplasmic CDK6 mRNA. In vivo experiments showed that overexpression of CDK6 reversed the protective effect of sh-YTHDC1 on STZ-induced retinal tissue damage. CONCLUSION: YTHDC1-mediated m6A methylation regulates diabetes-induced RVECs dysfunction. YTHDC1-CDK6 signaling axis could be therapeutically targeted for treating DR.


Subject(s)
Adenosine , Cyclin-Dependent Kinase 6 , Diabetic Retinopathy , Endothelial Cells , Glucose , Endothelial Cells/metabolism , Animals , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase 6/genetics , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/genetics , Adenosine/analogs & derivatives , Adenosine/metabolism , Glucose/metabolism , Glucose/pharmacology , Humans , Retina/metabolism , Male , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , Cell Proliferation , Nerve Tissue Proteins
9.
J Clin Invest ; 134(14)2024 May 16.
Article in English | MEDLINE | ID: mdl-39007267

ABSTRACT

Emerging evidence has linked the dysregulation of N6-methyladenosine (m6A) modification to inflammation and inflammatory diseases, but the underlying mechanism still needs investigation. Here, we found that high levels of m6A modification in a variety of hyperinflammatory states are p65-dependent because Wilms tumor 1-associated protein (WTAP), a key component of the "writer" complex, is transcriptionally regulated by p65, and its overexpression can lead to increased levels of m6A modification. Mechanistically, upregulated WTAP is more prone to phase separation to facilitate the aggregation of the writer complex to nuclear speckles and the deposition of m6A marks on transcriptionally active inflammatory transcripts, thereby accelerating the proinflammatory response. Further, a myeloid deficiency in WTAP attenuates the severity of LPS-induced sepsis and DSS-induced IBD. Thus, the proinflammatory effect of WTAP is a general risk-increasing mechanism, and interrupting the assembly of the m6A writer complex to reduce the global m6A levels by targeting the phase separation of WTAP may be a potential and promising therapeutic strategy for alleviating hyperinflammation.


Subject(s)
Adenosine , Cell Cycle Proteins , Inflammation , RNA Splicing Factors , Animals , Humans , Mice , Adenosine/metabolism , Adenosine/analogs & derivatives , Disease Models, Animal , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides , Mice, Knockout , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Sepsis/metabolism , Sepsis/genetics , Sepsis/pathology , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics
10.
EMBO J ; 43(16): 3494-3522, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38951610

ABSTRACT

Cells have evolved a robust and highly regulated DNA damage response to preserve their genomic integrity. Although increasing evidence highlights the relevance of RNA regulation, our understanding of its impact on a fully efficient DNA damage response remains limited. Here, through a targeted CRISPR-knockout screen, we identify RNA-binding proteins and modifiers that participate in the p53 response. Among the top hits, we find the m6A reader YTHDC1 as a master regulator of p53 expression. YTHDC1 binds to the transcription start sites of TP53 and other genes involved in the DNA damage response, promoting their transcriptional elongation. YTHDC1 deficiency also causes the retention of introns and therefore aberrant protein production of key DNA damage factors. While YTHDC1-mediated intron retention requires m6A, TP53 transcriptional pause-release is promoted by YTHDC1 independently of m6A. Depletion of YTHDC1 causes genomic instability and aberrant cancer cell proliferation mediated by genes regulated by YTHDC1. Our results uncover YTHDC1 as an orchestrator of the DNA damage response through distinct mechanisms of co-transcriptional mRNA regulation.


Subject(s)
DNA Damage , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Adenosine/metabolism , Adenosine/analogs & derivatives , Genomic Instability , Cell Proliferation , Gene Expression Regulation , Nerve Tissue Proteins
11.
Front Immunol ; 15: 1423378, 2024.
Article in English | MEDLINE | ID: mdl-39081311

ABSTRACT

Periodontitis, delineated by the destruction of structures that support teeth, is predominantly propelled by intricate immune responses. Immunomodulatory treatments offer considerable promise for the management of this ailment; however, the modulation of the periodontal immune microenvironment to facilitate tissue regeneration presents a substantial biomedical challenge. Herein, our study investigates the role of Wilms' tumor 1-associating protein (WTAP), a critical m6A methyltransferase, in the immunomodulation of periodontitis and assesses its viability as a therapeutic target. We observed heightened expression of WTAP in macrophages extracted from gingival tissues impacted by periodontitis, with a strong association with M1 polarization. Via loss-of-function experiments, we demonstrated that diminishing WTAP expression precipitates a transition from M1 to M2 macrophage phenotypes amidst inflammatory conditions, thus improving the periodontal immune landscape. Further, RNA sequencing and indirect co-culture assays indicated that suppressing of WTAP expression modulates osteoimmune responses and enhances the osteogenic differentiation of bone marrow stromal cells. The local deployment of adeno-associated virus-shWTAP in murine models of periodontitis robustly validated the therapeutic promise of targeting WTAP in this disease. Collectively, our findings highlight the crucial role of WTAP in orchestrating macrophage-mediated osteoimmune responses and tissue regeneration in periodontitis, proposing novel avenues for immunotherapeutic interventions in its treatment.


Subject(s)
Cell Cycle Proteins , Macrophages , Osteogenesis , Periodontitis , RNA Splicing Factors , Animals , Humans , Male , Mice , Cell Differentiation , Disease Models, Animal , Gingiva/immunology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Osteogenesis/immunology , Osteogenesis/genetics , Periodontitis/immunology , Periodontitis/therapy , Regeneration , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism
12.
J Bioenerg Biomembr ; 56(4): 405-418, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38842666

ABSTRACT

BACKGROUND: Ferritinophagy-mediated ferroptosis plays a crucial role in fighting pathogen aggression. The long non-coding RNA Mir22hg is involved in the regulation of ferroptosis and aberrantly overexpression in lipopolysaccharide (LPS)-induced sepsis mice, but whether it regulates sepsis through ferritinophagy-mediated ferroptosis is unclear. METHODS: Mir22hg was screened by bioinformatics analysis. Ferroptosis was assessed by assaying malondialdehyde (MDA), reactive oxygen species (ROS), and Fe2+ levels, glutathione (GSH) activity, as well as ferroptosis-related proteins GPX4 and SLC3A2 by using matched kits and performing western blot. Ferritinophagy was assessed by Lyso tracker staining and FerroOrange staining, immunofluorescence analysis of Ferritin and LC-3, and western blot analysis of LC-3II/I, p62, FTH1, and NCOA4. The bind of YTH domain containing 1 (YTHDC1) to Mir22hg or angiopoietin-like-4 (Angptl4) was verified by RNA pull-down and/or immunoprecipitation (RIP) assays. RESULTS: Mir22hg silencing lightened ferroptosis and ferritinophagy in LPS-induced MLE-12 cells and sepsis mouse models, as presented by the downregulated MDA, ROS, Fe2+, NCOA4, and SLC3A2 levels, upregulated GPX4, GSH, and FTH1 levels, along with a decrease in autophagy. Mir22hg could bind to the m6A reader YTHDC1 without affecting its expression. Mechanistically, Mir22hg enhanced Angptl4 mRNA stability through recruiting the m6A reader YTHDC1. Furthermore, Angptl4 overexpression partly overturned Mir22hg inhibition-mediated effects on ferroptosis and ferritinophagy in LPS-induced MLE-12 cells. CONCLUSION: Mir22hg contributed to in ferritinophagy-mediated ferroptosis in sepsis via recruiting the m6A reader YTHDC1 and strengthening Angptl4 mRNA stability, highlighting that Mir22hg may be a potential target for sepsis treatment based on ferroptosis.


