Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
1.
Cell Death Dis ; 15(10): 728, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39368995

ABSTRACT

Patients with lung adenocarcinoma (LUAD) generally have poor prognosis. Abnormal cellular energy metabolism is a hallmark of LUAD. Glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1) is a member of the γ-glutamylcyclotransferase family and an unfolded protein response pathway regulatory gene. Its biological function and molecular regulatory mechanism, especially regarding energy metabolism underlying LUAD, remain unclear. By utilizing tissue microarray and data from The Cancer Genome Atlas and Gene Expression Omnibus, we found that CHAC1 expression was markedly higher in LUAD tissues than in non-tumor tissues, and was positively correlated with poor prognosis. Phenotypically, CHAC1 overexpression enhanced the proliferation, migration, invasion, tumor sphere formation, and glycolysis ability of LUAD cells, resulting in tumor growth both in vitro and in vivo. Mechanistically, through a shotgun mass spectrometry-based proteomic approach and high-throughput RNA sequencing, we found that CHAC1 acted as a bridge connecting UBA2 and PKM2, enhancing the SUMOylation of PKM2. The SUMOylated PKM2 then transferred from the cytoplasm to the nucleus, activating the expression of glycolysis-related genes and enhancing the Warburg effect. Lastly, E2F Transcription Factor 1 potently activated CHAC1 transcription by directly binding to the CHAC1 promoter in LUAD cells. The results of this study implied that CHAC1 regulates energy metabolism and promotes glycolysis in LUAD progression.


Subject(s)
Adenocarcinoma of Lung , Carrier Proteins , Glucose , Lung Neoplasms , Membrane Proteins , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Humans , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Thyroid Hormones/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Glucose/metabolism , Carrier Proteins/metabolism , Carrier Proteins/genetics , Animals , Disease Progression , gamma-Glutamylcyclotransferase/metabolism , gamma-Glutamylcyclotransferase/genetics , Mice , Cell Line, Tumor , Cell Proliferation , Mice, Nude , Cell Nucleus/metabolism , Male , Gene Expression Regulation, Neoplastic , Glycolysis , Female , Cell Movement , Mice, Inbred BALB C
2.
Cancer Genomics Proteomics ; 21(5): 474-484, 2024.
Article in English | MEDLINE | ID: mdl-39191500

ABSTRACT

BACKGROUND/AIM: Glioblastoma is the most frequent type of adult-onset malignant brain tumor and has a very poor prognosis. Glioblastoma stem cells have been shown to be one of the mechanisms by which glioblastoma acquires therapy resistance. Therefore, there is a need to establish novel therapeutic strategies useful for inhibiting this cell population. γ-Glutamylcyclotransferase (GGCT) is an enzyme involved in the synthesis and metabolism of glutathione, which is highly expressed in a wide range of cancer types, including glioblastoma, and inhibition of its expression has been reported to have antitumor effects on various cancer types. The aim of this study was to clarify the function of GGCT in glioblastoma stem cells. MATERIALS AND METHODS: We searched for pathways affected by GGCT overexpression in mouse embryonic fibroblasts NIH-3T3 by comprehensive gene expression analysis. Knockdown of GGCT and overexpression of desert hedgehog (DHH), a representative ligand of the pathway, were performed in glioblastoma stem cells derived from a mouse glioblastoma model. RESULTS: GGCT overexpression activated the hedgehog pathway. Knockdown of GGCT inhibited proliferation of glioblastoma stem cells and reduced expression of DHH and the downstream target GLI family zinc finger 1 (GLI1). DHH overexpression significantly restored the growth-suppressive effect of GGCT knockdown. CONCLUSION: High GGCT expression is important for expression of DHH and activation of the hedgehog pathway, which is required to maintain glioblastoma stem cell proliferation. Therefore, inhibition of GGCT function may be useful in suppressing stemness of glioblastoma stem cells accompanied by activation of the hedgehog pathway.


Subject(s)
Cell Proliferation , Down-Regulation , Glioblastoma , Hedgehog Proteins , Neoplastic Stem Cells , gamma-Glutamylcyclotransferase , Animals , Mice , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , gamma-Glutamylcyclotransferase/metabolism , gamma-Glutamylcyclotransferase/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioblastoma/pathology , Glioblastoma/genetics , Glioblastoma/metabolism , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction
3.
Int J Oncol ; 65(1)2024 07.
Article in English | MEDLINE | ID: mdl-38847236

ABSTRACT

Glutathione (GSH)­degrading enzymes are essential for starting the first stages of GSH degradation. These enzymes include extracellular γ­glutamyl transpeptidase (GGT) and intracellular GSH­specific γ­glutamylcyclotransferase 1 (ChaC1) and 2. These enzymes are essential for cellular activities, such as immune response, differentiation, proliferation, homeostasis regulation and programmed cell death. Tumor tissue frequently exhibits abnormal expression of GSH­degrading enzymes, which has a key impact on the development and spread of malignancies. The present review summarizes gene and protein structure, catalytic activity and regulation of GSH­degrading enzymes, their vital roles in tumor development (including regulation of oxidative and endoplasmic reticulum stress, control of programmed cell death, promotion of inflammation and tumorigenesis and modulation of drug resistance in tumor cells) and potential role as diagnostic biomarkers and therapeutic targets.


