Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.686
Filter
2.
Biomolecules ; 14(6)2024 May 23.
Article in English | MEDLINE | ID: mdl-38927017

ABSTRACT

Renal interstitial fibrosis (RIF) is a classic pathophysiological process of chronic kidney disease (CKD). However, the mechanisms underlying RIF remain unclear. The present study found that a novel circular RNA, cirInpp5b, might be involved in RIF by high-throughput sequencing. Subsequent experiments revealed that circInpp5b was reduced in UUO mouse kidney tissues and TGF-ß1-treated proximal tubular cells. The overexpression of circInpp5b inhibited RIF in UUO mice and prevented extracellular matrix (ECM) deposition in TGF-ß1-treated proximal tubular cells. Furthermore, overexpression of circInpp5b down-regulated the protein level of DDX1. Mechanistically, circInpp5b bound to the DDX1 protein and promoted its lysosomal degradation. Collectively, the findings of our study demonstrate that circInpp5b ameliorates RIF by binding to the DDX1 protein and promoting its lysosomal degradation.


Subject(s)
DEAD-box RNA Helicases , Fibrosis , Lysosomes , RNA, Circular , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Animals , Mice , Lysosomes/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Proteolysis , Male , Mice, Inbred C57BL , Humans , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/genetics
3.
Int J Mol Sci ; 25(12)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38928053

ABSTRACT

The innate immune response in Salmo salar, mediated by pattern recognition receptors (PRRs), is crucial for defending against pathogens. This study examined DDX41 protein functions as a cytosolic/nuclear sensor for cyclic dinucleotides, RNA, and DNA from invasive intracellular bacteria. The investigation determined the existence, conservation, and functional expression of the ddx41 gene in S. salar. In silico predictions and experimental validations identified a single ddx41 gene on chromosome 5 in S. salar, showing 83.92% homology with its human counterpart. Transcriptomic analysis in salmon head kidney confirmed gene transcriptional integrity. Proteomic identification through mass spectrometry characterized three unique peptides with 99.99% statistical confidence. Phylogenetic analysis demonstrated significant evolutionary conservation across species. Functional gene expression analysis in SHK-1 cells infected by Piscirickettsia salmonis and Renibacterium salmoninarum indicated significant upregulation of DDX41, correlated with increased proinflammatory cytokine levels and activation of irf3 and interferon signaling pathways. In vivo studies corroborated DDX41 activation in immune responses, particularly when S. salar was challenged with P. salmonis, underscoring its potential in enhancing disease resistance. This is the first study to identify the DDX41 pathway as a key component in S. salar innate immune response to invading pathogens, establishing a basis for future research in salmonid disease resistance.


Subject(s)
Fish Diseases , Immunity, Innate , Phylogeny , Piscirickettsia , Piscirickettsiaceae Infections , Renibacterium , Salmo salar , Animals , Piscirickettsia/genetics , Immunity, Innate/genetics , Salmo salar/microbiology , Salmo salar/genetics , Salmo salar/immunology , Fish Diseases/microbiology , Fish Diseases/immunology , Fish Diseases/genetics , Piscirickettsiaceae Infections/microbiology , Piscirickettsiaceae Infections/immunology , Piscirickettsiaceae Infections/genetics , Piscirickettsiaceae Infections/veterinary , Renibacterium/genetics , Renibacterium/immunology , Fish Proteins/genetics , Fish Proteins/metabolism , Fish Proteins/immunology , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Evolution, Molecular
4.
Biochem Soc Trans ; 52(3): 1131-1148, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38884803

ABSTRACT

The RIG-I-like receptors (RLRs), comprising retinoic acid-inducible gene I (RIG-I), melanoma differentiation-associated gene 5 (MDA5), and laboratory of genetics and physiology 2 (LGP2), are pattern recognition receptors belonging to the DExD/H-box RNA helicase family of proteins. RLRs detect viral RNAs in the cytoplasm and respond by initiating a robust antiviral response that up-regulates interferon and cytokine production. RIG-I and MDA5 complement each other by recognizing different RNA features, and LGP2 regulates their activation. RIG-I's multilayered RNA recognition and proofreading mechanisms ensure accurate viral RNA detection while averting harmful responses to host RNAs. RIG-I's C-terminal domain targets 5'-triphosphate double-stranded RNA (dsRNA) blunt ends, while an intrinsic gating mechanism prevents the helicase domains from non-specifically engaging with host RNAs. The ATPase and RNA translocation activity of RIG-I adds another layer of selectivity by minimizing the lifetime of RIG-I on non-specific RNAs, preventing off-target activation. The versatility of RIG-I's ATPase function also amplifies downstream signaling by enhancing the signaling domain (CARDs) exposure on 5'-triphosphate dsRNA and promoting oligomerization. In this review, we offer an in-depth understanding of the mechanisms RIG-I uses to facilitate viral RNA sensing and regulate downstream activation of the immune system.


