Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 184
Filter
1.
Viruses ; 16(3)2024 03 05.
Article in English | MEDLINE | ID: mdl-38543768

ABSTRACT

LTR-retrotransposons are transposable elements characterized by the presence of long terminal repeats (LTRs) directly flanking an internal coding region. They share genome organization and replication strategies with retroviruses. Steamer-like Element-1 (MchSLE-1) is an LTR-retrotransposon identified in the genome of the Chilean blue mussel Mytilus chilensis. MchSLE-1 is transcribed; however, whether its RNA is also translated and the mechanism underlying such translation remain to be elucidated. Here, we characterize the MchSLE-1 translation mechanism. We found that the MchSLE-1 5' and 3'LTRs command transcription of sense and antisense RNAs, respectively. Using luciferase reporters commanded by the untranslated regions (UTRs) of MchSLE-1, we found that in vitro 5'UTR sense is unable to initiate translation, whereas the antisense 5'UTR initiates translation even when the eIF4E-eIF4G interaction was disrupted, suggesting the presence of an internal ribosomal entry site (IRES). The antisense 5'UTR IRES activity was tested using bicistronic reporters. The antisense 5'UTR has IRES activity only when the mRNA is transcribed in the nucleus, suggesting that nuclear RNA-binding proteins are required to modulate its activity. Indeed, heterogeneous nuclear ribonucleoprotein K (hnRNPK) was identified as an IRES trans-acting factor (ITAF) of the MchSLE-1 IRES. To our knowledge, this is the first report describing an IRES in an antisense mRNA derived from a mussel LTR-retrotransposon.


Subject(s)
Internal Ribosome Entry Sites , Mytilus , Animals , RNA, Messenger/genetics , RNA, Messenger/metabolism , Internal Ribosome Entry Sites/genetics , Retroelements/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , 5' Untranslated Regions , Mytilus/genetics , Mytilus/metabolism , Protein Biosynthesis
2.
Mol Ther ; 32(5): 1461-1478, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38414246

ABSTRACT

Osteoarthritis (OA) is an age-related or post-traumatic degenerative whole joint disease characterized by the rupture of articular cartilage homeostasis, the regulatory mechanisms of which remain elusive. This study identifies the essential role of heterogeneous nuclear ribonucleoprotein K (hnRNPK) in maintaining articular cartilage homeostasis. Hnrnpk expression is markedly downregulated in human and mice OA cartilage. The deletion of Hnrnpk effectively accelerates the development of post-traumatic and age-dependent OA in mice. Mechanistically, the KH1 and KH2 domain of Hnrnpk bind and degrade the mRNA of WWC1. Hnrnpk deletion increases WWC1 expression, which in turn leads to the activation of Hippo signaling and ultimately aggravates OA. In particular, intra-articular injection of LPA and adeno-associated virus serotype 5 expressing WWC1 RNA interference ameliorates cartilage degeneration induced by Hnrnpk deletion, and intra-articular injection of adeno-associated virus serotype 5 expressing Hnrnpk protects against OA. Collectively, this study reveals the critical roles of Hnrnpk in inhibiting OA development through WWC1-dependent downregulation of Hippo signaling in chondrocytes and defines a potential target for the prevention and treatment of OA.


Subject(s)
Cartilage, Articular , Chondrocytes , Heterogeneous-Nuclear Ribonucleoprotein K , Hippo Signaling Pathway , Osteoarthritis , Protein Serine-Threonine Kinases , Signal Transduction , Animals , Osteoarthritis/metabolism , Osteoarthritis/genetics , Osteoarthritis/etiology , Osteoarthritis/pathology , Osteoarthritis/therapy , Mice , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Disease Models, Animal , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Dependovirus/genetics , Gene Expression Regulation , Male
3.
Nucleic Acids Res ; 52(5): 2625-2647, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38165048

ABSTRACT

Translation initiation of the human immunodeficiency virus-type 1 (HIV-1) genomic mRNA (vRNA) is cap-dependent or mediated by an internal ribosome entry site (IRES). The HIV-1 IRES requires IRES-transacting factors (ITAFs) for function. In this study, we evaluated the role of the heterogeneous nuclear ribonucleoprotein K (hnRNPK) as a potential ITAF for the HIV-1 IRES. In HIV-1-expressing cells, the depletion of hnRNPK reduced HIV-1 vRNA translation. Furthermore, both the depletion and overexpression of hnRNPK modulated HIV-1 IRES activity. Phosphorylations and protein arginine methyltransferase 1 (PRMT1)-induced asymmetrical dimethylation (aDMA) of hnRNPK strongly impacted the protein's ability to promote the activity of the HIV-1 IRES. We also show that hnRNPK acts as an ITAF for the human T cell lymphotropic virus-type 1 (HTLV-1) IRES, present in the 5'UTR of the viral sense mRNA, but not for the IRES present in the antisense spliced transcript encoding the HTLV-1 basic leucine zipper protein (sHBZ). This study provides evidence for a novel role of the host hnRNPK as an ITAF that stimulates IRES-mediated translation initiation for the retroviruses HIV-1 and HTLV-1.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein K , Retroviridae , Humans , 5' Untranslated Regions , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Internal Ribosome Entry Sites/genetics , Phosphorylation , Protein Biosynthesis , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Retroviridae/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Biochim Biophys Acta Gene Regul Mech ; 1867(1): 195004, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38008244