Subject(s)
Glutathione , Neoplasms , gamma-Glutamylcyclotransferase , gamma-Glutamyltransferase , Humans , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/enzymology , Glutathione/metabolism , gamma-Glutamylcyclotransferase/metabolism , gamma-Glutamylcyclotransferase/genetics , gamma-Glutamyltransferase/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Animals , Gene Expression Regulation, Neoplastic , Oxidative Stress , Endoplasmic Reticulum Stress
4.
Cancer Sci ; 115(9): 2961-2971, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38924236

ABSTRACT

Osteosarcoma (OS) in humans is characterized by alterations in the TP53 gene. In mice, loss of p53 triggers OS development, for which c-Myc (Myc) oncogenicity is indispensable. However, little is known about which genes are targeted by Myc to promote tumorigenesis. Here, we examined the role of γ-glutamylcyclotransferase (Ggct) which is a component enzyme of the γ-glutamyl cycle essential for glutathione homeostasis, in human and mouse OS development. We found that GGCT is a poor prognostic factor for human OS, and that deletion of Ggct suppresses p53-deficient osteosarcomagenesis in mice. Myc upregulates Ggct directly by binding to the Ggct promoter, and deletion of a Myc binding site therein by genome editing attenuated the tumorigenic potential of p53-deficient OS cells. Taken together, these results show a rationale that GGCT is widely upregulated in cancer cells and solidify its suitability as a target for anticancer drugs.


Subject(s)
Bone Neoplasms , Osteosarcoma , Proto-Oncogene Proteins c-myc , Tumor Suppressor Protein p53 , Up-Regulation , gamma-Glutamylcyclotransferase , Osteosarcoma/genetics , Osteosarcoma/pathology , Osteosarcoma/metabolism , gamma-Glutamylcyclotransferase/metabolism , gamma-Glutamylcyclotransferase/genetics , Animals , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Mice , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Promoter Regions, Genetic/genetics , Female , Male , Prognosis , Carcinogenesis/genetics
5.
J Cancer Res Clin Oncol ; 150(6): 318, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38914714

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is renowned for its formidable and lethal nature, earning it a notorious reputation among malignant tumors. Due to its challenging early diagnosis, high malignancy, and resistance to chemotherapy drugs, the treatment of pancreatic cancer has long been exceedingly difficult in the realm of oncology. γ-Glutamyl cyclotransferase (GGCT), a vital enzyme in glutathione metabolism, has been implicated in the proliferation and progression of several tumor types, while the biological function of GGCT in pancreatic ductal adenocarcinoma remains unknown. METHODS: The expression profile of GGCT was validated through western blotting, immunohistochemistry, and RT-qPCR in both pancreatic cancer tissue samples and cell lines. Functional enrichment analyses including GSVA, ssGSEA, GO, and KEGG were conducted to explore the biological role of GGCT. Additionally, CCK8, Edu, colony formation, migration, and invasion assays were employed to evaluate the impact of GGCT on the proliferation and migration abilities of pancreatic cancer cells. Furthermore, the LASSO machine learning algorithm was utilized to develop a prognostic model associated with GGCT. RESULTS: Our study revealed heightened expression of GGCT in pancreatic cancer tissues and cells, suggesting an association with poorer patient prognosis. Additionally, we explored the immunomodulatory effects of GGCT in both pan-cancer and pancreatic cancer contexts, found that GGCT may be associated with immunosuppressive regulation in various types of tumors. Specifically, in patients with high expression of GGCT in pancreatic cancer, there is a reduction in the infiltration of various immune cells, leading to poorer responsiveness to immunotherapy and worse survival rates. In vivo and in vitro assays indicate that downregulation of GGCT markedly suppresses the proliferation and metastasis of pancreatic cancer cells. Moreover, this inhibitory effect appears to be linked to the regulation of GGCT on c-Myc. A prognostic model was constructed based on genes derived from GGCT, demonstrating robust predictive ability for favorable survival prognosis and response to immunotherapy.