Subject(s)
Angiopoietin-Like Protein 4 , Ferroptosis , MicroRNAs , Sepsis , Animals , Humans , Male , Mice , Angiopoietin-Like Protein 4/metabolism , Angiopoietin-Like Protein 4/genetics , Autophagy/physiology , Ferritins/metabolism , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics , RNA Stability , Sepsis/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism
13.
J Affect Disord ; 361: 97-103, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38834091

ABSTRACT

BACKGROUND: Multiple genes might interact to determine the age at onset of bipolar disorder. We investigated gene-gene interactions related to age at onset of bipolar disorder in the Korean population, using genome-wide association study (GWAS) data. METHODS: The study population consisted of 303 patients with bipolar disorder. First, the top 1000 significant single-nucleotide polymorphisms (SNPs) associated with age at onset of bipolar disorder were selected through single SNP analysis by simple linear regression. Subsequently, the QMDR method was used to find gene-gene interactions. RESULTS: The best 10 SNPs from simple regression were located in chromosome 1, 2, 3, 10, 11, 14, 19, and 21. Only five SNPs were found in several genes, such as FOXN3, KIAA1217, OPCML, CAMSAP2, and PTPRS. On QMDR analyses, five pairs of SNPs showed significant interactions with a CVC exceeding 1/5 in a two-locus model. The best interaction was found for the pair of rs60830549 and rs12952733 (CVC = 1/5, P < 1E-07). In three-locus models, four combinations of SNPs showed significant associations with age at onset, with a CVC of >1/5. The best three-locus combination was rs60830549, rs12952733, and rs12952733 (CVC = 2/5, P < 1E-6). The SNPs showing significant interactions were located in the KIAA1217, RBFOX3, SDK2, CYP19A1, NTM, SMYD3, and RBFOX1 genes. CONCLUSIONS: Our analysis confirmed genetic interactions influencing the age of onset for bipolar disorder and identified several potential candidate genes. Further exploration of the functions of these promising genes, which may have multiple roles within the neuronal network, is necessary.


Subject(s)
Age of Onset , Bipolar Disorder , Epistasis, Genetic , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Adult , Female , Humans , Male , Middle Aged , Bipolar Disorder/genetics , Genetic Predisposition to Disease , Republic of Korea , RNA Splicing Factors/genetics , East Asian People/genetics
14.
Int J Biol Sci ; 20(8): 3173-3184, 2024.
Article in English | MEDLINE | ID: mdl-38904016

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) poses significant challenges in terms of prognosis and treatment. Recent research has identified splicing deregulation as a new cancer hallmark. Herein, we investigated the largely uncharacterized alternative splicing profile and the key splicing factor SF3B1 in PDAC pancreatic cells and tissues as a potential discovery source of plausible drug targets and new predictive biomarkers of clinical outcome. The research involved a transcriptome-wide analysis, comparing profiles of splicing profiles in PDAC primary cells with normal ductal cells. This revealed more than 400 significant differential splicing events in genes involved in regulation of gene expression, primarily related to mRNA splicing, and metabolism of nucleic acids. PDAC cultures were highly sensitive to the SF3B1 modulators, E7107 and Pladienolide-B, showing IC50s in the low nanomolar range. These compounds induced apoptosis, associated to induction of the MCL-1/S splice variant. and reduced cell migration, associated to RON mis-splicing. In an orthotopic mouse model, E7107 showed promising results. Furthermore, we evaluated SF3B1 expression in specimens from 87 patients and found a significant association of SF3B1 expression with progression-free and overall survival. In conclusion, SF3B1 emerges as both a potential prognostic factor and therapeutic target in PDAC, impacting cell proliferation, migration, and apoptosis. These findings warrant future studies on this new therapeutic strategy against PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , RNA Splicing Factors , Humans , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/drug therapy , Animals , Mice , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Prognosis , Phosphoproteins/metabolism , Phosphoproteins/genetics , Macrolides/therapeutic use , Macrolides/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , RNA Splicing , Alternative Splicing , Female , Cell Movement/genetics
15.
Discov Med ; 36(185): 1289-1297, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38926115