Subject(s)
DEAD Box Protein 58 , Immunity, Innate , RNA, Viral , Receptors, Immunologic , Humans , RNA, Viral/metabolism , DEAD Box Protein 58/metabolism , Receptors, Immunologic/metabolism , Animals , RNA, Double-Stranded/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , DEAD-box RNA Helicases/metabolism , RNA Helicases/metabolism
5.
Genes (Basel) ; 15(6)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38927693

ABSTRACT

The identification and expression of germ cells are important for studying sex-related mechanisms in fish. The vasa gene, encoding an ATP-dependent RNA helicase, is recognized as a molecular marker of germ cells and plays a crucial role in germ cell development. Silurus asotus, an important freshwater economic fish species in China, shows significant sex dimorphism with the female growing faster than the male. However, the molecular mechanisms underlying these sex differences especially involving in the vasa gene in this fish remain poorly understood. In this work, the vasa gene sequence of S. asotus (named as Savasa) was obtained through RT-PCR and rapid amplification of cDNA end (RACE), and its expression in embryos and tissues was analyzed using qRT-PCR and an in situ hybridization method. Letrozole (LT) treatment on the larvae fish was also conducted to investigate its influence on the gene. The results revealed that the open reading frame (ORF) of Savasa was 1989 bp, encoding 662 amino acids. The SaVasa protein contains 10 conserved domains unique to the DEAD-box protein family, showing the highest sequence identity of 95.92% with that of Silurus meridionalis. In embryos, Savasa is highly expressed from the two-cell stage to the blastula stage in early embryos, with a gradually decreasing trend from the gastrula stage to the heart-beating stage. Furthermore, Savasa was initially detected at the end of the cleavage furrow during the two-cell stage, later condensing into four symmetrical cell clusters with embryonic development. At the gastrula stage, Savasa-positive cells increased and began to migrate towards the dorsal side of the embryo. In tissues, Savasa is predominantly expressed in the ovaries, with almost no or lower expression in other detected tissues. Moreover, Savasa was expressed in phase I-V oocytes in the ovaries, as well as in spermatogonia and spermatocytes in the testis, implying a specific expression pattern of germ cells. In addition, LT significantly upregulated the expression of Savasa in a concentration-dependent manner during the key gonadal differentiation period of the fish. Notably, at 120 dph after LT treatment, Savasa expression was the lowest in the testis and ovary of the high concentration group. Collectively, findings from gene structure, protein sequence, phylogenetic analysis, RNA expression patterns, and response to LT suggest that Savasa is maternally inherited with conserved features, serving as a potential marker gene for germ cells in S.asotus, and might participate in LT-induced early embryonic development and gonadal development processes of the fish. This would provide a basis for further research on the application of germ cell markers and the molecular mechanisms of sex differences in S. asotus.


Subject(s)
Catfishes , DEAD-box RNA Helicases , Fish Proteins , Letrozole , Animals , Letrozole/pharmacology , Female , Male , Fish Proteins/genetics , Fish Proteins/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Catfishes/genetics , Catfishes/growth & development , Catfishes/metabolism , Gene Expression Regulation, Developmental/drug effects , Germ Cells/metabolism , Germ Cells/drug effects , Germ Cells/growth & development , Phylogeny
6.
Biochem Biophys Res Commun ; 725: 150228, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38936167

ABSTRACT

The DEAD-box family is the largest family of RNA helicases (RHs), playing crucial roles in RNA metabolism and plant stress resistance. In this study, we report that an RNA helicase, RH12, positively regulates plant salt tolerance, as rh12 knockout mutants exhibit heightened sensitivity to salt stress. Further analysis indicates that RH12 is involved in the abscisic acid (ABA) response, as rh12 knockout mutants show increased sensitivity to ABA. Examination of reactive oxygen species (ROS) revealed that RH12 helps inhibit ROS accumulation under salt stress during seed germination. Additionally, RH12 accelerates the degradation of specific germination-related transcripts. In conclusion, our results demonstrate that RH12 plays multiple roles in the salt stress response in Arabidopsis.


Subject(s)
Abscisic Acid , Arabidopsis Proteins , Arabidopsis , DEAD-box RNA Helicases , Germination , Salt Tolerance , Seeds , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/growth & development , Germination/genetics , Salt Tolerance/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Seeds/genetics , Seeds/growth & development , Seeds/metabolism , Abscisic Acid/metabolism , Gene Expression Regulation, Plant , Reactive Oxygen Species/metabolism
7.
Nat Commun ; 15(1): 4772, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858384

ABSTRACT

The underlying mechanisms of atherosclerosis, the second leading cause of death among Werner syndrome (WS) patients, are not fully understood. Here, we establish an in vitro co-culture system using macrophages (iMφs), vascular endothelial cells (iVECs), and vascular smooth muscle cells (iVSMCs) derived from induced pluripotent stem cells. In co-culture, WS-iMφs induces endothelial dysfunction in WS-iVECs and characteristics of the synthetic phenotype in WS-iVSMCs. Transcriptomics and open chromatin analysis reveal accelerated activation of type I interferon signaling and reduced chromatin accessibility of several transcriptional binding sites required for cellular homeostasis in WS-iMφs. Furthermore, the H3K9me3 levels show an inverse correlation with retrotransposable elements, and retrotransposable element-derived double-stranded RNA activates the DExH-box helicase 58 (DHX58)-dependent cytoplasmic RNA sensing pathway in WS-iMφs. Conversely, silencing type I interferon signaling in WS-iMφs rescues cell proliferation and suppresses cellular senescence and inflammation. These findings suggest that Mφ-specific inhibition of type I interferon signaling could be targeted to treat atherosclerosis in WS patients.


Subject(s)
Atherosclerosis , Inflammation , Interferon Type I , Macrophages , Retroelements , Werner Syndrome , Interferon Type I/metabolism , Werner Syndrome/genetics , Werner Syndrome/metabolism , Humans , Atherosclerosis/metabolism , Atherosclerosis/immunology , Atherosclerosis/genetics , Atherosclerosis/pathology , Macrophages/metabolism , Macrophages/immunology , Retroelements/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , Induced Pluripotent Stem Cells/metabolism , Signal Transduction , Coculture Techniques , Myocytes, Smooth Muscle/metabolism , Endothelial Cells/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Cellular Senescence , Cell Proliferation
8.
Mol Med ; 30(1): 75, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834947

ABSTRACT

BACKGROUND: Liver kinase B1 (LKB1) is frequently mutated in lung adenocarcinoma, and its loss contributes to tumor progression. METHODS: To identify LKB1 downstream genes that promote lung adenocarcinoma aggressiveness, we performed bioinformatical analysis using publicly available datasets. RESULTS: Rab3B was upregulated in LKB1-depleted lung adenocarcinoma cells and suppressed by LKB1 overexpression. CREB protein was enriched at the promoter of Rab3B in lung cancer cells. Silencing of CREB abrogated the upregulation of Rab3B upon LKB1 loss. Immunohistochemistry revealed the elevated expression of Rab3B in lung adenocarcinomas relative to adjacent normal tissues. Upregulation of Rab3B was significantly associated with lymph node metastasis, advanced tumor stage, and reduced overall survival in lung adenocarcinoma patients. Knockdown of Rab3B suppressed and overexpression of Rab3B promoted the proliferation, colony formation, and migration of lung adenocarcinoma cells in vitro. In a mouse xenograft model, Rab3B depletion restrained and Rab3B overexpression augmented the growth of lung adenocarcinoma tumors. Mechanistically, Rab3B interacted with DDX6 and enhanced its protein stability. Ectopic expression of DDX6 significantly promoted the proliferation, colony formation, and migration of lung adenocarcinoma cells. DDX6 knockdown phenocopied the effects of Rab3B depletion on lung adenocarcinoma cells. Additionally, DDX6 overexpression partially rescued the aggressive phenotype of Rab3B-depleted lung adenocarcinoma cells. CONCLUSION: LKB1 deficiency promotes Rab3B upregulation via a CREB-dependent manner. Rab3B interacts with and stabilizes DDX6 protein to accelerate lung adenocarcinoma progression. The Rab3B-DDX6 axis may be potential therapeutic target for lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung , DEAD-box RNA Helicases , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Animals , Female , Humans , Male , Mice , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , AMP-Activated Protein Kinase Kinases/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cyclic AMP Response Element-Binding Protein/metabolism , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Stability
9.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38928290

ABSTRACT

Influenza virus infection poses a great threat to human health globally each year. Non-coding RNAs (ncRNAs) in the human genome have been reported to participate in the replication process of the influenza virus, among which there are still many unknowns about Long Intergenic Non-Coding RNAs (LincRNAs) in the cell cycle of viral infections. Here, we observed an increased expression of Linc01615 in A549 cells upon influenza virus PR8 infection, accompanied by the successful activation of the intracellular immune system. The knockdown of Linc01615 using the shRNAs promoted the proliferation of the influenza A virus, and the intracellular immune system was inhibited, in which the expressions of IFN-ß, IL-28A, IL-29, ISG-15, MX1, and MX2 were decreased. Predictions from the catRAPID website suggested a potential interaction between Linc01615 and DHX9. Also, knocking down Linc01615 promoted influenza virus proliferation. The subsequent transcriptome sequencing results indicated a decrease in Linc01615 expression after influenza virus infection when DHX9 was knocked down. Further analysis through cross-linking immunoprecipitation and high-throughput sequencing (CLIP-seq) in HEK293 cells stably expressing DHX9 confirmed the interaction between DHX9 and Linc01615. We speculate that DHX9 may interact with Linc01615 to partake in influenza virus replication and that Linc01615 helps to activate the intracellular immune system. These findings suggest a deeper connection between DHX9 and Linc01615, which highlights the significant role of Linc01615 in the influenza virus replication process. This research provides valuable insights into understanding influenza virus replication and offers new targets for preventing influenza virus infections.


Subject(s)
DEAD-box RNA Helicases , Influenza, Human , RNA, Long Noncoding , Virus Replication , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , A549 Cells , HEK293 Cells , Influenza, Human/virology , Influenza, Human/genetics , Influenza, Human/immunology , Influenza, Human/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Influenza A virus/physiology , Animals , Dogs , Gene Knockdown Techniques , Neoplasm Proteins
10.
Molecules ; 29(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38931009

ABSTRACT

The DEAD-box RNA helicase Ded1 is an essential yeast protein involved in translation initiation that belongs to the DDX3 subfamily. The purified Ded1 protein is an ATP-dependent RNA-binding protein and an RNA-dependent ATPase, but it was previously found to lack substrate specificity and enzymatic regulation. Here we demonstrate through yeast genetics, yeast extract pull-down experiments, in situ localization, and in vitro biochemical approaches that Ded1 is associated with, and regulated by, the signal recognition particle (SRP), which is a universally conserved ribonucleoprotein complex required for the co-translational translocation of polypeptides into the endoplasmic reticulum lumen and membrane. Ded1 is physically associated with SRP components in vivo and in vitro. Ded1 is genetically linked with SRP proteins. Finally, the enzymatic activity of Ded1 is inhibited by SRP21 in the presence of SCR1 RNA. We propose a model where Ded1 actively participates in the translocation of proteins during translation. Our results provide a new understanding of the role of Ded1 during translation.


Subject(s)
DEAD-box RNA Helicases , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Signal Recognition Particle , Signal Recognition Particle/metabolism , Signal Recognition Particle/genetics , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Protein Binding , Protein Biosynthesis , Protein Transport
11.
Cancer Res Commun ; 4(6): 1495-1504, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38767454