Subject(s)
Carcinoma, Pancreatic Ductal , Disease Progression , Immunotherapy , Pancreatic Neoplasms , gamma-Glutamylcyclotransferase , Humans , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/drug therapy , gamma-Glutamylcyclotransferase/metabolism , gamma-Glutamylcyclotransferase/genetics , Immunotherapy/methods , Cell Proliferation , Prognosis , Cell Line, Tumor , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Female , Gene Expression Regulation, Neoplastic , Male , Cell Movement , Multiomics
6.
Biochem Biophys Res Commun ; 714: 149977, 2024 06 25.
Article in English | MEDLINE | ID: mdl-38663093

ABSTRACT

Malignant tumors are characterized by a hypoxic microenvironment, and metabolic reprogramming is necessary to ensure energy production and oxidative stress resistance. Although the microenvironmental properties of tumors vary under acute and chronic hypoxia, studies on chronic hypoxia-induced metabolic changes are limited. In the present study, we performed a comprehensive metabolic analysis in a chronic hypoxia model using colorectal cancer (CRC) organoids, and identified an amino acid supply system through the γ-glutamyl cycle, a glutathione recycling pathway. We analyzed the metabolic changes caused by hypoxia over time and observed that chronic hypoxia resulted in an increase in 5-oxoproline and a decrease in oxidized glutathione (GSSG) compared to acute hypoxia. These findings suggest that chronic hypoxia induces metabolic changes in the γ-glutamyl cycle. Moreover, inhibition of the γ-glutamyl cycle via γ-glutamyl cyclotransferase (GGCT) and γ-glutamyl transferase 1 (GGT1) knockdown significantly reversed chronic hypoxia-induced upregulation of 5-oxoproline and several amino acids. Notably, GGT1 knockdown downregulated the intracellular levels of γ-glutamyl amino acids. Conclusively, these results indicate that the γ-glutamyl cycle serves as an amino acid supply system in CRC under chronic hypoxia, which provides fresh insight into cancer metabolism under chronic hypoxia.


Subject(s)
Amino Acids , Colorectal Neoplasms , Organoids , gamma-Glutamyltransferase , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Organoids/metabolism , Organoids/pathology , gamma-Glutamyltransferase/metabolism , Amino Acids/metabolism , Cell Hypoxia , Tumor Microenvironment , Glutathione/metabolism , Hypoxia/metabolism , Tumor Hypoxia , gamma-Glutamylcyclotransferase/metabolism , gamma-Glutamylcyclotransferase/genetics
7.
Carcinogenesis ; 45(4): 247-261, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38190483

ABSTRACT

We previously reported that RNF148 was involved in the ubiquitination-mediated degradation of CHAC2. However, its molecular mechanism was not determined. In this study, we investigated the role and mechanism of RNF148 in the progression of colorectal cancer (CRC), especially in the process of ubiquitination-mediated degradation of CHAC2. Our results revealed that RNF148 was upregulated in most CRC tissues, and its expression significantly correlated with the 3-year overall survival rate and most clinicopathological parameters of CRC patients. Furthermore, RNF148 served as an independent prognostic biomarker of CRC and promoted CRC cell proliferation and migration while inhibiting cell apoptosis and sensitivity to 5-FU. Mechanistically, RNF148 used its protease-associated domain to bind to the CHAC domain of CHAC2 and target it for degradation. In addition, we identified two phosphorylation and three ubiquitination residues of CHAC2 and identified Y118 and K102 as the critical phosphorylation and ubiquitination residues, respectively. We also identified CHAC2's and RNF148's interacting proteins and discovered their potential interaction network. In conclusion, our current study unveiled the role of RNF148 in CRC and the mechanism of RNF148 in the ubiquitination-mediated degradation of CHAC2, which shed light on providing potential prognostic biomarkers and molecular targets for CRC patients.


Subject(s)
Colorectal Neoplasms , Ubiquitin-Protein Ligases , gamma-Glutamylcyclotransferase , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Oncogenes , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , gamma-Glutamylcyclotransferase/metabolism
8.
Mol Med Rep ; 28(5)2023 Nov.
Article in English | MEDLINE | ID: mdl-37772365

ABSTRACT

Glutathione­specific γ­glutamylcyclotransferase 1 (CHAC1), is an unfolded protein response­induced gene. Although it has been previously reported that CHAC1 transcription is regulated by activating transcription factor (ATF) 4, ATF3 and CCAAT/enhancer­binding protein ß (C/EBPß), the signaling pathways that regulate CHAC1 are largely unknown. It was revealed that 3­(5'­hydroxymethyl­2'­furyl)­1­benzylindazole (YC­1; PubChem ID: 5712), a nitric oxide­independent activator of soluble guanylyl cyclase (sGC), increases CHAC1 levels in cultured human kidney proximal tubular cells (HK­2). Therefore, in the present study, the signaling pathways that induce CHAC1 by YC­1 were investigated in HK­2 cells. YC­1 induced CHAC1 expression in a dose­ and time­dependent manner. KT5823, an inhibitor of cGMP­dependent protein kinase (PKG), partially inhibited CHAC1 upregulation, indicating that the sGC­cGMP­PKG pathway participates in CHAC1 regulation. These results also suggested that other signaling pathways are involved in the regulation of CHAC1. Since antibody array analysis showed the activation of p38, mTOR and Akt, the involvement of these factors was further investigated. Although LY294002 and KU0063794 (inhibitors of Akt and mTOR, respectively) inhibited YC­1­induced CHAC1 expression, SB203580 (an inhibitor of p38) did not. These results indicated that CHAC1 is regulated by the Akt­mTOR pathway. In addition, YC­1 induced endoplasmic reticulum (ER) stress, a regulator of CHAC1 induction. These findings suggested that CHAC1 is regulated by YC­1 through the sGC­cGMP­PKG, Akt­mTOR and ER stress pathways. The present study demonstrated that CHAC1 induction reduced the intracellular glutathione concentration, indicating that CHAC1 plays an important role in intracellular redox homeostasis in tubular cells.


Subject(s)
Proto-Oncogene Proteins c-akt , gamma-Glutamylcyclotransferase , Humans , gamma-Glutamylcyclotransferase/genetics , gamma-Glutamylcyclotransferase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Glutathione/metabolism , Endoplasmic Reticulum Stress/genetics
9.
PLoS One ; 18(4): e0283806, 2023.
Article in English | MEDLINE | ID: mdl-37014882

ABSTRACT

Muscle wasting is one of the main characteristics of cachexia associated with cancer and other chronic diseases and is often exacerbated by antineoplastic agents. Increased oxidative stress is associated with muscle wasting, along with depletion of glutathione, the most abundant endogenous antioxidant. Therefore, boosting endogenous glutathione has been proposed as a therapeutic strategy to prevent muscle wasting. Here, we tested this hypothesis by inactivating CHAC1, an intracellular glutathione degradation enzyme. We found CHAC1 expression is increased under multiple muscle wasting conditions in animal models, including fasting, cancer cachexia, and chemotherapy. The elevation of muscle Chac1 expression is associated with reduced glutathione level. CHAC1 inhibition via CRSPR/Cas9 mediated knock-in of an enzyme inactivating mutation demonstrates a novel strategy to preserve muscle glutathione levels under wasting conditions but fails to prevent muscle wasting in mice. These results suggest that preserving intracellular glutathione level alone may not be sufficient to prevent cancer or chemotherapy induced muscle wasting.


Subject(s)
Cachexia , Neoplasms , gamma-Glutamylcyclotransferase , Animals , Mice , Cachexia/prevention & control , Cachexia/metabolism , Glutathione/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/pathology , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/metabolism , gamma-Glutamylcyclotransferase/metabolism
10.
Neoplasma ; 70(6): 777-786, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38247333

ABSTRACT

ChaC glutathione-specific γ-glutamylcyclotransferase 1 (CHAC1) is involved in intracellular glutathione depletion, ferroptosis, and tumorigenesis. The functional role of CHAC1 expression in thyroid carcinoma has not yet been established. The present study aimed to investigate the impact and mechanisms of CHAC1 on ferroptosis and radiation sensitivity in thyroid carcinoma. CHAC1 expression was examined in tumor tissue specimens and microarrays and thyroid carcinoma cell lines. CHAC1 was silenced or overexpressed by lentivirus transfection in thyroid carcinoma cells. Cell viability and lipid ROS levels were evaluated by Cell Counting Kit-8 and flow cytometry, respectively. The effect of CHAC1 on tumor growth in vivo was also measured. Ferroptosis-related proteins were measured by western blotting. CHAC1 expression was decreased in patients with thyroid carcinoma, and overexpression of CHAC1 suppressed cell viability of BCPAP cells and tumor growth in xenografted nude mice. Exposure to Ferrostatin-1, a ferroptosis inhibitor, significantly attenuated the inhibitory effects of CHAC1 overexpression on cell viability. In CHAC1-overexpressing BCPAP cells, ferroptosis was induced as indicated by increased lipid ROS production and PTGS2 expression. Knocking down of CHAC1 in K1 cells significantly induced cell viability, reduced lipid ROS production and PTGS2 expression, and enhanced GPX4 expression. Such effects were attenuated by RSL3, a ferroptosis inducer. Furthermore, we showed that CHAC1 overexpression enhanced radiation sensitivity in BCPAP cells as indicated by decreased cell viability, while CHAC1 knockdown had reversed effects in K1 cells as indicated by increased cell viability. Taken together, CHAC1 overexpression promoted ferroptosis and enhanced radiation sensitivity in thyroid carcinoma.


