Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
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
2.
PLoS Genet ; 15(3): e1007984, 2019 03.
Article in English | MEDLINE | ID: mdl-30893314

ABSTRACT

Inorganic arsenic (iAs) is a carcinogen, and exposure to iAs via food and water is a global public health problem. iAs-contaminated drinking water alone affects >100 million people worldwide, including ~50 million in Bangladesh. Once absorbed into the blood stream, most iAs is converted to mono-methylated (MMA) and then di-methylated (DMA) forms, facilitating excretion in urine. Arsenic metabolism efficiency varies among individuals, in part due to genetic variation near AS3MT (arsenite methyltransferase; 10q24.32). To identify additional arsenic metabolism loci, we measured protein-coding variants across the human exome for 1,660 Bangladeshi individuals participating in the Health Effects of Arsenic Longitudinal Study (HEALS). Among the 19,992 coding variants analyzed exome-wide, the minor allele (A) of rs61735836 (p.Val101Met) in exon 3 of FTCD (formiminotransferase cyclodeaminase) was associated with increased urinary iAs% (P = 8x10-13), increased MMA% (P = 2x10-16) and decreased DMA% (P = 6x10-23). Among 2,401 individuals with arsenic-induced skin lesions (an indicator of arsenic toxicity and cancer risk) and 2,472 controls, carrying the low-efficiency A allele (frequency = 7%) was associated with increased skin lesion risk (odds ratio = 1.35; P = 1x10-5). rs61735836 is in weak linkage disequilibrium with all nearby variants. The high-efficiency/major allele (G/Valine) is human-specific and eliminates a start codon at the first 5´-proximal Kozak sequence in FTCD, suggesting selection against an alternative translation start site. FTCD is critical for catabolism of histidine, a process that generates one-carbon units that can enter the one-carbon/folate cycle, which provides methyl groups for arsenic metabolism. In our study population, FTCD and AS3MT SNPs together explain ~10% of the variation in DMA% and support a causal effect of arsenic metabolism efficiency on arsenic toxicity (i.e., skin lesions). In summary, this work identifies a coding variant in FTCD associated with arsenic metabolism efficiency, providing new evidence supporting the established link between one-carbon/folate metabolism and arsenic toxicity.


Subject(s)
Ammonia-Lyases/genetics , Arsenic/toxicity , Glutamate Formimidoyltransferase/genetics , Methyltransferases/genetics , Adult , Alleles , Ammonia-Lyases/physiology , Arsenic/metabolism , Arsenic Poisoning , Bangladesh , Environmental Exposure , Female , Folic Acid/metabolism , Gene Frequency/genetics , Glutamate Formimidoyltransferase/physiology , Humans , Male , Methylation , Methyltransferases/metabolism , Multifunctional Enzymes , Mutation, Missense , Odds Ratio , Phenotype , Polymorphism, Single Nucleotide/genetics , Risk Factors , Skin Diseases/chemically induced , Skin Diseases/genetics , Water Pollutants, Chemical
3.
Apoptosis ; 10(5): 1105-10, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16151643

ABSTRACT

The Golgi complex is thought to play an important role in the apoptotic process of osteoarthritic (OA) chondrocytes. However, the exact relationship between modifications of the Golgi complex and apoptosis in human OA cartilage requires to be established. We compared the patterns and immunolabeling intensities for anti-Golgi 58 K protein with apoptosis markers such as TUNEL and caspase-2L in OA cartilage removed from patients during knee total replacement surgery. We observed important modifications in labeling of the Golgi 58 K protein in OA chondrocytes compared with normal cell. Immunohistochemical analysis revealed co-localization between 58 K protein and caspase-2L, suggesting that this enzyme was localized in Golgi complex of OA chondrocytes. In addition, these cells labeled positive with the TUNEL technique, but in different proportions to caspase-2L. Our results support the concept, previously reported, that apoptosis in OA cartilage (chondroptosis) might be a variant of the classical apoptosis.


Subject(s)
Apoptosis/physiology , Cartilage, Articular/pathology , Chondrocytes/cytology , Golgi Apparatus/drug effects , Osteoarthritis/physiopathology , Aged , Ammonia-Lyases/physiology , Caspases/analysis , Female , Humans , In Situ Nick-End Labeling , Male , Middle Aged , Osteoarthritis/pathology
4.
Eur J Biochem ; 184(3): 635-41, 1989 Oct 01.
Article in English | MEDLINE | ID: mdl-2806247

ABSTRACT

Agrobacterium nopaline Ti plasmids code for three enzymes of nopaline [N2-(1,3-dicarboxypropyl)-L-arginine] degradation: nopaline oxidase, arginase, and ornithine cyclodeaminase. We describe the DNA sequence of the arginase gene, a comparison of the deduced protein sequence with eucaryotic arginases, and properties of the procaryotic enzyme. The results show that the agrobacterial arginase is related with arginases from yeast, rat liver, and human liver (28-33% identity). The Ti plasmid enzyme revealed several properties which appear common to all arginases, but it does not utilize L-canavanine as substrate, and its Mn2+ requirement is not satisfied by Fe2+, Co2+, or Ni2+. The properties of arginase and ornithine cyclodeaminase are discussed as part of the mechanisms which avoid depletion of L-arginine and L-ornithine pools for biosynthetic reactions during catabolic utilization of nopaline.


Subject(s)
Arginase/genetics , DNA/analysis , Plasmids , Rhizobium/enzymology , Amino Acid Sequence , Ammonia-Lyases/physiology , Animals , Arginase/analysis , Base Sequence , Humans , Kinetics , Molecular Sequence Data , Rats , Substrate Specificity
5.
J Bacteriol ; 162(1): 98-101, 1985 Apr.
Article in English | MEDLINE | ID: mdl-3884601

ABSTRACT

Histidine ammonia-lyase catalyzes the first step in histidine catabolism, the deamination of histidine to urocanate and ammonia. In vitro experiments have shown that histidine ammonia-lyase also can catalyze the reverse (amination) reaction, histidine synthesis, relatively efficiently under extreme reaction conditions (4 M NH4OH, pH 10). An Escherichia coli hisB deletion strain was transformed with a pBR322 derivative plasmid (pCB101) containing the entire Klebsiella aerogenes histidine utilization (hut) operon to determine whether the catabolic histidine ammonia-lyase could function biosynthetically in vivo to satisfy the histidine auxotrophy. Although the initial construct did not grow on media containing urocanate and ammonia as a source of histidine, spontaneous mutants possessing this ability were isolated. Four mutants characterized grew at doubling times of 4 h compared with 1 h when histidine was present, suggesting that histidine synthesis, although unequivocally present, remained growth limiting. Each mutant contained a plasmid-encoded mutation which eliminated urocanase activity, the second enzyme in the Hut catabolic pathway. This genetic block led to the accumulation of high intracellular levels of urocanate, which was subsequently converted to histidine via histidine ammonia-lyase, thus satisfying the histidine auxotrophic requirement.


Subject(s)
Ammonia-Lyases/physiology , Escherichia coli/metabolism , Histidine Ammonia-Lyase/physiology , Histidine/biosynthesis , Mutation , Plasmids , Urocanic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL