Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
J Biochem ; 175(6): 629-641, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38299728

ABSTRACT

Proper regulation of cellular response to environmental stress is crucial for maintaining biological homeostasis and is achieved by the balance between cell death processes, such as the formation of the pyroptosis-inducing NLRP3 inflammasome, and pro-survival processes, such as stress granule (SG) assembly. However, the functional interplay between these two stress-responsive organelles remains elusive. Here, we identified DHX33, a viral RNA sensor for the NLRP3 inflammasome, as a SG component, and the SG-nucleating protein G3BP as an NLRP3 inflammasome component. We also found that a decrease in intracellular potassium (K+) concentration, a key 'common' step in NLRP3 inflammasome activation, markedly inhibited SG assembly. Therefore, when macrophages are exposed to stress stimuli with the potential to induce both SGs and the NLRP3 inflammasome, such as cytoplasmic poly(I:C) stimulation, they preferentially form the NLRP3 inflammasome but avoid SG assembly by sequestering G3BP into the inflammasome and by inducing a reduction in intracellular K+ levels. Thus, under such conditions, DHX33 is primarily utilized as a viral RNA sensor for the inflammasome. Our data reveal the functional crosstalk between NLRP3 inflammasome-mediated pyroptosis and SG-mediated cell survival pathways and delineate a molecular mechanism that regulates cell-fate decisions and anti-viral innate immunity under stress.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Stress Granules , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Humans , Stress Granules/metabolism , Mice , Animals , Potassium/metabolism , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Pyroptosis , RNA Helicases/metabolism , Macrophages/metabolism , Macrophages/virology , RNA Recognition Motif Proteins/metabolism , Poly I-C/pharmacology , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , DNA Helicases
2.
Biochim Biophys Acta Gen Subj ; 1868(3): 130547, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38143011

ABSTRACT

Tumor suppressor p53 is frequently null or mutated in human cancers. Here in this study, DHX33 protein was found to be induced in p53 null cells in vitro, and in p53 mutant lung tumorigenesis in vivo. Cholesterol metabolism through mevalonate pathway is pivotal for cell proliferation and is frequently altered in human cancers. Mice carrying mutant p53 and KrasG12D alleles showed upregulation of mevalonate pathway gene expression. However upon DHX33 loss, their upregulation was significantly debilitated. Additionally, in many human cancer cells, DHX33 knockdown caused inhibition of mavelonate pathway gene transcription. We propose DHX33 locates downstream of mutant p53 and Ras to regulate mevalonate pathway gene transcription and thereby supports cancer development in vivo.


Subject(s)
Mevalonic Acid , Tumor Suppressor Protein p53 , Humans , Mice , Animals , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Lung/metabolism , Carcinogenesis , Transcription, Genetic , DEAD-box RNA Helicases/genetics
3.
Bioorg Med Chem Lett ; 96: 129505, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37838340

ABSTRACT

RNA helicase DHX33 has been identified to be a critical factor in promoting cancer development. Genetic deletion of DHX33 significantly blocks tumorigenesis. Importantly, its helicase activity was found to be pivotal for exerting cellular functions. Herein we used a helicase-based high throughput screening (HTS) to discover DHX33 inhibitors from Chembridge chemical library containing 15,000 small molecules. We identified a hit compound containing benzimidazole ring that demonstrated activity against DHX33 with certain selectivity. Further structural optimization led to the design and synthesis of a series of analog inhibitors. Considering the potential role of DHX33 in cancer development, the compounds were evaluated based on the cytotoxicity activity in U251-MG cancer cells in vitro. Among them, compound IVa (KY386) was identified to be a selective inhibitor for DHX33 helicase with potent anti-cancer activity and moderate metabolic stability. These results support the promising role of DHX33 inhibitors for development of novel anti-cancer drugs.


Subject(s)
Antineoplastic Agents , Antineoplastic Agents/pharmacology
4.
Cell Signal ; 110: 110832, 2023 10.
Article in English | MEDLINE | ID: mdl-37543097

ABSTRACT

RNA helicase DHX33 has been shown to be aberrantly expressed in various human cancers, however, its role in tumorigenesis remains incompletely understood. In this report, we uncovered that a family of DNA architecture proteins, HMGBs, can be regulated by DHX33 in cancer cells but not in normal cells. Specifically, DHX33 knockdown caused the downregulation of HMGBs at the levels of both gene transcription and protein expression. Notably, in RAS driven lung tumorigenesis, nuclear HMGBs proteins can be induced via DHX33. When DHX33 was knocked out, HMGBs overexpression was debilitated. Mechanistically, DHX33 was found to bind to the promoters of HMGB family genes and regulated their transcription through demethylation on gene promoters. Our study reveals a novel mechanism for DHX33 to promote tumorigenesis and highlights its therapeutic value in human cancers.