ABSTRACT

BACKGROUND: Genetic mutations play a crucial role in the development and progression of myelodysplastic syndromes (MDS), impacting the immune microenvironment and influencing the choice of treatment regimen, as well as the efficacy and prognosis of patients. The objective of this study was to examine variations in hematological and immunological characteristics associated with common gene mutations in MDS patients and establish a foundation for the precise treatment of MDS. METHODS: The hematological, immunological, and other clinical features of 71 recently diagnosed MDS patients from January 1, 2019, to July 31, 2023, were retrospectively analyzed. These patients were categorized based on their gene mutations, and the variances in hematological and immunological characteristics among distinct groups were compared. RESULTS: Hematological variances were observed among different gene mutation groups. Specifically, platelet counts in the splicing factor 3B subunit 1 (SF3B1) mutation group were notably higher compared to the wild-type group (p = 0.009). Conversely, in the additional sex combs like 1 (ASXL1) mutation groups, monocyte ratios were significantly elevated in comparison to the wild-type group (p = 0.046), and in the ten-eleven translocation 2 (TET2) mutation group, lymphocyte ratios were significantly lower (p = 0.022). Additionally, the leukocyte (p = 0.005), neutrophil ratio (p = 0.002), and lymphocyte ratio (p = 0.001) were significantly higher in the Runt-related transcription factor 1 (RUNX1) mutation group. Regarding immunological distinctions, the Natural Killer (NK) cell ratio demonstrated a significant increase in the SF3B1 mutation group (p = 0.005). Moreover, the TET2 mutation group exhibited a significantly higher Interleukin-8 (IL-8) level (p = 0.017). In contrast, the U2 small nuclear RNA auxiliary factor 1 (U2AF1) group displayed significantly lower levels of IL-1ß (p = 0.033), IL-10 (p = 0.033), and Tumour Necrosis Factor-α (TNF-α) (p = 0.009). CONCLUSION: Distinct variations exist in the immune microenvironment of MDS associated with different genetic mutations. Further studies are imperative to delve into the underlying mechanisms that drive these differences.


Subject(s)
Dioxygenases , Mutation , Myelodysplastic Syndromes , RNA Splicing Factors , Humans , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/blood , Female , Male , Middle Aged , Aged , RNA Splicing Factors/genetics , Retrospective Studies , Adult , Aged, 80 and over , DNA-Binding Proteins/genetics , Phosphoproteins/genetics , Phosphoproteins/immunology , Killer Cells, Natural/immunology , Core Binding Factor Alpha 2 Subunit/genetics , Platelet Count , Repressor Proteins
16.
J Exp Med ; 221(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38861022

ABSTRACT

The IL-17 receptor adaptor molecule Act1, an RNA-binding protein, plays a critical role in IL-17-mediated cancer progression. Here, we report a novel mechanism of how IL-17/Act1 induces chemoresistance by modulating redox homeostasis through epitranscriptomic regulation of antioxidant RNA metabolism. Transcriptome-wide mapping of direct Act1-RNA interactions revealed that Act1 binds to the 5'UTR of antioxidant mRNAs and Wilms' tumor 1-associating protein (WTAP), a key regulator in m6A methyltransferase complex. Strikingly, Act1's binding sites are located in proximity to m6A modification sites, which allows Act1 to promote the recruitment of elF3G for cap-independent translation. Loss of Act1's RNA binding activity or Wtap knockdown abolished IL-17-induced m6A modification and translation of Wtap and antioxidant mRNAs, indicating a feedforward mechanism of the Act1-WTAP loop. We then developed antisense oligonucleotides (Wtap ASO) that specifically disrupt Act1's binding to Wtap mRNA, abolishing IL-17/Act1-WTAP-mediated antioxidant protein production during chemotherapy. Wtap ASO substantially increased the antitumor efficacy of cisplatin, demonstrating a potential therapeutic strategy for chemoresistance.


Subject(s)
Antioxidants , Drug Resistance, Neoplasm , Homeostasis , Oxidation-Reduction , Drug Resistance, Neoplasm/genetics , Humans , Antioxidants/metabolism , Antioxidants/pharmacology , Animals , Cell Line, Tumor , RNA, Messenger/metabolism , RNA, Messenger/genetics , Interleukin-17/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mice , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , 5' Untranslated Regions , Cisplatin/pharmacology , RNA Splicing Factors
17.
Genes (Basel) ; 15(6)2024 May 23.
Article in English | MEDLINE | ID: mdl-38927612

ABSTRACT

The current investigation endeavors to identify differentially expressed alternatively spliced (DAS) genes that exhibit concordant expression with splicing factors (SFs) under diverse multifactorial abiotic stress combinations in Arabidopsis seedlings. SFs serve as the post-transcriptional mechanism governing the spatiotemporal dynamics of gene expression. The different stresses encompass variations in salt concentration, heat, intensive light, and their combinations. Clusters demonstrating consistent expression profiles were surveyed to pinpoint DAS/SF gene pairs exhibiting concordant expression. Through rigorous selection criteria, which incorporate alignment with documented gene functionalities and expression patterns observed in this study, four members of the serine/arginine-rich (SR) gene family were delineated as SFs concordantly expressed with six DAS genes. These regulated SF genes encompass cactin, SR1-like, SR30, and SC35-like. The identified concordantly expressed DAS genes encode diverse proteins such as the 26.5 kDa heat shock protein, chaperone protein DnaJ, potassium channel GORK, calcium-binding EF hand family protein, DEAD-box RNA helicase, and 1-aminocyclopropane-1-carboxylate synthase 6. Among the concordantly expressed DAS/SF gene pairs, SR30/DEAD-box RNA helicase, and SC35-like/1-aminocyclopropane-1-carboxylate synthase 6 emerge as promising candidates, necessitating further examinations to ascertain whether these SFs orchestrate splicing of the respective DAS genes. This study contributes to a deeper comprehension of the varied responses of the splicing machinery to abiotic stresses. Leveraging these DAS/SF associations shows promise for elucidating avenues for augmenting breeding programs aimed at fortifying cultivated plants against heat and intensive light stresses.


Subject(s)
Alternative Splicing , Arabidopsis Proteins , Arabidopsis , Gene Expression Regulation, Plant , Stress, Physiological , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Stress, Physiological/genetics , Seedlings/genetics , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism
18.
Sci Adv ; 10(26): eadl1049, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38924416

ABSTRACT

A long-standing goal of evolutionary biology is to decode how changes in gene regulatory networks contribute to human-specific traits. Human accelerated regions (HARs) are prime candidates for driving gene regulatory modifications in human development. The RBFOX1 locus is densely populated with HARs, providing a set of potential regulatory elements that could have changed its expression in the human lineage. Here, we examined the role of RBFOX1-HARs using transgenic zebrafish reporter assays and identified 15 transcriptional enhancers that are active in the developing nervous system, 9 of which displayed differential activity between the human and chimpanzee sequences. The engineered loss of two selected RBFOX1-HARs in knockout mouse models modified Rbfox1 expression at specific developmental stages and tissues in the brain, influencing the expression and splicing of a high number of Rbfox1 target genes. Our results provided insight into the spatial and temporal changes in gene expression driven by RBFOX1-HARs.


Subject(s)
Enhancer Elements, Genetic , Evolution, Molecular , RNA Splicing Factors , Zebrafish , Humans , Animals , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Zebrafish/genetics , Mice , Gene Expression Regulation, Developmental , Mice, Knockout , Animals, Genetically Modified , Gene Regulatory Networks , Pan troglodytes/genetics , Genetic Loci
19.
Viruses ; 16(6)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38932230

ABSTRACT

Type I interferons (IFN-Is) are pivotal in innate immunity against human immunodeficiency virus I (HIV-1) by eliciting the expression of IFN-stimulated genes (ISGs), which encompass potent host restriction factors. While ISGs restrict the viral replication within the host cell by targeting various stages of the viral life cycle, the lesser-known IFN-repressed genes (IRepGs), including RNA-binding proteins (RBPs), affect the viral replication by altering the expression of the host dependency factors that are essential for efficient HIV-1 gene expression. Both the host restriction and dependency factors determine the viral replication efficiency; however, the understanding of the IRepGs implicated in HIV-1 infection remains greatly limited at present. This review provides a comprehensive overview of the current understanding regarding the impact of the RNA-binding protein families, specifically the two families of splicing-associated proteins SRSF and hnRNP, on HIV-1 gene expression and viral replication. Since the recent findings show specifically that SRSF1 and hnRNP A0 are regulated by IFN-I in various cell lines and primary cells, including intestinal lamina propria mononuclear cells (LPMCs) and peripheral blood mononuclear cells (PBMCs), we particularly discuss their role in the context of the innate immunity affecting HIV-1 replication.


Subject(s)
HIV Infections , HIV-1 , Immunity, Innate , Virus Replication , HIV-1/genetics , HIV-1/physiology , Humans , HIV Infections/virology , HIV Infections/genetics , HIV Infections/immunology , Gene Expression Regulation, Viral , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , Interferon Type I/metabolism , Interferon Type I/genetics , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Interferons/metabolism , Interferons/genetics , Interferons/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
20.
Neuroscience ; 553: 56-73, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-38945353

ABSTRACT

BACKGROUND: N6-methyladenosine (m6A) is one of the most extensive RNA methylation modifications in eukaryotes and participates in the pathogenesis of numerous diseases including ischemic stroke. Peripheral blood neutrophils are forerunners after ischemic brain injury and exert crucial functions. This study aims to explore the transcriptional profiles of m6A modification in neutrophils of patients with ischemic stroke. RESULTS: We found that the expression levels of m6A regulators FTO and YTHDC1 were notably decreased in the neutrophils following ischemic stroke, and FTO expression was negatively correlated with neutrophil counts and neutrophil-to-lymphocyte ratio (NLR). The m6A mRNA&lncRNA epigenetic transcriptome microarray identified 416 significantly upregulated and 500 significantly downregulated mRNA peaks in neutrophils of ischemic stroke patients. Moreover, 48 mRNAs and 18 lncRNAs were hypermethylated, and 115 mRNAs and 29 lncRNAs were hypomethylated after cerebral ischemia. Gene ontology (GO) analysis identified that these m6A-modified mRNAs were primarily enriched in calcium ion transport, long-term synaptic potentiation, and base-excision repair. The signaling pathways involved were EGFR tyrosine kinase inhibitor resistance, ErbB, and base excision repair signaling pathway. MeRIP-qPCR validation results showed that NRG1 and GDPD1 were significantly hypermethylated, and LIG1, CHRND, lncRNA RP11-442J17.2, and lncRNA RP11-600P1.2 were significantly hypomethylated after cerebral ischemia. Moreover, the expression levels of major m6A regulators Mettl3, Fto, Ythdf1, and Ythdf3 were obviously declined in the brain and leukocytes of post-stroke mouse models. CONCLUSION: This study explored the RNA m6A methylation pattern in the neutrophils of ischemic stroke patients, indicating that it is an intervention target of epigenetic regulation in ischemic stroke.


Subject(s)
Adenosine , Ischemic Stroke , Neutrophils , RNA-Binding Proteins , Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , Ischemic Stroke/metabolism , Ischemic Stroke/genetics , Neutrophils/metabolism , Male , Humans , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Mice , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Middle Aged , RNA, Messenger/metabolism , Female , Mice, Inbred C57BL , Aged , Brain Ischemia/metabolism , Brain Ischemia/genetics , Transcriptome , RNA Splicing Factors , Nerve Tissue Proteins
SELECTION OF CITATIONS
SEARCH DETAIL