ABSTRACT

Deletions on the long arm of chromosome 9 (del(9q)) are recurrent abnormalities in about 2 % of acute myeloid leukemia cases, which usually involve HNRNPK and are frequently associated with other known aberrations. Based on an Hnrnpk haploinsufficient mouse model, a recent study demonstrated a function of hnRNP K in pathogenesis of myeloid malignancies via the regulation of cellular proliferation and myeloid differentiation programs. Here, we provide evidence that reduced hnRNP K expression results in the dysregulated expression of C/EBPα and additional transcription factors. CyTOF analysis revealed monocytic skewing with increased levels of mature myeloid cells. To explore the role of hnRNP K during normal and pathological myeloid differentiation in humans, we characterized hnRNP K-interacting RNAs in human AML cell lines. Notably, RNA-sequencing revealed several mRNAs encoding key transcription factors involved in the regulation of myeloid differentiation as targets of hnRNP K. We showed that specific sequence motifs confer the interaction of SPI1 and CEBPA 5' and 3'UTRs with hnRNP K. The siRNA mediated reduction of hnRNP K in human AML cells resulted in an increase of PU.1 and C/EBPα that is most pronounced for the p30 isoform. The combinatorial treatment with the inducer of myeloid differentiation valproic acid resulted in increased C/EBPα expression and myeloid differentiation. Together, our results indicate that hnRNP K post-transcriptionally regulates the expression of myeloid master transcription factors. These novel findings can inaugurate novel options for targeted treatment of AML del(9q) by modulation of hnRNP K function.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha , Leukemia, Myeloid, Acute , Animals , Mice , Humans , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Transcription Factors/metabolism , Heterogeneous-Nuclear Ribonucleoproteins , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism
5.
Neurochem Int ; 169: 105589, 2023 10.
Article in English | MEDLINE | ID: mdl-37543308

ABSTRACT

As a ubiquitous RNA-binding protein, heterogeneous nuclear ribonucleoprotein K (hnRNPK) interacts with numerous nucleic acids and proteins and is involved in various cellular functions. Available literature indicates that it can regulate dendritic spine density through the extracellular signal-regulating kinase (ERK) - brain-derived neurotrophic factor (BDNF) pathway, which is crucial to retain the synaptic plasticity in patients with major depressive disorder (MDD) and mouse depression models. However, ERK upstream regulatory kinase has not been fully elucidated. Furthermore, it remains unexplored whether hnRNPK may impact the depressive condition via the ERK pathway. The present study addressed this issue by integrating approaches of genetics, molecular biology, behavioral testing. We found that hnRNPK in the brain was mainly distributed in the hippocampal neurons; that it was significantly downregulated in mice that displayed stress-induced depression-like behaviors; and that the level of hnRNPK markedly decreased in MDD patients from the GEO database. Further in vivo and in vitro analyses revealed that the changes in the expressions of BDNF and PSD95 and in the phosphorylation of ERK (Thr202/Tyr204) paralleled the variation of hnRNPK levels in the ventral hippocampal neurons in mice with depression-like behaviors. Finally, esketamine treatment significantly increased the level of hnRNPK in mice. These findings evidence that hnRNPK involved in the pathogenesis of depression via the ERK-BDNF pathway, pinpointing hnRNPK as a potential therapeutic target in treating MDD patients.


Subject(s)
Depression , Depressive Disorder, Major , Animals , Mice , Brain-Derived Neurotrophic Factor/metabolism , Depression/drug therapy , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Hippocampus/metabolism , Signal Transduction , MAP Kinase Signaling System
6.
BMC Mol Cell Biol ; 24(1): 26, 2023 Aug 17.
Article in English | MEDLINE | ID: mdl-37592256

ABSTRACT

BACKGROUND: Heterogeneous nuclear ribonucleoprotein K (HNRNPK) regulates pre-mRNA processing and long non-coding RNA localization in the nucleus. It was previously shown that shuttling of HNRNPK to the cytoplasm promotes cell proliferation and cancer metastasis. However, the mechanism of HNRNPK cytoplasmic localization, its cytoplasmic RNA ligands, and impact on post-transcriptional gene regulation remain uncharacterized. RESULTS: Here we show that the intermediate filament protein Keratin 19 (K19) directly interacts with HNRNPK and sequesters it in the cytoplasm. Correspondingly, in K19 knockout breast cancer cells, HNRNPK does not localize in the cytoplasm, resulting in reduced cell proliferation. We comprehensively mapped HNRNPK binding sites on mRNAs and showed that, in the cytoplasm, K19-mediated HNRNPK-retention increases the abundance of target mRNAs bound to the 3' untranslated region (3'UTR) at the expected cytidine-rich (C-rich) sequence elements. Furthermore, these mRNAs protected by HNRNPK in the cytoplasm are typically involved in cancer progression and include the p53 signaling pathway that is dysregulated upon HNRNPK knockdown (HNRNPK KD) or K19 knockout (KRT19 KO). CONCLUSIONS: This study identifies how a cytoskeletal protein can directly regulate gene expression by controlling the subcellular localization of RNA-binding proteins to support pathways involved in cancer progression.


Subject(s)
Triple Negative Breast Neoplasms , Humans , RNA, Messenger/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Keratin-19 , Cytoplasm , 3' Untranslated Regions/genetics
7.
J Mol Biol ; 435(10): 168050, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36933825

ABSTRACT

In the present investigation, we have identified the functional significance of the highly conserved miR-140 binding site on the Hepatitis E Virus (HEV) genome. Multiple sequence alignment of the viral genome sequences along with RNA folding prediction indicated that the putative miR-140 binding site has significant conservation for sequence and secondary RNA structure among HEV genotypes. Site-directed mutagenesis and reporter assays indicated that an intact sequence of the miR-140 binding site is essential for HEV translation. Provision of mutant miR-140 oligos carrying same mutation as on mutant HEV successfully rescued mutant HEV replication. In vitro cell-based assays with modified oligos proved that host factor-miR-140 is a critical requirement for HEV replication. Biotinylated RNA pulldown and RNA immunoprecipitation assays proved that the predicted secondary RNA structure of the miR-140 binding site allows the recruitment of hnRNP K, which is a key protein of the HEV replication complex. We predicted the model from the obtained results that the miR-140 binding site can serve as a platform for recruitment of hnRNP K and other proteins of HEV replication complex only in the presence of miR-140.


Subject(s)
Hepatitis E virus , Heterogeneous-Nuclear Ribonucleoprotein K , MicroRNAs , Hepatitis E virus/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Viral/metabolism , Virus Replication/genetics , Humans
8.
J Mol Biol ; 435(6): 167993, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36736887

ABSTRACT

Heterogeneous nuclear ribonucleoprotein K (HNRNPK, hnRNP K), a multifunctional RNA/DNA binding protein, mainly regulates transcription, translation and RNA splicing, and then plays oncogenic roles in many cancers. However, the related mechanisms remain largely unknown. Here, we found that HNRNPK can partially epigenetically regulate cancer cell proliferation via increasing transcription and exon 4-inclusion of SPIN1, an important oncogenic histone code reader. This exon 4 skipping event of SPIN1 generates a long non-coding RNA, followed by the downregulation of SPIN1 protein. SPIN1 is one of the most significantly co-expressed genes of HNRNPK in thirteen TCGA cancers. Our further studies revealed HNRNPK knockdown significantly inhibited cell growth and cell cycle progression in oral squamous cell carcinoma (OSCC) cells and promoted cell apoptosis. Overexpression of SPIN1 was able to partially rescue the growth inhibition triggered by HNRNPK knockdown. Moreover, CCND1 (Cyclin D1), a key cell cycle regulator and oncogene, epigenetically up-regulated by SPIN1, was also positively regulated by HNRNPK. In addition, we discovered that HNRNPK promoted SPIN1 exon 4 inclusion by interacting with an intronic splicing enhancer in intron 4. Collectively, our study suggests a novel epigenetic regulatory pathway of HNRNPK in OSCC, mediated by controlling the transcription activity and alternative splicing of SPIN1 gene.


Subject(s)
Carcinoma, Squamous Cell , Cell Cycle Proteins , Epigenesis, Genetic , Heterogeneous-Nuclear Ribonucleoprotein K , Mouth Neoplasms , Humans , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Cycle/genetics , Cell Proliferation/genetics , Exons/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Histone Code/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Cell Cycle Proteins/genetics
9.
Elife ; 122023 02 03.
Article in English | MEDLINE | ID: mdl-36735291

ABSTRACT

Peroxiredoxin 5 (Prdx5) is involved in pathophysiological regulation via the stress-induced cellular response. However, its function in the bone remains largely unknown. Here, we show that Prdx5 is involved in osteoclast and osteoblast differentiation, resulting in osteoporotic phenotypes in Prdx5 knockout (Prdx5Ko) male mice. To investigate the function of Prdx5 in the bone, osteoblasts were analyzed through immunoprecipitation (IP) and liquid chromatography combined with tandem mass spectrometry (LC-MS/MS) methods, while osteoclasts were analyzed through RNA-sequencing. Heterogeneous nuclear ribonucleoprotein K (hnRNPK) was identified as a potential binding partner of Prdx5 during osteoblast differentiation in vitro. Prdx5 acts as a negative regulator of hnRNPK-mediated osteocalcin (Bglap) expression. In addition, transcriptomic analysis revealed that in vitro differentiated osteoclasts from the bone marrow-derived macrophages of Prdx5Ko mice showed enhanced expression of several osteoclast-related genes. These findings indicate that Prdx5 might contribute to the maintenance of bone homeostasis by regulating osteoblast differentiation. This study proposes a new function of Prdx5 in bone remodeling that may be used in developing therapeutic strategies for bone diseases.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein K , Osteogenesis , Animals , Male , Mice , Bone Regeneration , Cell Differentiation , Chromatography, Liquid , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Tandem Mass Spectrometry
10.
Virology ; 581: 15-25, 2023 04.
Article in English | MEDLINE | ID: mdl-36842269

ABSTRACT

HnRNP K is a well-known member of HnRNP family proteins that has been implicated in the regulation of protein expression. Currently, the impact of HnRNP K on the reproduction cycle of a broad range of virus were reported, while the precise function for PRRSV was lacking. In this study, we determined that both PRRSV infection and ectopic expression of N protein induced an enrichment of HnRNP K in the cytoplasm. Using RNA pulldown and RNA immunoprecipitation, we described the interactions between the KH2 domain of HnRNP K and cytosine-rich sequences (CRS) in PRRSV genomic RNA corresponding to Nsp7α coding region. Meanwhile, overexpression of HnRNP K inhibited viral gene expression and PRRSV replication, while silencing of HnRNP K resulted in an increased in virus yield. Taken together, this study assists in the understanding of PRRSV-host interactions, and the development of vaccines based on viral genome engineering.


Subject(s)
Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Animals , Swine , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/metabolism , Cell Line , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication/physiology , RNA , Gene Expression
11.
Commun Biol ; 6(1): 82, 2023 01 21.
Article in English | MEDLINE | ID: mdl-36681772

ABSTRACT

RNA-binding proteins (RBPs) are key players of gene expression and perturbations of RBP-RNA regulatory network have been observed in various cancer types. Here, we propose a computational method, RBPreg, to identify the RBP regulators by integration of single cell RNA-Seq (N = 233,591) and RBP binding data. Pan-cancer analyses suggest that RBP regulators exhibit cancer and cell specificity and perturbations of RBP regulatory network are involved in cancer hallmark-related functions. We prioritize an oncogenic RBP-HNRNPK, which is highly expressed in tumors and associated with poor prognosis of patients. Functional assays performed in cancer cells reveal that HNRNPK promotes cancer cell proliferation, migration, and invasion in vitro and in vivo. Mechanistic investigations further demonstrate that HNRNPK promotes tumorigenesis and progression by directly binding to MYC and perturbed the MYC targets pathway in lung cancer. Our results provide a valuable resource for characterizing RBP regulatory networks in cancer, yielding potential biomarkers for precision medicine.


Subject(s)
Lung Neoplasms , RNA , Humans , RNA/genetics , Carcinogenesis , Cell Transformation, Neoplastic , Heterogeneous-Nuclear Ribonucleoprotein K/genetics
12.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 40(2): 226-229, 2023 Feb 10.
Article in Chinese | MEDLINE | ID: mdl-36709946

ABSTRACT

OBJECTIVE: To explore the clinical phenotype and genetic basis of a neonate with Au-Kline syndrome (AKS). METHODS: Clinical data and result of genetic testing of a neonate with AKS who was admitted to the Affiliated Provincial Children's Hospital of Anhui Medical University in January 2021 were retrospectively analyzed. Relevant literature was searched from the Wanfang Data Knowledge Service Platform, China National Knowledge Infrastructure and PubMed databases using key words "Au Kline syndrome", "Au-Kline syndrome", "HNRNPK" and "AKS". The research period was set as from January 1, 2000 to December 31, 2020. RESULTS: The male newborn has manifested feeding difficulties, hypotonia, absence of the upper jaw to the uvula and facial dysmorphism. Trio-whole exome sequencing revealed that he has harbored a frameshift c.478dupA (p.Ile160AsnfsTer7) variant of the HNRNPK gene, which was varified by Sanger sequencing to have a de novo origin. The variant has not been included in the databases. Based on the guidelines from the American College of Medical Genetics and Genomics, the variant was rated as pathogenic (PVS1+PS2+PM2_Supporting). Literature retrieval has identified 14 children with AKS and de novo mutations of the HNRNPK gene. Their clinical manifestations have included growth and motor retardation, various degree of mental retardation, facial dysmorphism and a high frequency of congenital heart malformations. CONCLUSION: The AKS in this child may be attributed to the c478dupA frameshifting variant of the HNRNPK gene. Diagnosis of AKS should be suspected for children with mental retardation and multiple congenital malformation syndromes including Kabuki syndrome.


Subject(s)
Abnormalities, Multiple , Intellectual Disability , Humans , Male , Abnormalities, Multiple/genetics , Genetic Testing , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Intellectual Disability/genetics , Mutation , Retrospective Studies , Infant, Newborn
13.
J Cancer Res Clin Oncol ; 149(9): 6225-6237, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36700980

ABSTRACT

PURPOSE: This study aims to explore the expression of hnRNP K in cervical carcinogenesis and to investigate the regulatory role of hnRNP K on HPV16 oncogene expression as well as biological changes in cervical cancer cells. METHODS: In total 1042 subjects, including 573 with the normal cervix and 469 with different grades of cervical lesions were enrolled in this study to explore the association between hnRNP K and HPV16 oncogene expression in cervical carcinogenesis. Additionally, the Gene Omnibus (GEO) database was used to analyze hnRNP K mRNA expression in cervical cancerization. Meanwhile, the effects of hnRNP K on cell biological functions and HPV16 oncogene expression were investigated in Siha cells. Moreover, Function analyses were conducted using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) databases after ChIP-seq. RESULTS: hnRNP K was highly expressed in cervical cancer and precancerous lesions, and positively correlated with HPV16 E6, but negatively correlated with HPV16 E2 and HPV16 E2/E6 ratio. hnRNP K induced cell proliferation, inhibited apoptosis and caused cell cycle arrest in the S phase, and particularly increased HPV16 E6 protein expression. CONCLUSION: This study revealed that hnRNP K overexpression has important warning significance for the malignant transformation of cervical lesions, and could be used as a potential therapeutic target for inhibiting the carcinogenicity of HPV16 and prevention of cervical carcinogenesis.


Subject(s)
Oncogene Proteins, Viral , Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Cervix Uteri/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Human papillomavirus 16/genetics , Human papillomavirus 16/metabolism , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Oncogenes/genetics , Carcinogenesis/genetics , Papillomavirus Infections/genetics
14.
Exp Biol Med (Maywood) ; 248(17): 1479-1491, 2023 09.
Article in English | MEDLINE | ID: mdl-35866661

ABSTRACT

HnRNP K is a heterogeneous nuclear ribonucleoprotein and has been identified as an oncogene in most solid tumors via regulating gene expression or alternative splicing of genes by binding both DNA and pre-mRNA. However, how hnRNP K affects tumorigenesis and regulates the gene expression in cervical cancer (CESC) remains to be elucidated. In these data, higher expression of hnRNP K was observed in CESC and was negatively correlated with the patient survival time. We then overexpressed hnRNP K (hnRNP K-OE) and found that its overexpression promoted cell proliferation in HeLa cells (P = 0.0052). Next, global transcriptome sequencing (RNA-seq) experiments were conducted to explore gene expression and alternative splicing profiles regulated by hnRNP K. It is shown that upregulated genes by hnRNP K-OE were associated with inflammatory response and an apoptotic process of neuron cells, which involves in cancer. In addition, the alternative splicing of those genes regulated by hnRNP K-OE was associated with transcriptional regulation. Analysis of the binding features of dysregulated transcription factors (TFs) in the promoter region of the inflammatory response genes regulated by hnRNP K revealed that hnRNP K may modulate the expression level of genes related to inflammatory response by influencing the alternative splicing of TFs. Among these hnRNP K-TFs-inflammatory gene regulatory networks, quantitative reverse transcription polymerase chain reaction (RT-qPCR) experiments and gene silencing were conducted to verify the hnRNP K-IRF1-CCL5 axis. In conclusion, the hnRNP K-TFs-inflammatory gene regulatory axis provides a novel molecular mechanism for hnRNP K in promoting CESC and offers a new therapeutic target.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein K , Transcription Factors , Humans , Alternative Splicing/genetics , Gene Expression , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Transcription Factors/metabolism
15.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-970910

ABSTRACT

OBJECTIVE@#To explore the clinical phenotype and genetic basis of a neonate with Au-Kline syndrome (AKS).@*METHODS@#Clinical data and result of genetic testing of a neonate with AKS who was admitted to the Affiliated Provincial Children's Hospital of Anhui Medical University in January 2021 were retrospectively analyzed. Relevant literature was searched from the Wanfang Data Knowledge Service Platform, China National Knowledge Infrastructure and PubMed databases using key words "Au Kline syndrome", "Au-Kline syndrome", "HNRNPK" and "AKS". The research period was set as from January 1, 2000 to December 31, 2020.@*RESULTS@#The male newborn has manifested feeding difficulties, hypotonia, absence of the upper jaw to the uvula and facial dysmorphism. Trio-whole exome sequencing revealed that he has harbored a frameshift c.478dupA (p.Ile160AsnfsTer7) variant of the HNRNPK gene, which was varified by Sanger sequencing to have a de novo origin. The variant has not been included in the databases. Based on the guidelines from the American College of Medical Genetics and Genomics, the variant was rated as pathogenic (PVS1+PS2+PM2_Supporting). Literature retrieval has identified 14 children with AKS and de novo mutations of the HNRNPK gene. Their clinical manifestations have included growth and motor retardation, various degree of mental retardation, facial dysmorphism and a high frequency of congenital heart malformations.@*CONCLUSION@#The AKS in this child may be attributed to the c478dupA frameshifting variant of the HNRNPK gene. Diagnosis of AKS should be suspected for children with mental retardation and multiple congenital malformation syndromes including Kabuki syndrome.


Subject(s)
Humans , Male , Infant, Newborn , Abnormalities, Multiple/genetics , Genetic Testing , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Intellectual Disability/genetics , Mutation , Retrospective Studies
16.
Cell Mol Biol (Noisy-le-grand) ; 69(14): 62-68, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38279484

ABSTRACT

The research aimed to the influences of the interaction between Y-box-binding protein 1 (YBX1) and heterogeneous nuclear ribonucleoprotein K (HNRNPK) on cell division cycle protein 25 phosphatase A (CDC25a) signal pathway and the regulatory mechanism of lung cancer (LC) metastasis. A total of 34 patients diagnosed with LC pathologically were selected as the research objects, and the expression levels of YBX1, HNRNP and CDC25a in LC non-metastasis tissues and LC metastasis tissues were detected by immunohistochemistry and Western blot (WB). High-expression stable cell lines including YBX1/A549 and HNRNPK /A549 were established in the LC A549 cell strain. The expression levels of YBX1 and HNRNP in YBX1/A549 and HNRNPK /A549 were tested by RT-PCR and WB. Besides, the number of migratory cells YBX1/A549 and HNRNPK /A549 was detected by cell migration experiment, and the influences of the interaction between YBX1 and HNRNP on the expression level of CDC25a were analyzed by co-immunoprecipitation (co-IP). The results showed that the expression level of YBX1 protein in LC metastasis tissues was higher than that in LC non-metastasis tissues (P<0.001). The expression level of HNRNPK protein in LC metastasis tissues was higher than that in LC non-metastasis tissues (P<0.01). The expression level of CDC25a protein in LC metastasis tissues was higher than that in LC non-metastasis tissues (P<0.05). Compared with the Control Group of A549 cell strain and transfected blank plasmid, mRNA levels and relative protein expression levels of YBX1 and HNRNPK in YBX1/A549 and HNRNPK/A549 cell lines were both increased (P<0.001). The number of migratory cells YBX1/A549 and HNRNPK/A549 was increased compared with A549 cells and those in Control Group (P<0.001), and cell migration rate of YBX1/A549 and HNRNPK/A549 was also enhanced compared with A549 cells and those in Control Group (P<0.001). The mRNA and protein levels of YBX1 in YBX1/A549 cell line were increased compared with those in Control Group (P<0.01), and the comparison of mRNA level and protein expression level of HNRNPK in YBX1/A549 cell line with the in Control Group showed no differences (P>0.05). The mRNA level and protein expression level of HNRNPK in HNRNPK/A549 cell line were enhanced compared with those in Control Group (P<0.01), and the comparison of YBX1 level and protein expression level in HNRNPK/A549 cell line with the in Control Group demonstrated no differences (P>0.05). YBX1 antibody adopted in co-IP was coated with magnetic beads, and numerous HNRNPK protein was abundant in YBX1/HNRNPK composite. The mRNA level and protein expression level of YBX1 and HNRNPK in YBX1/A549 and HNRNPK/A549 cell lines were enhanced compared with those in Control Group (P<0.001), and the comparison of mRNA level and protein expression level of CDC25 with those in Control Group showed no differences (P>0.05). The mRNA level and protein expression level of CDC25a in YBX1/HNRNPK/A549 were both higher than those in YBX1/A549 cell line and HNRNPK/A549 (P<0.001). With being induced by YBX1 or HNRNPK, the number of migratory cells CDC25/A549 was increased compared with that in Control Group (P<0.05). The mRNA level and protein expression level of CDC25a in YBX1/HNRNPK/A549 were both significantly higher than those in YBX1/A549 cell line and HNRNPK/A549 (P<0.001). All the above results indicated that the interaction between YBX1 and HNRNP regulated the expression of CDC25a, and further got involved in LC metastasis.


Subject(s)
Lung Neoplasms , Humans , Cell Line, Tumor , Cell Proliferation , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Lung Neoplasms/pathology , RNA, Messenger/genetics , Signal Transduction , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism
17.
Int J Biol Sci ; 18(16): 6084-6101, 2022.
Article in English | MEDLINE | ID: mdl-36439880

ABSTRACT

Background: Chloride channel 3 (CLCN3) is regulated by transcription-coactivator, however, it is unclear which core transcription factor regulates CLCN3. The role of CLCN3 in lung adenocarcinoma (LUAD) is unexplored and the relationship between CLCN3 and tumor microenvironment is unknown. Methods: A 5'-biotin-labeled promoter probe of CLCN3 was used to pull down the promoter-binding transcription factor. Further study was investigated using LUAD samples, cell lines, and xenograft mice models, and the mechanism was explored. Results: CLCN3 was upregulated in human LUAD, and CLCN3 knockdown inhibited tumor proliferation and migration in vitro. Next, heterogeneous nuclear ribonucleoprotein K (HNRNPK) was first validated as a CLCN3 promoter-binding transcription factor. Mechanistically, HNRNPK knockdown suppressed the promoter activity of CLCN3, thus regulating CLCN3 expression at the transcriptional level, and the binding motif 'GCGAGG' and binding site '-538/-248 bp' were identified. Subsequently, the RNA-seq data illustrated that the primary functions of HNRNPK were similar to those of CLCN3. The results from in vitro and in vivo trials indicated that the expression and function of CLCN3 were regulated by HNRNPK. By isolating primary cancer-associated fibroblasts (CAFs) from human LUAD, we confirmed that decreased extracellular CLCN3 secretion induced by HNRNPK knockdown inhibited CAFs activation and TGF-ß1 production, thus suppressing nuclear HNRNPK expression and LUAD progression in a feedback way. Furthermore, this phenomenon was rescued after the addition of TGF-ß1, revealing that the HNRNPK/CLCN3 axis facilitated LUAD progression through intercellular interactions. Finally, we identified that CLCN3 and HNRNPK were upregulated and correlated with poor prognosis in LUAD patients. Conclusions: HNRNPK/CLCN3 axis facilitates the progression of LUAD through CAF-tumor interaction.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Lung Neoplasms , Humans , Mice , Animals , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Transforming Growth Factor beta1/metabolism , Gene Expression Regulation, Neoplastic/genetics , Adenocarcinoma of Lung/metabolism , Transcription Factors/metabolism , Lung Neoplasms/metabolism , Tumor Microenvironment
18.
Nat Commun ; 13(1): 6614, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36329064

ABSTRACT

Heterogeneous Nuclear Ribonucleoprotein K (hnRNPK) is a multifunctional RNA binding protein (RBP) localized in the nucleus and the cytoplasm. Abnormal cytoplasmic enrichment observed in solid tumors often correlates with poor clinical outcome. The mechanism of cytoplasmic redistribution and ensuing functional role of cytoplasmic hnRNPK remain unclear. Here we demonstrate that the SCFFbxo4 E3 ubiquitin ligase restricts the pro-oncogenic activity of hnRNPK via K63 linked polyubiquitylation, thus limiting its ability to bind target mRNA. We identify SCFFbxo4-hnRNPK responsive mRNAs whose products regulate cellular processes including proliferation, migration, and invasion. Loss of SCFFbxo4 leads to enhanced cell invasion, migration, and tumor metastasis. C-Myc was identified as one target of SCFFbxo4-hnRNPK. Fbxo4 loss triggers hnRNPK-dependent increase in c-Myc translation, thereby contributing to tumorigenesis. Increased c-Myc positions SCFFbxo4-hnRNPK dysregulated cancers for potential therapeutic interventions that target c-Myc-dependence. This work demonstrates an essential role for limiting cytoplasmic hnRNPK function in order to maintain translational and cellular homeostasis.


Subject(s)
Carcinogenesis , Heterogeneous-Nuclear Ribonucleoprotein K , Humans , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Carcinogenesis/genetics , Ubiquitination , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Oncogenes , RNA, Messenger/metabolism
19.
J Virol ; 96(22): e0155522, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36317879

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is a re-emerging enteric coronavirus currently spreading in several nations and inflicting substantial financial damages on the swine industry. The currently available coronavirus vaccines do not provide adequate protection against the newly emerging viral strains. It is essential to study the relationship between host antiviral factors and the virus and to investigate the mechanisms underlying host immune response against PEDV infection. This study shows that heterogeneous nuclear ribonucleoprotein K (hnRNP K), the host protein determined by the transcription factor KLF15, inhibits the replication of PEDV by degrading the nucleocapsid (N) protein of PEDV in accordance with selective autophagy. hnRNP K was found to be capable of recruiting the E3 ubiquitin ligase, MARCH8, aiming to ubiquitinate N protein. Then, it was found that the ubiquitinated N protein could be delivered into autolysosomes for degradation by the cargo receptor NDP52, thereby inhibiting PEDV proliferation. Moreover, based on the enhanced MyD88 expression, we found that hnRNP K activated the interferon 1 (IFN-1) signaling pathway. Overall, the data obtained revealed a new mechanism of hnRNP K-mediated virus restriction wherein hnRNP K suppressed PEDV replication by degradation of viral N protein using the autophagic degradation pathway and by induction of IFN-1 production based on upregulation of MyD88 expression. IMPORTANCE The spread of the highly virulent PEDV in many countries is still leading to several epidemic and endemic outbreaks. To elucidate effective antiviral mechanisms, it is important to study the relationship between host antiviral factors and the virus and to investigate the mechanisms underlying host immune response against PEDV infection. In the work, we detected hnRNP K as a new host restriction factor which can hinder PEDV replication through degrading the nucleocapsid protein based on E3 ubiquitin ligase MARCH8 and the cargo receptor NDP52. In addition, via the upregulation of MyD88 expression, hnRNP K could also activate the interferon (IFN) signaling pathway. This study describes a previously unknown antiviral function of hnRNP K and offers a new vision toward host antiviral factors that regulate innate immune response as well as a protein degradation pathway against PEDV infection.


Subject(s)
Coronavirus Infections , Heterogeneous-Nuclear Ribonucleoprotein K , Interferon Type I , Porcine epidemic diarrhea virus , Virus Replication , Animals , Antiviral Agents , Chlorocebus aethiops , Coronavirus Infections/veterinary , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Interferons , Myeloid Differentiation Factor 88 , Nucleocapsid Proteins/physiology , Porcine epidemic diarrhea virus/physiology , Swine , Swine Diseases/virology , Ubiquitin-Protein Ligases , Vero Cells , Interferon Type I/immunology
20.
J Biol Chem ; 298(12): 102670, 2022 12.
Article in English | MEDLINE | ID: mdl-36334628

ABSTRACT

I-motifs are four-strand noncanonical secondary structures formed by cytosine (C)-rich sequences in living cells. The structural dynamics of i-motifs play essential roles in many cellular processes, such as telomerase inhibition, DNA replication, and transcriptional regulation. In cells, the structural dynamics of the i-motif can be modulated by the interaction of poly(C)-binding proteins (PCBPs), and the interaction is closely related to human health, through modulating the transcription of oncogenes and telomere stability. Therefore, the mechanisms of how PCBPs interact with i-motif structures are fundamentally important. However, the underlying mechanisms remain elusive. I-motif structures in the promoter of the c-MYC oncogene can be unfolded by heterogeneous nuclear ribonucleoprotein K (hnRNP K), a PCBP, to activate its transcription. Here, we selected this system as an example to comprehensively study the unfolding mechanisms. We found that the promoter sequence containing 5 C-runs preferred folding into type-1245 to type-1234 i-motif structures based on their folding stability, which was further confirmed by single-molecule FRET. In addition, we first revealed that the c-MYC i-motif structure was discretely resolved by hnRNP K through two intermediate states, which were assigned to the opposite hairpin and neighboring hairpin, as further confirmed by site mutations. Furthermore, we found all three KH (hnRNP K homology) domains of hnRNP K could unfold the c-MYC i-motif structure, and KH2 and KH3 were more active than KH1. In conclusion, this study may deepen our understanding of the interactions between i-motifs and PCBPs and may be helpful for drug development.


Subject(s)
Carrier Proteins , Heterogeneous-Nuclear Ribonucleoprotein K , Humans , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Carrier Proteins/metabolism , RNA-Binding Proteins/metabolism , DNA/metabolism , Protein Structure, Secondary
SELECTION OF CITATIONS
SEARCH DETAIL
...