Subject(s)
Neoplasms , Humans , Lung , Carcinogenesis , Cell Transformation, Neoplastic , HMGB Proteins , DEAD-box RNA Helicases/genetics
5.
Environ Toxicol ; 38(8): 2011-2021, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37256545

ABSTRACT

DEAH-box helicase 33 (DHX33) is a potent oncogenic agent on patients with hepatocellular carcinoma and colon cancer. Nevertheless, the prognostic significance of DHX33 in human pan-cancers remains unclear. Various bioinformatics tools have been used to clarify the oncogenic effects of DHX33 in pan-cancers. The Cancer Genome Atlas (TCGA) and Human Protein Atlas (HPA) were used to evaluate DHX33 at the mRNA and protein levels, respectively. The pan-cancer prognostic prediction value of DHX33 was evaluated using the Kaplan-Meier plotter. The association between DHX33 levels and clinicopathological features was then determined using the UALCAN database. In addition, gene set enrichment analysis and nomogram prediction models were constructed. The association between DHX33 levels and immune cell infiltration was then assessed using TISIDB. We determined that DHX33 is aberrantly overexpressed in pan-cancers and related to lung adenocarcinoma (LUAD) clinical stage (p < .001). Moreover, DHX33 overexpression was indicative of poor overall survival, disease-free survival, and progression-free interval in patients with LUAD (p < .05). Furthermore, DHX33 levels were associated with age, N stage, T stage, and pathologic stage in patients with LUAD (p < .001). Additionally, multivariate Cox analysis revealed that DHX33 was an independent risk factor for OS in patients with LUAD. A nomogram model between DHX33 levels and characteristic clinical parameters was developed. Additionally, DHX33 levels correlated with immunomodulators and chemokines. Finally, gene set enrichment analysis revealed that the amyloid fiber formation pathway and the WICH complex positively regulates RNA expression pathway, which was the most enriched in LUAD. Our data revealed oncogenic effects of DHX33 in various cancers. We suggest that DHX33 may serve as a biomarker for poor prognosis in LUAD.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Hepatocellular , Liver Neoplasms , Lung Neoplasms , Humans , Prognosis , DEAD-box RNA Helicases/genetics
6.
Cell Signal ; 101: 110526, 2023 01.
Article in English | MEDLINE | ID: mdl-36403931

ABSTRACT

DHX33 is a member of DEAD/H box protein family, and is involved in both RNA and DNA metabolism. It plays diverse roles in multiple cellular activities. DHX33 overexpression has been found to promote the development of many human cancers. However, the underlying mechanism to explain its high expression in cancer cells remains incompletely resolved. In this study, with both human cancer cell lines and normal fibroblasts, we found glycogen synthase kinase 3ß (GSK-3ß) regulates DHX33 protein stability. This is through its direct phosphorylation of DHX33 on T482, which triggers ubiquitination mediate protein degradation. We further identified one of the major ubiquitination sites of DHX33 to be on its N-terminal K94, a critical residue previously found to be important and highly conserved for ATP binding and helicase activity. Our study for the first time reveals an important upstream regulator, GSK-3ß, as a critical kinase to phosphorylate DHX33 at the post-translational level leading to its degradation. Moreover, cancer cells have frequent GSK3ß deactivation to disrupt this signaling cascade. Therefore, DHX33 is stabilized in many cancer cells as compared to normal cells. Our study unveils an important post-translational regulation of DHX33 in cells and further unveils a novel mechanism for DHX33 overexpression in cancer cells.