ABSTRACT

Cancer cells with DNA repair defects (e.g., BRCA1/2 mutant cells) are vulnerable to PARP inhibitors (PARPi) due to induction of synthetic lethality. However, recent clinical evidence has shown that PARPi can prevent the growth of some cancers irrespective of their BRCA1/2 status, suggesting alternative mechanisms of action. We previously discovered one such mechanism in breast cancer involving DDX21, an RNA helicase that localizes to the nucleoli of cells and is a target of PARP1. We have now extended this observation in endometrial and ovarian cancers and provided links to patient outcomes. When PARP1-mediated ADPRylation of DDX21 is inhibited by niraparib, DDX21 is mislocalized to the nucleoplasm resulting in decreased rDNA transcription, which leads to a reduction in ribosome biogenesis, protein translation, and ultimately endometrial and ovarian cancer cell growth. High PARP1 expression was associated with high nucleolar localization of DDX21 in both cancers. High nucleolar DDX21 negatively correlated with calculated IC50s for niraparib. By studying endometrial cancer patient samples, we were able to show that high DDX21 nucleolar localization was significantly associated with decreased survival. Our study suggests that the use of PARPi as a cancer therapeutic can be expanded to further types of cancers and that DDX21 localization can potentially be used as a prognostic factor and as a biomarker for response to PARPi. SIGNIFICANCE: Currently, there are no reliable biomarkers for response to PARPi outside of homologous recombination deficiency. Herein we present a unique potential biomarker, with clear functional understanding of the molecular mechanism by which DDX21 nucleolar localization can predict response to PARPi.


Subject(s)
Cell Nucleolus , DEAD-box RNA Helicases , Poly(ADP-ribose) Polymerase Inhibitors , Humans , Female , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Cell Line, Tumor , Ovarian Neoplasms/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Endometrial Neoplasms/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/mortality , Endometrial Neoplasms/metabolism , Piperidines/pharmacology , Piperidines/therapeutic use , Prognosis , Cell Proliferation/drug effects , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/pathology , Genital Neoplasms, Female/drug therapy , Genital Neoplasms, Female/mortality , Genital Neoplasms, Female/metabolism , Indazoles
12.
Life Sci ; 349: 122716, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38762067

ABSTRACT

RNA helicases are involved in almost all biological events, and the DDXs family is one of the largest subfamilies of RNA helicases. Recently, studies have reported that RNA helicase DDX21 is involved in several biological events, specifically in orchestrating gene expression. Hence, in this review, we provide a comprehensive overview of the function of DDX21 in health and diseases. In the genome, DDX21 contributes to genome stability by promoting DNA damage repair and resolving R-loops. It also facilitates transcriptional regulation by directly binding to promoter regions, interacting with transcription factors, and enhancing transcription through non-coding RNA. Moreover, DDX21 is involved in various RNA metabolism such as RNA processing, translation, and decay. Interestingly, the activity and function of DDX21 are regulated by post-translational modifications, which affect the localization and degradation of DDX21. Except for its role of RNA helicase, DDX21 also acts as a non-enzymatic function in unwinding RNA, regulating transcriptional modifications and promoting transcription. Next, we discuss the potential application of DDX21 as a clinical predictor for diseases, which may facilitate providing novel pharmacological targets for molecular therapy.


Subject(s)
DEAD-box RNA Helicases , Gene Expression Regulation , Humans , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Animals , Genomic Instability , Protein Processing, Post-Translational/genetics
13.
Neoplasma ; 71(3): 231-242, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38764294

ABSTRACT

Pancreatic cancer (PAAD) is a fatal malignancy with a poor prognosis. The treatment strategies are quite limited and gemcitabine is the canonical one, which has been proven to improve the prognosis of PAAD patients. However, the treatment efficiency of gemcitabine is far from satisfactory and remains to be further improved. DEAD-Box Helicase 46 (DDX46) is a kind of RNA helicase, which promotes multiple cancers development. However, its role in PAAD is largely unknown. In the present study, we found DDX46 was highly expressed in PAAD tissues and correlated with poor prognosis. Knockdown of DDX46 repressed PAAD cell growth in vitro and in vivo and sensitized PAAD cells to gemcitabine treatment. Mechanically, DDX46 bound to JMJD6 and promoted JMJD6/CDK4 signaling pathway. Overexpression of JMJD6 reversed the anti-tumor function of DDX46 knockdown. Our study found a novel pathological mechanism of PAAD progression and provided a potential therapeutic target to improve gemcitabine efficiency.


Subject(s)
Cyclin-Dependent Kinase 4 , DEAD-box RNA Helicases , Deoxycytidine , Drug Resistance, Neoplasm , Gemcitabine , Jumonji Domain-Containing Histone Demethylases , Pancreatic Neoplasms , Signal Transduction , Humans , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 4/metabolism , Animals , Mice , Cell Proliferation , Prognosis , Antimetabolites, Antineoplastic/pharmacology , Male , Gene Expression Regulation, Neoplastic
14.
Signal Transduct Target Ther ; 9(1): 127, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38782919

ABSTRACT

DEAD-box helicase 17 (DDX17) is a typical member of the DEAD-box family with transcriptional cofactor activity. Although DDX17 is abundantly expressed in the myocardium, its role in heart is not fully understood. We generated cardiomyocyte-specific Ddx17-knockout mice (Ddx17-cKO), cardiomyocyte-specific Ddx17 transgenic mice (Ddx17-Tg), and various models of cardiomyocyte injury and heart failure (HF). DDX17 is downregulated in the myocardium of mouse models of heart failure and cardiomyocyte injury. Cardiomyocyte-specific knockout of Ddx17 promotes autophagic flux blockage and cardiomyocyte apoptosis, leading to progressive cardiac dysfunction, maladaptive remodeling and progression to heart failure. Restoration of DDX17 expression in cardiomyocytes protects cardiac function under pathological conditions. Further studies showed that DDX17 can bind to the transcriptional repressor B-cell lymphoma 6 (BCL6) and inhibit the expression of dynamin-related protein 1 (DRP1). When DDX17 expression is reduced, transcriptional repression of BCL6 is attenuated, leading to increased DRP1 expression and mitochondrial fission, which in turn leads to impaired mitochondrial homeostasis and heart failure. We also investigated the correlation of DDX17 expression with cardiac function and DRP1 expression in myocardial biopsy samples from patients with heart failure. These findings suggest that DDX17 protects cardiac function by promoting mitochondrial homeostasis through the BCL6-DRP1 pathway in heart failure.


Subject(s)
DEAD-box RNA Helicases , Heart Failure , Myocytes, Cardiac , Animals , Humans , Mice , Apoptosis/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Dynamins/genetics , Dynamins/metabolism , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/metabolism , Homeostasis/genetics , Mice, Knockout , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Dynamics/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism
15.
Biochem Pharmacol ; 225: 116251, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38701867

ABSTRACT

Hepatocellular carcinoma (HCC) is the main histological subtype of primary liver cancer and remains one of the most common solid malignancies globally. Ferroptosis was recently defined as an iron-catalyzed form of regulated necrosis. Because cancer cells exhibit higher iron requirements than noncancer cells, treatment with ferroptosis-inducing compounds may be a feasible strategy for cancer therapy. However, cancer cells develop acquired resistance to evade ferroptosis, and the mechanisms responsible for ferroptosis resistance are not fully clarified. In the current study, we reported that DDX39B was downregulated during sorafenib-induced ferroptosis in a dose- and time-dependent manner. Exogenous introduction of DDX39B ensured the survival of HCC cells upon exposure to sorafenib, while the opposite phenomenon was observed in DDX39B-silenced HCC cells. Mechanistically, we demonstrated that DDX39B increased GPX4 levels by promoting the splicing and cytoplasmic translocation of GPX4 pre-mRNA, which was sufficient to detoxify sorafenib-triggered excess lipid ROS production, lipid peroxidation accumulation, ferrous iron levels, and mitochondrial damage. Inhibition of DDX39B ATPase activity by CCT018159 repressed the splicing and cytoplasmic export of GPX4 pre-mRNA and synergistically assisted sorafenib-induced ferroptotic cell death in HCC cells. Taken together, our data uncover a novel role for DDX39B in ferroptosis resistance by modulating the maturation of GPX4 mRNA via a posttranscriptional approach and suggest that DDX39B inhibition may be a promising therapeutic strategy to enhance the sensitivity and vulnerability of HCC cells to sorafenib.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , DEAD-box RNA Helicases , Ferroptosis , Liver Neoplasms , Phospholipid Hydroperoxide Glutathione Peroxidase , RNA Precursors , Sorafenib , Ferroptosis/drug effects , Ferroptosis/physiology , Sorafenib/pharmacology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , RNA Precursors/metabolism , RNA Precursors/genetics , Antineoplastic Agents/pharmacology , Animals , Mice , RNA Splicing/drug effects , Mice, Nude , Cell Line, Tumor , Dose-Response Relationship, Drug , Mice, Inbred BALB C , Male , Cytoplasm/metabolism , Cytoplasm/drug effects
16.
J Mol Biol ; 436(12): 168604, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38729260

ABSTRACT

Posttranscriptional maturation and export from the nucleus to the cytoplasm are essential steps in the normal processing of many cellular RNAs. The RNA helicase UAP56 (U2AF associated protein 56; also known as DDX39B) has emerged as a critical player in facilitating and co-transcriptionally linking these steps. Originally identified as a helicase involved in pre-mRNA splicing, UAP56 has been shown to facilitate formation of the A complex during spliceosome assembly. Additionally, it has been found to be critical for interactions between components of the exon junction and transcription and export complexes to promote the loading of export receptors. Although it appears to be structurally similar to other helicase superfamily 2 members, UAP56's ability to interact with multiple different protein partners allows it to perform its various cellular functions. Herein, we describe the structure-activity relationship studies that identified protein interactions of UAP56 and its human paralog URH49 (UAP56-related helicase 49; also known as DDX39A) and are beginning to reveal molecular mechanisms by which interacting proteins and substrate RNAs may regulate these helicases. We also provide an overview of reports that have demonstrated less well-characterized roles for UAP56, including R-loop resolution and telomere maintenance. Finally, we discuss studies that indicate a potential pathogenic effect of UAP56 in the development of autoimmune diseases and cancer, and identify the association of somatic and genetic mutations in UAP56 with neurodevelopmental disorders.


Subject(s)
DEAD-box RNA Helicases , RNA Splicing , Humans , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/chemistry , Structure-Activity Relationship , Spliceosomes/metabolism , Protein Binding , Animals
17.
Sci Rep ; 14(1): 10963, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38745066

ABSTRACT

MicroRNAs (miRNAs) are sequence-specific inhibitors of post-transcriptional gene expression. However, the physiological functions of these non-coding RNAs in renal interstitial mesenchymal cells remain unclear. To conclusively evaluate the role of miRNAs, we generated conditional knockout (cKO) mice with platelet-derived growth factor receptor-ß (PDGFR-ß)-specific inactivation of the key miRNA pathway gene Dicer. The cKO mice were subjected to unilateral ureteral ligation, and renal interstitial fibrosis was quantitatively evaluated using real-time polymerase chain reaction and immunofluorescence staining. Compared with control mice, cKO mice had exacerbated interstitial fibrosis exhibited by immunofluorescence staining and mRNA expression of PDGFR-ß. A microarray analysis showed decreased expressions of miR-9-5p, miR-344g-3p, and miR-7074-3p in cKO mice compared with those in control mice, suggesting an association with the increased expression of PDGFR-ß. An analysis of the signaling pathways showed that the major transcriptional changes in cKO mice were related to smooth muscle cell differentiation, regulation of DNA metabolic processes and the actin cytoskeleton, positive regulation of fibroblast proliferation and Ras protein signal transduction, and focal adhesion-PI3K/Akt/mTOR signaling pathways. Depletion of Dicer in mesenchymal cells may downregulate the signaling pathway related to miR-9-5p, miR-344g-3p, and miR-7074-3p, which can lead to the progression of chronic kidney disease. These findings highlight the possibility for future diagnostic or therapeutic developments for renal fibrosis using miR-9-5p, miR-344g-3p, and miR-7074-3p.


Subject(s)
Fibrosis , Kidney , Mesenchymal Stem Cells , Mice, Knockout , MicroRNAs , Receptor, Platelet-Derived Growth Factor beta , Ribonuclease III , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Mice , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Kidney/pathology , Kidney/metabolism , Mesenchymal Stem Cells/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Signal Transduction , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Diseases/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Male
18.
Oncotarget ; 15: 313-325, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38753413

ABSTRACT

The diheteroarylamide-based compound 1C8 and the aminothiazole carboxamide-related compound GPS167 inhibit the CLK kinases, and affect the proliferation of a broad range of cancer cell lines. A chemogenomic screen previously performed with GPS167 revealed that the depletion of components associated with mitotic spindle assembly altered sensitivity to GPS167. Here, a similar screen performed with 1C8 also established the impact of components involved in mitotic spindle assembly. Accordingly, transcriptome analyses of cells treated with 1C8 and GPS167 indicated that the expression and RNA splicing of transcripts encoding mitotic spindle assembly components were affected. The functional relevance of the microtubule connection was confirmed by showing that subtoxic concentrations of drugs affecting mitotic spindle assembly increased sensitivity to GPS167. 1C8 and GPS167 impacted the expression and splicing of transcripts in pathways relevant to tumor progression, including MYC targets and the epithelial mesenchymal transition (EMT). Finally, 1C8 and GPS167 altered the expression and alternative splicing of transcripts involved in the antiviral immune response. Consistent with this observation, depleting the double-stranded RNA sensor DHX33 suppressed GPS167-mediated cytotoxicity on HCT116 cells. Our study uncovered molecular mechanisms through which 1C8 and GPS167 affect cancer cell proliferation as well as processes critical for metastasis.


Subject(s)
Cell Proliferation , Epithelial-Mesenchymal Transition , Protein Kinase Inhibitors , Protein-Tyrosine Kinases , Humans , Epithelial-Mesenchymal Transition/drug effects , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Protein Kinase Inhibitors/pharmacology , Cell Proliferation/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Thiazoles/pharmacology , Antiviral Agents/pharmacology , HCT116 Cells , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Gene Expression Profiling
19.
Cell Death Dis ; 15(5): 342, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760378

ABSTRACT

U3 snoRNA is essential for ribosome biogenesis during interphase. Upon mitotic onset, the nucleolus disassembles and U3 snoRNA relocates to the perichromosomal region (PR) to be considered as a chromosome passenger. Whether U3 controls mitosis remains unknown. Here, we demonstrate that U3 snoRNA is required for mitotic progression. We identified DDX21 as the predominant U3-binding protein during mitosis and confirmed that U3 snoRNA colocalizes with DDX21 in the PR. DDX21 knockdown induces mitotic catastrophe and similar mitotic defects caused by U3 snoRNA depletion. Interestingly, the uniform PR distribution of U3 snoRNA and DDX21 is interdependent. DDX21 functions in mitosis depending on its PR localization. Mechanistically, U3 snoRNA regulates DDX21 PR localization through maintaining its mobility. Moreover, Cy5-U3 snoRNA downsizes the fibrous condensates of His-DDX21 at proper molecular ratios in vitro. This work highlights the importance of the equilibrium between U3 snoRNA and DDX21 in PR formation and reveals the potential relationship between the PR assembly and mitotic regulation.


Subject(s)
DEAD-box RNA Helicases , Mitosis , RNA, Small Nucleolar , Humans , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , RNA, Small Nucleolar/metabolism , RNA, Small Nucleolar/genetics , HeLa Cells
20.
Int J Mol Sci ; 25(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38732173

ABSTRACT

Lung adenocarcinoma (LUAD) is the most prevalent and aggressive subtype of lung cancer, exhibiting a dismal prognosis with a five-year survival rate below 5%. DEAD-box RNA helicase 18 (DDX18, gene symbol DDX18), a crucial regulator of RNA metabolism, has been implicated in various cellular processes, including cell cycle control and tumorigenesis. However, its role in LUAD pathogenesis remains elusive. This study demonstrates the significant upregulation of DDX18 in LUAD tissues and its association with poor patient survival (from public databases). Functional in vivo and in vitro assays revealed that DDX18 knockdown potently suppresses LUAD progression. RNA sequencing and chromatin immunoprecipitation experiments identified cyclin-dependent kinase 4 (CDK4), a cell cycle regulator, as a direct transcriptional target of DDX18. Notably, DDX18 depletion induced G1 cell cycle arrest, while its overexpression promoted cell cycle progression even in normal lung cells. Interestingly, while the oncogenic protein c-Myc bound to the DDX18 promoter, it did not influence its expression. Collectively, these findings establish DDX18 as a potential oncogene in LUAD, functioning through the CDK4-mediated cell cycle pathway. DDX18 may represent a promising therapeutic target for LUAD intervention.


Subject(s)
Adenocarcinoma of Lung , Cyclin-Dependent Kinase 4 , DEAD-box RNA Helicases , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Animals , Humans , Mice , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 4/genetics , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Nude , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...