Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Biomolecules ; 14(1)2023 Dec 28.
Article in English | MEDLINE | ID: mdl-38254641

ABSTRACT

BACKGROUND: Exosomes are essential for hepatocellular carcinoma (HCC) progression and have garnered significant interest as novel targets for diagnostic, prognostic, and therapeutic approaches. This study aims to identify potential exosome-related biomarkers for the development of useful strategies for HCC diagnosis and therapy. METHODS: Three datasets obtained from the Gene Expression Omnibus (GEO) were utilized to identify differentially expressed genes (DEGs) in HCC. Through Gene Ontology (GO) analysis and protein-protein interaction (PPI) network, overall survival (OS) analysis, Cox analyses, and diethylnitrosamine (DEN)-induced HCC mouse model detection, exosome-related hub gene was screened out, followed by a prognostic value assessment and immune-correlates analysis based on the Cancer Genome Atlas (TCGA) dataset. The hub gene-containing exosomes derived from Hepa1-6 cells were isolated and characterized using differential ultracentrifugation, transmission electron microscopy scanning, and Western blot. Ultrasound-guided intrahepatic injection, cell co-culture, CCK-8, and flow cytometry were performed to investigate the effects of the hub gene on macrophage infiltration and polarization in HCC. RESULTS: A total of 83 DEGs enriched in the extracellular exosome term, among which, FTCD, HRA, and C8B showed the strongest association with the progression of HCC. FTCD was independently associated with a protective effect in HCC and selected as the hub gene. The presence of FTCD in exosomes was confirmed. FTCD-stimulated macrophages were polarized towards the M1 type and suppressed HCC cells proliferation. CONCLUSIONS: FTCD is a potential exosome-related biomarker for HCC diagnosis, prognosis, and treatment. The crosstalk between FTCD-containing exosomes and macrophages in HCC progression deserves further investigation.


Subject(s)
Carcinoma, Hepatocellular , Exosomes , Glutamate Formimidoyltransferase , Liver Neoplasms , Animals , Mice , Blotting, Western , Carcinoma, Hepatocellular/genetics , Exosomes/genetics , Liver Neoplasms/genetics , Mice, Inbred Strains , Glutamate Formimidoyltransferase/metabolism
2.
EMBO J ; 40(9): e105853, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33555040

ABSTRACT

p97ATPase-mediated membrane fusion is required for the biogenesis of the Golgi complex. p97 and its cofactor p47 function in soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein receptor (SNARE) priming, but the tethering complex for p97/p47-mediated membrane fusion remains unknown. In this study, we identified formiminotransferase cyclodeaminase (FTCD) as a novel p47-binding protein. FTCD mainly localizes to the Golgi complex and binds to either p47 or p97 via its association with their polyglutamate motifs. FTCD functions in p97/p47-mediated Golgi reassembly at mitosis in vivo and in vitro via its binding to p47 and to p97. We also showed that FTCD, p47, and p97 form a big FTCD-p97/p47-FTCD tethering complex. In vivo tethering assay revealed that FTCD that was designed to localize to mitochondria caused mitochondria aggregation at mitosis by forming a complex with endogenous p97 and p47, which support a role for FTCD in tethering biological membranes in cooperation with the p97/p47 complex. Therefore, FTCD is thought to act as a tethering factor by forming the FTCD-p97/p47-FTCD complex in p97/p47-mediated Golgi membrane fusion.


Subject(s)
Ammonia-Lyases/metabolism , Glutamate Formimidoyltransferase/metabolism , Golgi Apparatus/metabolism , Multifunctional Enzymes/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Valosin Containing Protein/metabolism , Ammonia-Lyases/chemistry , Binding Sites , Glutamate Formimidoyltransferase/chemistry , HeLa Cells , Hep G2 Cells , Humans , Membrane Fusion , Mitochondria , Mitosis , Multifunctional Enzymes/chemistry , Multiprotein Complexes/metabolism , Protein Binding
3.
Med Sci Monit ; 25: 4474-4484, 2019 Jun 16.
Article in English | MEDLINE | ID: mdl-31203308

ABSTRACT

BACKGROUND Formiminotransferase cyclodeaminase (FTCD) is a candidate tumor suppressor gene in hepatocellular carcinoma (HCC). However, the mechanism for reduced expression of FTCD and its functional role in HCC remains unclear. In this study, we explored the biological functions of FTCD in HCC. MATERIAL AND METHODS The expression and clinical correlation of FTCD in HCC tissue were analyzed using TCGA (The Cancer Genome Atlas) and a cohort of 60 HCC patients. The MEXPRESS platform was accessed to identify the methylation level in promoter region FTCD. CCK-8 assay and flow cytometry analysis were used to explore the proliferation, cell apoptosis proportion, and DNA damage in HCC cells with FTCD overexpression. Western blot analysis was performed to identify the downstream target of FTCD. RESULTS FTCD is significantly downregulated in HCC tissues and cell lines. Low FTCD expression is correlated with a poor prognosis (P<0.001) and an aggressive tumor phenotype, including AFP levels (P=0.009), tumor size (P=0.013), vascular invasion (P=0.001), BCLC stage (P=0.024), and pTNM stage (P<0.001). Bioinformatics analysis indicated promoter hypermethylation can result in decreased expression of FTCD. FTCD overexpression suppressed cell proliferation by promoting DNA damage and inducing cell apoptosis in HCC cells. FTCD overexpression resulted in increased level of PTEN protein, but a decrease in PI3K, total Akt, and phosphorylated Akt protein in HCC cells, suggesting involvement of the PI3K/Akt pathway. CONCLUSIONS FTCD acts as a tumor suppressor gene in HCC pathogenesis and progression and is a candidate prognostic marker and a possible therapeutic target for this disease.


Subject(s)
Ammonia-Lyases/metabolism , Carcinoma, Hepatocellular/metabolism , Glutamate Formimidoyltransferase/metabolism , Multifunctional Enzymes/metabolism , Aged , Ammonia-Lyases/physiology , Apoptosis/physiology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , China , DNA Damage/physiology , Female , Gene Expression Regulation, Neoplastic/genetics , Glutamate Formimidoyltransferase/physiology , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Multifunctional Enzymes/physiology , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology
4.
Nature ; 559(7715): 632-636, 2018 07.
Article in English | MEDLINE | ID: mdl-29995852

ABSTRACT

The chemotherapeutic drug methotrexate inhibits the enzyme dihydrofolate reductase1, which generates tetrahydrofolate, an essential cofactor in nucleotide synthesis2. Depletion of tetrahydrofolate causes cell death by suppressing DNA and RNA production3. Although methotrexate is widely used as an anticancer agent and is the subject of over a thousand ongoing clinical trials4, its high toxicity often leads to the premature termination of its use, which reduces its potential efficacy5. To identify genes that modulate the response of cancer cells to methotrexate, we performed a CRISPR-Cas9-based screen6,7. This screen yielded FTCD, which encodes an enzyme-formimidoyltransferase cyclodeaminase-that is required for the catabolism of the amino acid histidine8, a process that has not previously been linked to methotrexate sensitivity. In cultured cancer cells, depletion of several genes in the histidine degradation pathway markedly decreased sensitivity to methotrexate. Mechanistically, histidine catabolism drains the cellular pool of tetrahydrofolate, which is particularly detrimental to methotrexate-treated cells. Moreover, expression of the rate-limiting enzyme in histidine catabolism is associated with methotrexate sensitivity in cancer cell lines and with survival rate in patients. In vivo dietary supplementation of histidine increased flux through the histidine degradation pathway and enhanced the sensitivity of leukaemia xenografts to methotrexate. The histidine degradation pathway markedly influences the sensitivity of cancer cells to methotrexate and may be exploited to improve methotrexate efficacy through a simple dietary intervention.


Subject(s)
Histidine/metabolism , Methotrexate/pharmacology , Methotrexate/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Ammonia-Lyases/deficiency , Ammonia-Lyases/genetics , Ammonia-Lyases/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Female , Folic Acid Antagonists/pharmacology , Folic Acid Antagonists/therapeutic use , Glutamate Formimidoyltransferase/deficiency , Glutamate Formimidoyltransferase/genetics , Glutamate Formimidoyltransferase/metabolism , Histidine/pharmacology , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Multifunctional Enzymes , Nucleotides/biosynthesis , Reduced Folate Carrier Protein/genetics , Reduced Folate Carrier Protein/metabolism , Tetrahydrofolate Dehydrogenase/metabolism , Tetrahydrofolates/deficiency , Tetrahydrofolates/metabolism , Xenograft Model Antitumor Assays
5.
Mol Cell Proteomics ; 14(5): 1400-10, 2015 May.
Article in English | MEDLINE | ID: mdl-25724911

ABSTRACT

The data-independent acquisition (DIA) approach has recently been introduced as a novel mass spectrometric method that promises to combine the high content aspect of shotgun proteomics with the reproducibility and precision of selected reaction monitoring. Here, we evaluate, whether SWATH-MS type DIA effectively translates into a better protein profiling as compared with the established shotgun proteomics. We implemented a novel DIA method on the widely used Orbitrap platform and used retention-time-normalized (iRT) spectral libraries for targeted data extraction using Spectronaut. We call this combination hyper reaction monitoring (HRM). Using a controlled sample set, we show that HRM outperformed shotgun proteomics both in the number of consistently identified peptides across multiple measurements and quantification of differentially abundant proteins. The reproducibility of HRM in peptide detection was above 98%, resulting in quasi complete data sets compared with 49% of shotgun proteomics. Utilizing HRM, we profiled acetaminophen (APAP)(1)-treated three-dimensional human liver microtissues. An early onset of relevant proteome changes was revealed at subtoxic doses of APAP. Further, we detected and quantified for the first time human NAPQI-protein adducts that might be relevant for the toxicity of APAP. The adducts were identified on four mitochondrial oxidative stress related proteins (GATM, PARK7, PRDX6, and VDAC2) and two other proteins (ANXA2 and FTCD). Our findings imply that DIA should be the preferred method for quantitative protein profiling.


Subject(s)
Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Hepatocytes/drug effects , Liver/drug effects , Peptides/analysis , Proteome/analysis , Amidinotransferases/analysis , Amidinotransferases/genetics , Amidinotransferases/metabolism , Ammonia-Lyases/analysis , Ammonia-Lyases/genetics , Ammonia-Lyases/metabolism , Annexin A2/analysis , Annexin A2/genetics , Annexin A2/metabolism , Gene Expression , Glutamate Formimidoyltransferase/analysis , Glutamate Formimidoyltransferase/genetics , Glutamate Formimidoyltransferase/metabolism , Hepatocytes/metabolism , Humans , Intracellular Signaling Peptides and Proteins/analysis , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Multifunctional Enzymes , Oncogene Proteins/analysis , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Peroxiredoxin VI/analysis , Peroxiredoxin VI/genetics , Peroxiredoxin VI/metabolism , Protein Deglycase DJ-1 , Proteolysis , Proteome/genetics , Proteome/metabolism , Proteomics/methods , Tissue Culture Techniques , Trypsin/chemistry , Voltage-Dependent Anion Channel 2/analysis , Voltage-Dependent Anion Channel 2/genetics , Voltage-Dependent Anion Channel 2/metabolism
6.
Cell Signal ; 26(7): 1560-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24686083

ABSTRACT

HIF-1α is implicated in hepatocellular carcinoma (HCC) pathologies. Here, we screened a human liver cDNA library for HIF-1α-interacting partners using a yeast two-hybrid system. We identified 53 genes, including formiminotransferase cyclodeaminase (FTCD), which was confirmed by co-immunoprecipitation. Moreover, our data indicated that HIF-1α mediated the effects of hypoxia on FTCD induction via binding to the hypoxia-responsive elements of the FTCD promoter. Knockdown of FTCD reduced the effects of HIF-1α in hypoxia and enhanced chemosensitivity in HepG2 cells. Our findings suggested crosstalk between FTCD and HIF signaling and promoted HCC progression, thus implicating FTCD as a therapeutic target for HCC.


Subject(s)
Ammonia-Lyases/metabolism , Carcinoma, Hepatocellular/pathology , Glutamate Formimidoyltransferase/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver Neoplasms/pathology , Two-Hybrid System Techniques , Ammonia-Lyases/biosynthesis , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Proliferation , Glutamate Formimidoyltransferase/biosynthesis , Hep G2 Cells , Humans , Multifunctional Enzymes , Neoplasm Invasiveness/pathology , Promoter Regions, Genetic , Protein Binding , RNA Interference , RNA, Small Interfering , Response Elements/genetics , Signal Transduction
7.
J Biol Chem ; 285(53): 41557-66, 2010 Dec 31.
Article in English | MEDLINE | ID: mdl-20952389

ABSTRACT

5-Formyltetrahydrofolate (5-CHO-THF) is formed by a side reaction of serine hydroxymethyltransferase. Unlike other folates, it is not a one-carbon donor but a potent inhibitor of folate enzymes and must therefore be metabolized. Only 5-CHO-THF cycloligase (5-FCL) is generally considered to do this. However, comparative genomic analysis indicated (i) that certain prokaryotes lack 5-FCL, implying that they have an alternative 5-CHO-THF-metabolizing enzyme, and (ii) that the histidine breakdown enzyme glutamate formiminotransferase (FT) might moonlight in this role. A functional complementation assay for 5-CHO-THF metabolism was developed in Escherichia coli, based on deleting the gene encoding 5-FCL (ygfA). The deletion mutant accumulated 5-CHO-THF and, with glycine as sole nitrogen source, showed a growth defect; both phenotypes were complemented by bacterial or archaeal genes encoding FT. Furthermore, utilization of supplied 5-CHO-THF by Streptococcus pyogenes was shown to require expression of the native FT. Recombinant bacterial and archaeal FTs catalyzed formyl transfer from 5-CHO-THF to glutamate, with k(cat) values of 0.1-1.2 min(-1) and K(m) values for 5-CHO-THF and glutamate of 0.4-5 µM and 0.03-1 mM, respectively. Although the formyltransferase activities of these proteins were far lower than their formiminotransferase activities, the K(m) values for both substrates relative to their intracellular levels in prokaryotes are consistent with significant in vivo flux through the formyltransferase reaction. Collectively, these data indicate that FTs functionally replace 5-FCL in certain prokaryotes.


Subject(s)
Carbon-Nitrogen Ligases/chemistry , Glutamate Formimidoyltransferase/metabolism , Animals , Archaea/metabolism , Escherichia coli/enzymology , Escherichia coli/metabolism , Folic Acid/chemistry , Genetic Complementation Test , Genomics , Glutamic Acid/chemistry , Histidine/chemistry , Kinetics , Models, Genetic , Mutation , Phenotype , Recombinant Proteins/chemistry , Streptococcus pyogenes/metabolism , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...