Subject(s)
DEAD-box RNA Helicases , Glycogen Synthase Kinase 3 beta , Proteolysis , Signal Transduction , Humans , DEAD-box RNA Helicases/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Phosphorylation , Ubiquitination
7.
Genes Genomics ; 43(8): 857-868, 2021 08.
Article in English | MEDLINE | ID: mdl-33843021

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common human cancers. Long non-coding RNAs (lncRNAs) play pivotal roles in progression of various cancers, including HCC. OBJECTIVE: We aimed to explore the exact role and underlying mechanism of lncRNA HOX transcript antisense intergenic RNA (HOTAIR) in HCC. METHODS: Quantitative real time polymerase chain reaction (qRT-PCR) was carried out to determine the levels of HOTAIR, DEAH-box helicase 33 (DHX33) and miR-526b-3p. Western blot assay was used to detect the protein level of DHX33. Besides, cell proliferation and apoptosis were assessed by methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay and flow cytometry analysis, respectively. Cell migration and invasion were detected by transwell assay. The interaction between miR-526b-3p and HOTAIR or DHX33 was predicted by starbase and confirmed by the dual-luciferase reporter assay. Murine xenograft model was established through injecting Huh7 cells transfected with sh-NC or sh-HOTAIR. RESULTS: The levels of HOTAIR and DHX33 were increased in HCC tissues and cells. Knockdown of either HOTAIR or DHX33 suppressed proliferation, migration and invasion but increased apoptosis in HCC cells. Moreover, DHX33 overexpression reversed the suppressive effect of HOTAIR knockdown on progression of HCC cells. Interestingly, miR-526b-3p could directly bind to HOTAIR, and DHX33 was a direct target of miR-526b-3p. Additionally, interference of HOTAIR restrained the tumor growth by upregulating miR-526b-3p and downregulating DHX33 in vivo. CONCLUSIONS: HOTAIR knockdown suppressed cell proliferation, migration and invasion, and promoted apoptosis via regulating miR-526b-3p/DHX33 axis in HCC cells, providing a potential avenue for treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , DEAD-box RNA Helicases/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Animals , Apoptosis/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Liver Neoplasms/pathology , Mice
8.
J Cell Physiol ; 236(2): 981-996, 2021 02.
Article in English | MEDLINE | ID: mdl-32617965

ABSTRACT

Cancer cells metabolize glucose through glycolysis to promote cell proliferation even with abundant oxygen. Multiple glycolysis genes are deregulated during cancer development. Despite intensive effort, the cause of their deregulation remains incompletely understood. Here in this study, we discovered that DHX33 plays a critical role in Warburg effect of cancer cells. DHX33 deficient cells have markedly reduced glycolysis activity. Through RNA-seq analysis, we found multiple critical genes involved in Warburg effect were downregulated after DHX33 deficiency. These genes include lactate dehydrogenase A (LDHA), pyruvate dehydrogenase kinase 1 (PDK1), pyruvate kinase muscle isoform 2 (PKM2), enolase 1 (ENO1), ENO2, hexokinase 1/2, among others. With LDHA, PDK1, and PKM2 as examples, we further revealed that DHX33 altered the epigenetic marks around the promoter of glycolytic genes. This is through DHX33 in complex with Gadd45a-a growth arrest and DNA damage protein. DHX33 is required for the loading of Gadd45a and DNA dioxygenase Tet1 at the promoter sites, which resulted in active DNA demethylation and enhanced histone H4 acetylation. We conclude that DHX33 changes local epigenetic marks in favor of the transcription of glycolysis genes to promote cancer cell proliferation. Our study highlights the significance of RNA helicase DHX33 in Warburg effect and cancer therapeutics.


Subject(s)
DEAD-box RNA Helicases/genetics , Glycolysis/genetics , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , DNA Damage/genetics , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Transcription, Genetic/genetics , Warburg Effect, Oncologic
9.
Cancer Sci ; 111(10): 3564-3575, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32767810

ABSTRACT

Ras has been found to be mutated in 30% of non-small cell lung cancers, and its mutation has been regarded as a causal factor underlying tumorigenesis. However, no successful medicine has been developed so far to inhibit Ras for lung cancer treatment. We have previously identified DHX33 as a Ras downstream effector, promoting cell cycle progression and cell growth. In this study, with the K-Ras (G12D);DHX33 (lox/lox) mouse model, we discovered that genetic ablation of DHX33 inhibited tumor development. We further found that ablation of DHX33 altered the expression of nearly 2000 genes which are critical in cancer development such as cell cycle, apoptosis, glycolysis, Wnt signaling, and cell migration. Our study for the first time demonstrates the pivotal role of the DHX33 in Ras-driven lung cancer development in vivo and highlights that pharmacological targeting DHX33 can be a feasible option in treating Ras-mutant lung cancers.


Subject(s)
Carcinogenesis/genetics , DEAD-box RNA Helicases/genetics , Lung Neoplasms/genetics , ras Proteins/genetics , Animals , Apoptosis/genetics , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Lung/pathology , Lung Neoplasms/pathology , Mice , Mice, Knockout , Wnt Signaling Pathway/genetics
10.
Mol Cell Biol ; 40(13)2020 06 15.
Article in English | MEDLINE | ID: mdl-32312884

ABSTRACT

RNA helicase DHX33 was found to regulate the transcription of multiple genes involved in cancer development. But the underlying molecular mechanism remains unclear. Here, we found DHX33 associated extensively with gene promoters at CG-rich region. Its deficiency reduced the loading of active RNA polymerase II at gene promoters. Furthermore, we observed a functional interaction between DHX33, AP-2ß, and DNA demethylation protein Gadd45a (growth arrest and DNA damage inductile protein 45a) at specific gene promoters. DHX33 is required to recruit GADD45a, thereby causing local DNA demethylation through further recruiting ten-eleven-translocation (Tet) methylcytosine dioxygenase enzyme, as manifested by reduced 5-hydroxymethyl cytosine levels for a subset of genes after DHX33 deficiency. This process might involve R-loop formation in GC skew as a guidance signal at promoter sites. Our report provides for the first time, to our knowledge, original evidence that DHX33 alters epigenetic marks and regulates specific gene transcription through interaction with Gadd45a.


Subject(s)
Cell Cycle Proteins/metabolism , DEAD-box RNA Helicases/metabolism , DNA Demethylation , Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Promoter Regions, Genetic , Protein Interaction Maps , Transcriptional Activation
11.
Gene ; 735: 144402, 2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32004669

ABSTRACT

Colon cancer is one of the most common malignancies in the world; there is no effective therapeutic treatment after surgery. Our previous studies indicate that RNA helicase DHX33 plays a critical role in cell proliferation and cell growth. Here in this study, DHX33 is found to be highly expressed in colon cancer tissues and colon cancer cell lines. Knockdown of DHX33 significantly decreased cell proliferation and triggered apoptosis. Mechanistically, DHX33 was found to transcriptionally control multiple critical genes involved in cell cycle, apoptosis and migration. DHX33 deficiency caused decreased tumor growth for colon cancer cells in a xenograft model in vivo. With Wnt/ß-cateninactivator and inhibitors, we further discovered that Wnt/ß-catenin pathway regulates DHX33 transcriptionally. This study for the first time demonstratesthe important role of DHX33 in colon cancer development and reveals the underlying molecular mechanism. We also provide the initial evidence for the relationship between DHX33 and Wnt/ß-catenin signaling pathway in colon cancer development.


Subject(s)
Carcinoma/genetics , Colonic Neoplasms/genetics , DEAD-box RNA Helicases/genetics , Wnt Signaling Pathway , Animals , Apoptosis , Carcinoma/metabolism , Carcinoma/pathology , Cell Proliferation , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DEAD-box RNA Helicases/metabolism , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude
12.
Front Oncol ; 9: 979, 2019.
Article in English | MEDLINE | ID: mdl-31632911

ABSTRACT

Transcriptional intermediary factor 1 γ (TIF1γ), also known as TRIM33, RFG7, PTC7, or Ectodermin, is an E3 ubiquitin-ligase family member with a ring-box-coiled-coil region. It can regulate TGF-ß/Smad signaling in two different ways in different cellular contexts. On one hand, TIF1γ can monoubiquitinate Smad4 to inhibit the formation of Smad2/3/4 nuclear complexes. On the other hand, TIF1γ can function as a cofactor of phosphorylated (p)-Smad2/3, competing with Smad4 to inhibit the formation of the Smad2/3/4 complex. In addition, TIF1γ has been reported to play a role in transcription elongation, cellular differentiation, embryonic development, and mitosis. As transforming growth factor-ß (TGF-ß) superfamily signaling plays an important role in the occurrence and development of cancer, and TIF1γ was reported to be involved in the regulation of TGF-ß superfamily signaling, studies on TIF1γ during the last decade have focused on its role in the development of cancer. However, TIF1γ can function either as a tumor suppressor or promoter in different cellular contexts, yet there are few reviews focusing on the roles of TIF1γ in cancer. Hence, in this paper we systematically review and discuss the roles of TIF1γ in cancer. Firstly, we review the biological features, the regulatory mechanisms and the related signaling pathways of TIF1γ. Next, we illustrate the roles of TIF1γ in different tumors. We then provide a tentative hypothesis that explains the dual roles of TIF1 γ in cancer. Finally, we provide our viewpoint regarding the future developments of cancer research focusing on TIF1γ, especially in relation to the effects of TIF1γ on tumoral immunity.

13.
Mol Cell Biol ; 39(17)2019 09 01.
Article in English | MEDLINE | ID: mdl-31182639

ABSTRACT

The RNA helicase DHX33 has been found to be overexpressed in human cancers, where it promotes cancer development. Previous reports have shown that DHX33 deficiency caused cancer cell apoptosis, but the underlying mechanism remains unknown. In this study, we discovered that DHX33 regulates Bcl-2 family protein expression. In multiple human cancer cell lines, DHX33 was found to stimulate the transcription of Bcl-2 Mechanistically, we found that DHX33 interacts with the AP-2ß transcription factor and acts as a coactivator to stimulate Bcl-2 gene transcription. DHX33 deficiency abolished the loading of AP-2ß onto the promoter of Bcl-2 and thereby reduced the recruitment of active RNA polymerase II during transcription initiation. Acute knockdown of DHX33 in multiple human cancer cells caused decreased Bcl-2 protein level, which ultimately triggered mitochondrion-mediated cellular apoptosis. In addition, we found that normal human lung and mammary epithelial cells were less sensitive to acute DHX33 knockdown, implying that cancer cells are uniquely responsive to DHX33 reduction. These data support the notion that disruption of DHX33 function could be an important application for cancer therapy.


Subject(s)
Breast Neoplasms/pathology , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Transcription Factor AP-2/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Mitochondria/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Polymerase II/metabolism
14.
J Cell Physiol ; 234(9): 15308-15319, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30684270

ABSTRACT

DEAH box protein DHX33 has been found to be necessary for cell proliferation and early development of multicellular organisms. It plays diverse roles in regulating gene transcription, ribosome RNA synthesis, and protein translation. Dysregulation of DHX33 has been observed in various human cancers. In this study, we identified a short DHX33 variant in cells. The short DHX33 (hereafter referred to as DHX33-2) has only 534 amino acids, which completely matches the C-terminal helicase domain of full-length DHX33 (DHX33-1). Different from DHX33-1, which mainly localizes to the nucleus, DHX33-2 preferentially localizes to the cytoplasm. Through protein immunoprecipitation and RNA- immunoprecipitation analysis, we found that DHX33-2 interacts with DDX3, eIF3, hnRNPs, poly (A) binding protein, and a subset of mRNAs. Further RNA sequencing analysis showed that DHX33 binds to a subset of mRNAs important in cell proliferation. DHX33-2 stimulates the translation for specific mRNAs. Our study for the first time demonstrates the function of a short DHX33 variant in protein translation.

15.
Cell Signal ; 54: 170-178, 2019 02.
Article in English | MEDLINE | ID: mdl-30552990

ABSTRACT

Human Glioblastoma is one deadly disease; the median survival time is reported to be 13.9 months after treatment. In the present study, we discovered that DHX33 is highly expressed in 84% of all Glioblastoma multiforme (GBM). Knockdown of DHX33 led to significant reduced proliferation and migration in glioblastoma cells in vitro and in vivo. Mechanistically, DHX33 regulated a set of critical genes involved in cell cycle and cell migration to promote glioblastoma development. Additionally, DHX33 was found to be induced by inhibitors of PI3K and mTOR whose activation has been detected in 50% of glioblastoma. Overexpression of wild type DHX33 protein, but not the helicase dead mutant, confers resistance to mTOR inhibitors in glioblastoma cells. DHX33 probably functions as a critical regulator to promote GBM development. Our results highlight its therapeutic potential in treating GBM.


Subject(s)
Brain Neoplasms/metabolism , DEAD-box RNA Helicases/physiology , Glioblastoma/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement , Cell Proliferation , Glioblastoma/drug therapy , HEK293 Cells , Humans , Mice, Nude
16.
Biochem Biophys Res Commun ; 502(4): 501-507, 2018 08 25.
Article in English | MEDLINE | ID: mdl-29864424

ABSTRACT

DHX33 has been shown to play key roles in promoting cell proliferation. We have previously found that DHX33 protein is a doublet. In this report, we discovered that DHX33 doublet is due to alternative translation initiation by two in-frame initiation codons. This is supported by studies from both cell lines and mouse models. DHX33 translation initiation from either AUG codon happens at equal efficiency. Short DHX33 protein has similar cellular location and functions with full-length DHX33. Our results suggest that leaky scanning normally occur in DHX33 mRNA translation, which may serve as a safeguard mechanism to ensure optimal DHX33 translation efficiency. This is the first report of DEAD/DEAH box proteins that can be regulated by alternative translation initiation.


Subject(s)
Codon, Initiator , DEAD-box RNA Helicases/biosynthesis , DEAD-box RNA Helicases/genetics , Peptide Chain Initiation, Translational , Animals , Cells, Cultured , DEAD-box RNA Helicases/chemistry , Humans , Isoenzymes/biosynthesis , Isoenzymes/chemistry , Isoenzymes/genetics , Mice , Mice, Knockout , NIH 3T3 Cells , RNA, Guide, Kinetoplastida/genetics , Reading Frames
17.
Mol Oncol ; 10(10): 1532-1541, 2016 12.
Article in English | MEDLINE | ID: mdl-27693040

ABSTRACT

Deregulation of microRNAs contributes to the aberrant growth of hepatocellular carcinoma (HCC). Here, we showed that miR-634 expression was frequently decreased in HCC. Low miR-634 expression was significantly associated with larger tumor size, poorer tumor differentiation, advanced TNM stage, vascular invasion, absence of tumor capsule and unfavorable overall survival. Overexpression of miR-634 markedly attenuated cell viability, colony formation, tumor growth and metastasis, whereas miR-634 inhibition resulted in the opposite phenotypes. Furthermore, re-introduction of miR-634 induced cell apoptosis in vitro and in vivo. Mechanistically, miR-634 inhibited the expression of Rab1A and DHX33 via directly binding to the 3'-UTR of both genes. In clinical samples, the expression of Rab1A or DHX33 was reversely correlated with miR-634. Re-expression of Rab1A or DHX33 abrogated the miR-634-mediated inhibition of cell proliferation and migration. Collectively, our data suggest a tumor suppressor role of miR-634 in HCC. The newly identified miR-634/Rab1A or miR-634/DHX33 axis serves as a potential therapeutic target for the clinical management.


Subject(s)
Carcinoma, Hepatocellular/genetics , DEAD-box RNA Helicases/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Liver/pathology , MicroRNAs/genetics , rab1 GTP-Binding Proteins/genetics , 3' Untranslated Regions , Animals , Apoptosis , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Genes, Tumor Suppressor , Humans , Liver/metabolism , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology
18.
Biochem Biophys Res Commun ; 473(4): 1163-1169, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27073163

ABSTRACT

DEAH-box helicase 33 (DHX33) has been implicated in ribosome biogenesis, mRNA translation and inflammation. However, the role of DHX33 in human cancer is rarely studied. Here, we showed that DHX33 expression was significantly increased in hepatocellular carcinoma (HCC), compared with the adjacent nontumorous tissues. In a cohort of 520 patients, DHX33 expression in HCC was closely associated with tumor size (P = 0.007), serum AFP level (P = 0.011), and tumor capsule (P = 0.030). Kaplan-Meier analysis indicated high DHX33 expression was correlated with worse overall and disease-free survival, and higher recurrence rate. The prognostic value of DHX33 was further confirmed by stratified survival analysis. Multivariate analysis revealed DHX33 as an independent prognostic factor for poor overall survival (Hazard Ratio = 1.772, 95% confident interval: 1.451-2.165, P < 0.0001). Our data therefore suggest DHX33 is overexpressed in HCC and serves as a promising prognostic biomarker for this deadly disease.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/mortality , DEAD-box RNA Helicases/genetics , Liver Neoplasms/diagnosis , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , China/epidemiology , DEAD-box RNA Helicases/metabolism , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Prevalence , Reproducibility of Results , Risk Factors , Sensitivity and Specificity , Survival Rate , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL