Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
J Biol Chem ; 298(4): 101757, 2022 04.
Article in English | MEDLINE | ID: mdl-35202654

ABSTRACT

The aminoacyl-tRNA synthetases are an ancient and ubiquitous component of all life. Many eukaryotic synthetases balance their essential function, preparing aminoacyl-tRNA for use in mRNA translation, with diverse roles in cell signaling. Herein, we use long-read sequencing to discover a leukocyte-specific exon skipping event in human leucyl-tRNA synthetase (LARS). We show that this highly expressed splice variant, LSV3, is regulated by serine-arginine-rich splicing factor 1 (SRSF1) in a cell-type-specific manner. LSV3 has a 71 amino acid deletion in the catalytic domain and lacks any tRNA leucylation activity in vitro. However, we demonstrate that this LARS splice variant retains its role as a leucine sensor and signal transducer for the proliferation-promoting mTOR kinase. This is despite the exon deletion in LSV3 including a portion of the previously mapped Vps34-binding domain used for one of two distinct pathways from LARS to mTOR. In conclusion, alternative splicing of LARS has separated the ancient catalytic activity of this housekeeping enzyme from its more recent evolutionary role in cell signaling, providing an opportunity for functional specificity in human immune cells.


Subject(s)
Alternative Splicing , Leucine-tRNA Ligase , Humans , Leucine-tRNA Ligase/genetics , Leucine-tRNA Ligase/metabolism , RNA, Transfer/metabolism , Serine-Arginine Splicing Factors/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
2.
J Transl Med ; 19(1): 210, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001131

ABSTRACT

BACKGROUND: As a significant cause of malignancy mortality, gastric carcinoma (GC) has been well documented to be an often-fatal diagnosis. Despite the limitations of effective therapy, immunotherapy has emerged as a promising therapeutic approach capable of killing cancer cells via the immune system. The current study was conducted to investigate the effect of cytokine C-C motif chemokine ligand 21 (CCL21) on GC progression through the metastasis-associated lung adenocarcinoma transcript 1/serine arginine-rich splicing factor 1/mammalian target of rapamycin (MALAT1/SRSF1/mTOR) axis. METHODS: Bioinformatics analysis was conducted to identify the key genes associated with GC and to subsequently predict their downstream genes. The effect of CCL21, MALAT1, and SRSF1 on the malignant phenotypes and epithelial-mesenchymal transition (EMT) of SGC-7901 and MGC-803 cells in-vitro and the tumorigenesis of SGC-7901 and MGC-803 cells in-vivo were assessed by expression determination and plasmid transfection. Additionally, RNA pull-down and RNA binding protein immunoprecipitation experiments were performed to determine the MALAT1-microRNA-202-3p (miR-203-3p) interaction and miR-202-3p-SRSF1 interaction followed by the analysis of their effect on the mTOR pathway. RESULTS: CCL21 was identified as a key GC immune gene. Overexpressed CCL21, MALAT1, and SRSF1 along with poorly expressed miR-202-3p were identified in the GC cells. CCL21 induced the MALAT1 expression in a time- and dose-dependent manner. Functionally, MALAT1 targeted miR-202-3p but upregulated SRSF1 and activated mTOR. Crucially, evidence was obtained indicating that CCL21 promoted both the malignant phenotypes and EMT of SGC-7901 and MGC-803 cells in-vitro and the tumorigenesis of SGC-7901 and MGC-803 cells in-vivo by increasing the MALAT1-induced upregulation of SRSF1. CONCLUSIONS: Taken together, the key observations of our study provide evidence that CCL21 enhances the progression of GC via the MALAT1/SRSF1/mTOR axis, providing a novel therapeutic target for the treatment of GC.


Subject(s)
Carcinoma , MicroRNAs , RNA, Long Noncoding , Cell Line, Tumor , Chemokine CCL21 , Chemokines , Gene Expression Regulation, Neoplastic , Humans , Ligands , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Serine-Arginine Splicing Factors , Stomach Neoplasms , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
3.
Mol Med Rep ; 24(1)2021 Jul.
Article in English | MEDLINE | ID: mdl-33982782

ABSTRACT

Atherosclerosis is a primary cause of multiple types of cardiovascular disease, including myocardial infarction. In addition, injury of human umbilical vein endothelial cells (HUVECs) can lead to the development of atherosclerosis. Circular (circ)RNAs participate in atherosclerosis. It has previously been shown that circRNA cSMARCA5 (hsa_circ_0001445) expression is downregulated in atherosclerosis. However, the effects of hsa_circ_0001445 on the proliferation of HUVECs remain unclear. In order to mimic atherosclerosis in vitro, HUVECs were treated with oxidized low­density lipoprotein (oxLDL). The expression levels of specific genes and proteins were detected in HUVECs by reverse transcription­quantitative PCR and western blot analysis, respectively. Cell proliferation was assessed by Cell Counting Kit­8 and 5­Ethynyl­2'­deoxyuridine staining. Cell apoptosis and 5,5',6,6'­Tetrachloro­1,1',3,3'­tetraethyl­imidacarbocyanine staining were examined by flow cytometry. In addition, the association between hsa_circ_0001445 and serine/arginine­rich splicing factor 1 (SRSF1) was investigated by RNA pull­down assay. hsa_circ_0001445 expression was downregulated in oxLDL­treated HUVECs. Moreover, oxLDL­induced inhibition of HUVEC proliferation was significantly reversed by overexpression of hsa_circ_0001445. oxLDL notably inhibited tube formation and mitochondrial membrane potential in HUVECs, while these effects were markedly reversed by hsa_circ_0001445 overexpression. Furthermore, overexpression of hsa_circ_0001445 reversed oxLDL­induced activation of ß­catenin by binding to SRSF1. Collectively, these data demonstrated that overexpression of hsa_circ_0001445 reversed oxLDL­induced inhibition of HUVEC proliferation via activation of the SRSF1/ß­catenin axis. These findings may provide novel targets for the treatment of atherosclerosis.


Subject(s)
Cell Proliferation/genetics , Human Umbilical Vein Endothelial Cells/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Serine-Arginine Splicing Factors/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cells, Cultured , Epithelial-Mesenchymal Transition/genetics , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Lipoproteins, LDL/toxicity , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/genetics , Neovascularization, Pathologic/genetics , beta Catenin/metabolism
4.
Eur J Pharmacol ; 890: 173669, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33098832

ABSTRACT

Glioma is the most common primary intracranial tumor, in which glioblastoma (GBM) is the most malignant and lethal. However, the current chemotherapy drugs are still unsatisfactory for GBM therapy. As the natural products mainly extracted from Eucalyptus species, phloroglucinol-terpene adducts have the potential to be anti-cancer lead compounds that attracted increasing attention. In order to discover the new lead compounds with the anti-GBM ability, we isolated Eucalyptal A with a phloroglucinol-terpene skeleton from the fruit of E. globulus and investigated its anti-GBM activity in vitro and in vivo. Functionally, we verified that Eucalyptal A could inhibit the proliferation, growth and invasiveness of GBM cells in vitro. Moreover, Eucalyptal A had the same anti-GBM activity in tumor-bearing mice as in vitro and prolonged the overall survival time by maintaining mice body weight. Further mechanism research revealed that Eucalyptal A downregulated SRSF1 expression and rectified SRSF1-guided abnormal alternative splicing of MYO1B mRNA, which led to anti-GBM activity through the PDK1/AKT/c-Myc and PAK/Cofilin axes. Taken together, we identified Eucalyptal A as an important anti-GBM lead compound, which represents a novel direction for glioma therapy.


Subject(s)
Brain Neoplasms/metabolism , Carcinogenesis/drug effects , Eucalyptol/therapeutic use , Glioma/metabolism , Myosin Type I/metabolism , Protein Splicing/drug effects , Serine-Arginine Splicing Factors/biosynthesis , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/therapeutic use , Brain Neoplasms/genetics , Brain Neoplasms/prevention & control , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Drugs, Chinese Herbal/isolation & purification , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Eucalyptol/isolation & purification , Eucalyptol/pharmacology , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/prevention & control , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Myosin Type I/genetics , Protein Splicing/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serine-Arginine Splicing Factors/antagonists & inhibitors , Serine-Arginine Splicing Factors/genetics , Xenograft Model Antitumor Assays/methods
5.
Neuromuscul Disord ; 30(8): 685-692, 2020 08.
Article in English | MEDLINE | ID: mdl-32690349

ABSTRACT

LGMD D2 is a disease caused by TNPO3 mutation. We describe the expression of TNPO3 and selected proteins, likely modified by TNPO3 mutation, in muscle biopsies of affected patients. We also aim to find other genes involved in pathways correlated to TNPO3. Our morphological study on LGMD D2 muscle described the expression of TNPO3 and SRSF1, a splicing factor transported by TNPO3. Moreover, we investigated some sarcomeric and nuclear proteins, likely altered by TNPO3 mutation. Through an in silico approach we tried to identify genes involved in pathways that include, besides TNPO3 and SRSF1, p62 and Murf-1, altered in LGMD D2. In patients' muscles TNPO3 appeared weaker and randomly organized, with sporadic cytoplasmic aggregates positive for TNPO3; both SRSF1 and sarcomeric alpha actinin showed a different expression, while there were no alterations in the expression of the nuclear proteins. The in silico study lead to identify five genes, all coding for proteins responsible for muscle contraction. Our data suggest a possible interference in the morphology and function of myofibrillar network by mutated TNPO3; these findings are supported by the in silico identification of genes involved in muscle contraction that could help to explain the pathogenic mechanisms of LGMD D2.


Subject(s)
Muscular Dystrophies, Limb-Girdle/pathology , beta Karyopherins/genetics , Biopsy , Computer Simulation , Exome , Female , Humans , In Vitro Techniques , Muscle Proteins , Muscles/pathology , Muscular Dystrophies, Limb-Girdle/genetics , Mutation/genetics , Nuclear Proteins/genetics , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
6.
Cell Mol Biol (Noisy-le-grand) ; 64(7): 43-50, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29974845

ABSTRACT

Abnormal proliferation of vascular smooth muscle cells (VSMCs) induced by high cyclic stretch is crucial in the vascular remodeling during hypertension. Vascular endothelial growth factor A (VEGFA) alternative splicing plays important roles in the pathological process of vascular diseases and remodeling. However, the roles of VEGFA isoforms in modulating VSMC functions in response to cyclic stretch remain unclear. We hypothesize that high cyclic stretch may induce VEGFA alternative splicing via Serine/arginine-rich splicing factor 1 (SRSF1) which subsequently induce VSMC proliferation. In the present research, hypertensive rat model was established using the abdominal aortic constriction method. In comparison with sham-operated group, immunohistology staining showed translocation of SRSF1 into nuclei in hypertensive rat thoracic aorta, and RT-PCR detected a shift of VEGFA expression pattern, including the increased expression of VEGFA120 and VEGFA164, but not VEGFA188.Then VSMCs were subjected to cyclic stretch in vitro using a Flexercell strain unit. VEGFA ELISA assay showed 15% cyclic stretch increased the secretion of VEGFA which significantly increased proliferation of VSMCs. Western blot and immunofluorescence detected accumulation of SRSF1 in nuclei after 15% cyclic stretch application. Furthermore, SRSF1-specific siRNA transfection reversed the VEGFA secretion induced by pathological high cyclic stretch. Our present results suggested that pathologically high cyclic stretch induces the shuttling of SRSF1 which results in the secretive pattern splicing of VEGFA and finally contributes to the proliferation of VSMCs.


Subject(s)
Alternative Splicing , Hypertension/pathology , Muscle, Smooth, Vascular/pathology , Serine-Arginine Splicing Factors/metabolism , Vascular Endothelial Growth Factor A/genetics , Animals , Aorta, Thoracic/pathology , Cell Nucleus/metabolism , Cell Proliferation , Disease Models, Animal , Humans , Hypertension/metabolism , Male , Muscle, Smooth, Vascular/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Serine-Arginine Splicing Factors/genetics , Vacuum
7.
Oncotarget ; 9(12): 10784-10807, 2018 Feb 13.
Article in English | MEDLINE | ID: mdl-29535843

ABSTRACT

Cancer is a complex, multi-factorial, multi-stage disease and a global threat to human health. Early detection of nature and stage of cancer is highly crucial for disease management. Recent studies have proved beyond any doubt about the involvement of the ubiquitous, myriad ligand binding, multi-functional human protein, hyaluronan-binding protein 1 (HABP1), which is identical to the splicing factor associated protein (p32) and the receptor of the globular head of the complement component (gC1qR) in tumorigenesis and cancer metastasis. Simultaneously three laboratories have discovered and named this protein separately as mentioned. Subsequently, different scientists have worked on the distinct functions in cellular processes ranging from immunological response, splicing mechanism, sperm-oocyte interactions, cell cycle regulation to cancer and have concentrated in their respective area of interest, referring it as either p32 or gC1qR or HABP1. HABP1 overexpression has been reported in almost all the tissue-specific forms of cancer and correlated with stage and poor prognosis in patients. In order to tackle this deadly disease and for therapeutic intervention, it is imperative to focus on all the regulatory aspects of this protein. Hence, this work is an attempt to combine an assortment of information on this protein to have an overview, which suggests its use as a diagnostic marker for cancer. The knowledge might assist in the designing of drugs for therapeutic intervention of HABP1/p32/gC1qR regulated specific ligand mediated pathways in cancer.

8.
Oncol Lett ; 14(5): 5393-5399, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29113173

ABSTRACT

Colorectal cancer is the third most common type of cancer and the fourth leading cause of cancer-associated mortality worldwide. Serine/arginine-rich splicing factor 1 (SRSF1) is a well-characterized oncogenic factor that promotes tumorigenesis by controlling a number of alternative splicing events. However, there is limited network analysis, from a global aspect, to study the effect of SRSF1 on colorectal cancer. In the present study, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of available gene regulation data from The Cancer Genome Atlas database revealed the enriched functions and signaling pathways of SRSF1. Subsequently, Oncomine analysis was performed, which demonstrated that SRSF1 was upregulated in a number of types of colon cancer. From overlapping the analysis of 2,678 SRSF1-related genes and 3,625 colorectal cancer genes in GeneCards, 468 genes were identified as SRSF1-related colorectal cancer genes. The GO results revealed that these overlapped genes were primarily enriched in metabolic processes, response to DNA damage, regulation of the cell cycle and a number of additional biological processes. KEGG pathway analysis revealed that SRSF1-related colorectal cancer genes were associated with the cell cycle, deregulated signaling pathways associated with cancer progression and colorectal cancer signaling pathways. In addition, the Search Tool for the Retrieval of Interacting Genes/Proteins database and Cytoscape analysis demonstrated that 468 SRSF1-related colorectal cancer genes exhibit potential interaction networks in which these genes were enriched in DNA metabolic processes, cell cycle regulation and regulation of apoptosis. The results of the present study suggested that SRSF1 exhibited an increased degree of interaction with key molecules, including NUF2 NDC80 kinetochore complex component, kinesin family member 2C, structural maintenance of chromosomes 3, ATM serine/threonine kinase, BRCA1 DNA repair associated, protein kinase DNA-activated catalytic polypeptide, heat shock protein 90 alpha family class A member 1, ras homolog family member A, and phosphatase and tensin homolog. Collectively, the bioinformatics analysis of the present study indicated that SRSF1 may have key functions in the progression and development of colorectal cancer.

9.
Alcohol Clin Exp Res ; 41(10): 1715-1724, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28800142

ABSTRACT

BACKGROUND: Heavy and chronic ethanol (EtOH) exposure can cause significant structural and functional damage to the adult brain. The most devastating consequence of EtOH exposure is the neurotoxicity associated with the depletion of neurons. Regulation of splice variants in the brain can modulate protein functions, which may ultimately affect behaviors associated with alcohol dependence and EtOH-mediated neurotoxicity. As alcohol consumption is associated with neurotoxicity, it is possible that altered splicing of survival and pro-survival factors during the development of alcoholism may contribute to the neurotoxicity. METHODS: Primary human neurons and a neuroblastoma cell line were exposed to different concentrations of EtOH for various time periods. Cell viability and neuronal marker expression were analyzed by MTT assay and immunoblotting, respectively. Effect of EtOH exposure on splicing regulatory protein expression and alternative splicing of candidate genes was analyzed by a biochemical approach. Transcriptional activity of serine/arginine-rich splicing factor 1 (SRSF1) gene was determined by reporter gene analysis. RESULTS: Our results suggest that EtOH exposure to neuronal cells at 25 mM and higher concentrations are detrimental. In addition, EtOH exposure caused a dramatic reduction in SRSF1 expression levels. Furthermore, EtOH exposure led to pre-mRNA missplicing of Mcl-1, a pro-survival member of the Bcl-2 family, by down-regulating the expression levels of SRSF1. Moreover, ectopic expression of both SRSF1 and Mcl-1L isoform was able to recover EtOH-mediated neurotoxicity. CONCLUSIONS: Our results suggest that EtOH exposure can lead to pre-mRNA missplicing of Mcl-1 in neuronal cells. Our results indicate that EtOH exposure of neurons leads to a decrease in the ratio of Mcl-1L/Mcl-1S by favoring pro-apoptotic Mcl-1S splicing over anti-apoptotic Mcl-1L isoform suggesting that Mcl-1S may play a crucial role in neurotoxicity associated with alcohol consumption.


Subject(s)
Alternative Splicing/physiology , Ethanol/toxicity , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neurons/physiology , RNA Precursors/genetics , Alternative Splicing/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Neurons/drug effects , RNA Precursors/biosynthesis
10.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-486781

ABSTRACT

Objective To test the expression of serine/arginine rich splicing factor 1(SRSF1)and apoptosis inhibiting factor(Survivin) in prostate cancer,and study their correlation with the pathological features of prostate cancer,so as to put forward the new targets in the treatment of prostate cancer. Methods SRSF1 and Survivin protein was determined in 20 prostate tissue samples including prostate cancer(n=12)and benign prostat?ic hyperplasia(n=8)by immunohistochemical SP method. SRSF1 and Survivin was correlated to pathological features,and both the relevance was analyzed(no related reports at home and abroad). Results The positive expression rate of SRSF1 protein in prostate cancer tissue cells was 76.37± 5.06%,which was significantly higher than that of benign prostatic hyperplasia(11.30%±1.09%,P<0.05);the positive expression rate of Survivin protein in prostate cancer tissue cells was 86.93%±3.21%,which was significantly higher than that of benign prostatic hyperplasia(17.67%±1.99%, P<0.05);SRSF1 and Survivin protein expressed in prostate cancer organizations and were positively correlated to pathological Gleason grading, and there was significant correlation(P<0.05). Conclusion SRSF1 and Survivin protein were highly expressed in adenocarcinoma tissue,which were significantly increased than that of benign hyperplasia of prostate tissue. The positive expression SRSF1 and Survivin protein were positively cor?related to pathological Gleason grading.The expression of Survivin protein was elevated with the expression of SRSF1 protein in prostate cancer. These preliminary evidence indicated that SRSF1 may up?regulate the expression of Survivin,and thus promote the occurrence and development of prostate cancer.

SELECTION OF CITATIONS
SEARCH DETAIL