Subject(s)
Ferroptosis , Thyroid Neoplasms , gamma-Glutamylcyclotransferase , Animals , Humans , Mice , Cyclooxygenase 2 , Ferroptosis/genetics , Glutathione , Lipids , Mice, Nude , Reactive Oxygen Species , Thyroid Neoplasms/genetics , Thyroid Neoplasms/radiotherapy , gamma-Glutamylcyclotransferase/genetics , gamma-Glutamylcyclotransferase/metabolism
11.
Neurosci Res ; 184: 62-69, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35988816

ABSTRACT

The PARK7 gene, which encodes DJ-1 protein, is the causative gene of autosomal recessive early-onset Parkinson's disease. DJ-1 has many biological functions, including regulating glutathione (GSH) levels. However, the molecular mechanism by which DJ-1 regulates GSH levels in astrocytes remains unclear. With high throughput sequencing, we discovered that DJ-1 knockout could significantly upregulate the expression of ChaC glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1). We demonstrate that DJ-1 can bind with the basic leucine zipper domain of activating transcription factor 3 (ATF3) through bimolecular fluorescence complementation. Besides, DJ-1 inhibits ATF3 binding to the CHAC1 promoter and downregulates the expression of CHAC1 to reduce GSH degradation. Our research suggests that the loss of DJ-1 in astrocytes promotes the degradation of GSH, leading neurons more vulnerable to oxidative damage. It provides a theoretical basis for developing drugs targeting DJ-1 and GSH in the brain.


Subject(s)
Astrocytes , gamma-Glutamylcyclotransferase , Activating Transcription Factor 3/metabolism , Astrocytes/metabolism , Glutathione/metabolism , Protein Deglycase DJ-1/genetics , gamma-Glutamylcyclotransferase/genetics , gamma-Glutamylcyclotransferase/metabolism
12.
Anticancer Res ; 42(3): 1221-1227, 2022 03.
Article in English | MEDLINE | ID: mdl-35220212

ABSTRACT

BACKGROUND/AIM: γ-Glutamyl cyclotransferase (GGCT) is up-regulated in various cancer types, including lung cancer. In this study, we evaluated efficacy of gapmer-type antisense oligonucleotides (ASOs) targeting GGCT in an A549 lung cancer xenograft mouse model and studied their mechanisms of action. MATERIALS AND METHODS: GGCT was inhibited using GGCT-ASOs and cell proliferation was evaluated by dye exclusion test. Western blot analysis was conducted to measure expression of GGCT, p21, p16 and p27, phosphorylation of AMP-activated protein kinase, and caspase activation in A549 cells. Induction of apoptosis and up-regulation of reactive oxygen species were assessed by flow cytometry using annexin V staining and 2',7'-dichlorodihydrofluorescein diacetate dye, respectively. RESULTS: GGCT-ASOs suppressed GGCT expression in A549 cells, inhibited proliferation, and induced apoptosis with activation of caspases. GGCT-ASOs also increased expression of cell-cycle regulating proteins, phospho-AMPK and ROS levels. Systemic administration of GGCT-ASOs to animals bearing A549 lung cancer xenografts showed significant antitumor effects without evident toxicity. CONCLUSION: GGCT-ASOs appear to be promising as novel cancer therapeutic agents.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Lung Neoplasms/drug therapy , Oligonucleotides, Antisense/pharmacology , gamma-Glutamylcyclotransferase/metabolism , A549 Cells , Animals , Apoptosis , Caspases/metabolism , Cell Cycle Proteins/metabolism , Cycloheximide/analogs & derivatives , Cycloheximide/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice, SCID , Signal Transduction , Tumor Burden , Xenograft Model Antitumor Assays , gamma-Glutamylcyclotransferase/genetics
13.
Endocrinology ; 163(4)2022 04 01.
Article in English | MEDLINE | ID: mdl-35213720

ABSTRACT

Papillary thyroid cancer (PTC) remains the most common endocrine malignancy, despite marked achieves in recent decades, and the mechanisms underlying the pathogenesis and progression for PTC are incompletely elucidated. Accumulating evidence show that γ-glutamylcyclotransferase (GGCT), an enzyme participating in glutathione homeostasis and is elevated in multiple types of tumors, represents an attractive therapeutic target. Using bioinformatics, immunohistochemistry, qRT-PCR, and Western blot assays, we found that GGCT expression was upregulated in PTC and correlated with more aggressive clinicopathological characteristics and worse prognosis. GGCT knockdown inhibited the growth and metastasis ability of PTC cells both in vitro and in vivo and reduced the expression of mesenchymal markers (N-cadherin, CD44, MMP2, and MMP9) while increasing epithelial marker (E-cadherin) in PTC cells. We confirmed binding of microRNA-205-5p (miR-205-5p) on the 3'-UTR regions of GGCT by dual-luciferase reporter assay and RNA-RNA pull-down assay. Delivery of miR-205-5p reversed the pro-malignant capacity of GGCT both in vitro and in vivo. Lastly, we found that GGCT interacted with and stabilized CD44 in PTC cells by co-immunoprecipitation and immunohistochemistry assays. Our findings illustrate a novel signaling pathway, miR-205-5p/GGCT/CD44, that involves in the carcinogenesis and progression of PTC. Development of miR-205-mimics or GGCT inhibitors as potential therapeutics for PTC may have remarkable applications.


Subject(s)
MicroRNAs , Thyroid Neoplasms , 3' Untranslated Regions , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Thyroid Cancer, Papillary/metabolism , Thyroid Neoplasms/pathology , gamma-Glutamylcyclotransferase/genetics , gamma-Glutamylcyclotransferase/metabolism
14.
Mol Pharmacol ; 101(2): 78-86, 2022 02.
Article in English | MEDLINE | ID: mdl-34862308

ABSTRACT

Prohibitin-2 (PHB2) is a scaffold protein that has pleiotropic functions, which include interacting with γ-glutamylcyclotransferase (GGCT) in the cytoplasm and repressing the transcriptional activities of the p21Waf1/Cip (p21) gene in the nucleus. The cytotoxic drug fluorizoline binds to PHB1/2 and exerts antiproliferative actions on cancer cells. However, the precise mechanism underlying the antiproliferative effects of fluorizoline is not fully elucidated. In the present study, we first show that fluorizoline induces p21 expression in several human cancer cell lines, including MCF7 breast cancer cells. Treatment of MCF7 cells with fluorizoline suppressed proliferation and prevented cells from entering into the DNA synthesis phase. Knockdown of p21 rescued the suppressed proliferation, indicating that fluorizoline inhibited MCF7 cell growth via the induction of p21. Overexpression of PHB2 in MCF7 cells prevented the induction of p21 expression by fluorizoline and restored the antiproliferative effects and blockade of cell cycle progression. Moreover, treatment of MCF7 cells with fluorizoline inhibited the interaction between endogenous PHB2 and GGCT proteins and reduced the level of nuclear localization of PHB2 proteins. These results indicate that targeting PHB2 with fluorizoline induces the expression of p21 and consequently blocks proliferation of cancer cells. SIGNIFICANCE STATEMENT: This study shows that fluorizoline may be a promising novel anticancer drug candidate that induces p21 expression and blocks cell-cycle progression in human cancer cell lines. In addition, we show that fluorizoline inhibits the interaction between PHB2 and GGCT and reduces the nuclear localization of PHB2 proteins.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Gene Expression Regulation, Neoplastic/physiology , Prohibitins/metabolism , gamma-Glutamylcyclotransferase/metabolism , Antineoplastic Agents/chemical synthesis , Breast Neoplasms/genetics , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Prohibitins/antagonists & inhibitors , gamma-Glutamylcyclotransferase/antagonists & inhibitors
15.
Molecules ; 26(24)2021 Dec 19.
Article in English | MEDLINE | ID: mdl-34946766

ABSTRACT

The development of the field of nanotechnology has revolutionized various aspects in the fields of modern sciences. Nano-medicine is one of the primary fields for the application of nanotechnology techniques. The current study sheds light on the reno-protective impacts of gold nano-particles; nanogold (AuNPs) against 5-flurouracil (5-FU)-induced renal toxicity. Indeed, the use of 5-FU has been associated with kidney injury which greatly curbs its therapeutic application. In the current study, 5-FU injection was associated with a significant escalation in the indices of renal injury, i.e., creatinine and urea. Alongside this, histopathological and ultra-histopathological changes confirmed the onset of renal injury. Both gene and/or protein expression of nuclear factor erythroid 2-related factor 2 (Nrf-2) and downstream antioxidant enzymes revealed consistent paralleled anomalies. AuNPs administration induced a significant renal protection on functional, biochemical, and structural levels. Renal expression of the major sensor of the cellular oxidative status Nrf-2 escalated with a paralleled reduction in the renal expression of the other contributor to this axis, known as Kelch-like ECH-associated protein 1 (Keap-1). On the level of the effector downstream targets, heme oxygenase 1 (HO-1) and gamma-glutamylcysteine synthetase (γ-GCS) AuNPs significantly restored their gene and protein expression. Additionally, combination of AuNPs with 5-FU showed better cytotoxic effect on MCF-7 cells compared to monotreatments. Thus, it can be inferred that AuNPs conferred reno-protective impact against 5-FU with an evident modulatory impact on Nrf-2/Keap-1 and its downstream effectors, HO-1 and γ-GCS, suggesting its potential use in 5-FU regimens to improve its therapeutic outcomes and minimize its underlying nephrotoxicity.


Subject(s)
Fluorouracil/antagonists & inhibitors , Gold/pharmacology , Kidney/drug effects , Metal Nanoparticles/chemistry , Animals , Disease Models, Animal , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Gold/administration & dosage , Gold/chemistry , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/metabolism , Injections, Intraperitoneal , Kidney/injuries , Kidney/pathology , Metal Nanoparticles/administration & dosage , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Nanotechnology , Particle Size , Rats , Rats, Sprague-Dawley , gamma-Glutamylcyclotransferase/antagonists & inhibitors , gamma-Glutamylcyclotransferase/metabolism
16.
Zhonghua Zhong Liu Za Zhi ; 43(11): 1183-1187, 2021 Nov 23.
Article in Chinese | MEDLINE | ID: mdl-34794221

ABSTRACT

Objective: To evaluate the expression and clinical significance of γ-glutamylcyclotransferase (GGCT) in patients with bladder urothelial cell carcinoma. Methods: Immunohistochemical staining for GGCT were performed on tissue sections of 86 patients with bladder urothelial cell carcinoma and 10 normal controls, and the correlations between GGCT and clinicopathological characteristics and the prognosis were analyzed. Results: The positive rate of the expression of GGCT in 86 cases of bladder urothelial cell carcinoma was 61.6% (53/86). GGCT protein was located mainly in cancer cell cytoplasm, and it can be seen in the nucleus of the tumor cells in some cases. The level of GGCT expression was positively related to pathological classification (P<0.001), stage (P=0.020), and tumor size (P=0.025). Immunohistochemical semiquantitative analysis showed that the expression of GGCT in patients with T1 stage of non-muscle invasion bladder urothelial cell carcinoma was significantly higher than that with Ta stage (P=0.034). Kaplan-Meier analysis showed that the expression of GGCT was correlated with the recurrence-free survival in patients with non-muscle invasive bladder cancer, the recurrence-free survival rate was lower in the GGCT positive group (P=0.029). Multivariate COX regression analysis showed that the pathological stage (OR=5.029, P=0.009) and the number of tumors (OR=3.320, P=0.024)were the independent risk factors for recurrence-free survival in patients with early urothelial cell carcinoma of the bladder. Conclusions: The expression of GGCT is significantly increased in bladder urothelial cell carcinoma and is related to the malignant biological behavior and progression of tumor. Patients with GGCT positive early bladder tumor are inclined to recur.


Subject(s)
Carcinoma, Transitional Cell , Urinary Bladder Neoplasms , gamma-Glutamylcyclotransferase/metabolism , Biomarkers, Tumor , Humans , Neoplasm Recurrence, Local , Prognosis
17.
Neurosci Lett ; 765: 136283, 2021 11 20.
Article in English | MEDLINE | ID: mdl-34624395

ABSTRACT

Intracerebral hemorrhage (ICH) is a leading medical problem and has no effective treatment approach up until now. The transcription factor androgen receptor (AR) has been indicated in the cerebrovascular function recently. However, its participation in ICH remains unclear. The present study aims to expound the regulation of AR in microglia/macrophage phenotypes and the secondary brain injury in a rat model with ICH, and to discuss the involved pathway. Following the induction of ICH in rats, we found that ICH led to increased mNSS score, enhanced microglial activity, and promoted levels of inflammatory factors and apoptosis of brain cells. Using microarray analysis, AR was found to be significantly overexpressed in ICH rat brain tissues. AR repressed the transcription of Jumonji d3 (JMJD3, histone 3 demethylase). JMJD3 inhibited the methylation of Botch and promoted the activity of Notch1. JMJD3 hampered microglial activity and ameliorated secondary brain injury in rats, whereas upregulation of AR or downregulation of Botch reversed the protective effects of JMJD3. In conclusion, we found that AR promoted microglial activation and secondary brain injury via transcriptionally repressing JMJD3 and mediating the subsequent Botch/Notch1 pathway, which may provide novel insights into therapeutic options for the treatment of ICH.


Subject(s)
Intracranial Hemorrhages/metabolism , Macrophage Activation/physiology , Microglia/metabolism , Receptors, Androgen/metabolism , Signal Transduction/physiology , Animals , Jumonji Domain-Containing Histone Demethylases/metabolism , Macrophages/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptor, Notch1/metabolism , gamma-Glutamylcyclotransferase/metabolism
18.
Oncol Rep ; 46(5)2021 Nov.
Article in English | MEDLINE | ID: mdl-34558645

ABSTRACT

Dihydroartemisinin (DHA), an artemisinin derivate, has been investigated as a potential antitumor drug in primary liver cancer (PLC). Ferroptosis is a form of iron­dependent cell death that can be driven by lipid peroxidation inducers. The present study aimed to determine whether and how DHA could promote the death of PLC cells by inducing ferroptosis. In total, four PLC cell lines with different p53 statuses, including Hep3B (p53 null), Huh7 (p53 mutant), PLC/PRF/5 (p53 mutant) and HepG2 (p53 wild­type), were treated with various concentrations of DHA. The effects of DHA on all three branches of the unfolded protein response (UPR) were evaluated. To deactivate the UPRs, small interfering RNA was used to knockdown the expression of activating transcription factor (ATF)4, X­box binding protein 1 (XBP1) or ATF6 in PLC cells. The effect of DHA on the promoter activity of Chac glutathione specific γ­glutamylcyclotransferase 1 (CHAC1) was evaluated using a dual luciferase reporter assay. The results revealed that DHA­induced death in PLC cells was irrelevant of the p53 status. PLC cells exposed to DHA displayed classic features of ferroptosis, such as increased lipid reactive oxygen species and malondialdehyde levels, an iron overload, and decreased activity or expression of glutathione (GSH), glutathione peroxidase 4, solute carrier family (SLC) 7 member 11 and SLC family 3 member 2. The antitumor effects of DHA in PLC cells were significantly weakened by two typical ferroptosis inhibitors, ferrostatin­1 and deferoxamine mesylate salt, whereas the antitumor effects were augmented following iron overload. Furthermore, DHA activated all three branches of the UPR (eukaryotic translation initiation factor 2 α kinase 3/eukaryotic translation initiation factor 2A/ATF4, inositol­requiring transmembrane kinase/endoribonuclease 1α/XBP1 and ATF6 branches) in vitro. Notably, DHA­induced ferroptosis was significantly attenuated following the knockdown of ATF4, XBP1 or ATF6 expression. In addition, the promoter activity of CHAC1, a gene capable of degrading GSH, was enhanced by DHA, but weakened when the aforementioned three UPR transcription factors were knocked down. In conclusion, the findings of the present study suggested that DHA may effectively induce ferroptosis in PLC cells through the activation of anti­survival UPRs and the upregulation of CHAC1 expression.


Subject(s)
Artemisinins/pharmacology , Ferroptosis/drug effects , Liver Neoplasms/drug therapy , Unfolded Protein Response/drug effects , gamma-Glutamylcyclotransferase/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , RNA, Small Interfering/metabolism , Up-Regulation
19.
Cell Death Dis ; 12(9): 834, 2021 09 04.
Article in English | MEDLINE | ID: mdl-34482365

ABSTRACT

Ferroptosis is a nonapoptotic form of programmed cell death triggered by the accumulation of reactive oxygen species (ROS) depended on iron overload. Although most investigations focus on the relationship between ferroptosis and cancer, neurodegenerative diseases, and ischemia/reperfusion injury, research on ferroptosis induced by immune-related inflammatory diseases, especially sepsis, is scarce. Sestrin2 (Sesn2), a highly evolutionary and stress-responsive protein, is critically involved in defense against oxidative stress challenges. Upregulated expression of Sesn2 has been observed in preliminary experiments to have an antioxidative function in the context of an inflammatory response. Nevertheless, the underlying function of Sesn2 in inflammation-mediated ferroptosis in the immune system remains uncertain. The current study aimed to demonstrate the protective effect of Sesn2 on ferroptosis and even correlations with ferroptosis and the functions of ferroptotic-dendritic cells (DCs) stimulated with lipopolysaccharide (LPS). The mechanism underlying DCs protection from LPS-induced ferroptosis by Sesn2 was further explored in this study. We found that the immune response of DCs assessed by co-stimulatory phenotypes was gradually enhanced at the peak time of 12 h upon 1 µg/ml LPS stimulation while ferroptosis in DCs treated with LPS at 24 h was significantly detected. LPS-induced ferroptosis showed a suppressive impact on DCs in phenotypic maturation, which was conversely relieved by the ferroptotic inhibitor. Compared with wild-type (WT) mice, DCs in genetic defective mice of Sesn2 (Sesn2-/-) exhibited exacerbated ferroptosis. Furthermore, the protective effect of Sesn2 on ferroptosis was noticed to be associated with the ATF4-CHOP-CHAC1 pathway, eventually exacerbating ferroptosis by degrading of glutathione. These results indicate that Sesn2 can suppress the ferroptosis of DCs in sepsis by downregulating the ATF4-CHOP-CHAC1 signaling pathway, and it might play an antioxidative role.


Subject(s)
Dendritic Cells/metabolism , Ferroptosis , Peroxidases/metabolism , Protective Agents/metabolism , Sepsis/metabolism , Sepsis/pathology , Activating Transcription Factor 4 , Animals , Cecum/pathology , Cell Differentiation , Dendritic Cells/immunology , Dendritic Cells/ultrastructure , Down-Regulation , Immunity , Ligation , Lipopolysaccharides , Male , Mice, Inbred C57BL , Phenotype , Punctures , Sepsis/immunology , Signal Transduction , Spleen/cytology , Transcription Factor CHOP/metabolism , gamma-Glutamylcyclotransferase/metabolism
20.
Br J Haematol ; 195(2): 267-275, 2021 10.
Article in English | MEDLINE | ID: mdl-34409610

ABSTRACT

The expression of GGCT (γ-glutamyl cyclotransferase) is upregulated in various human cancers. γ-glutamyl cyclotransferase enzyme activity was originally purified from human red blood cells (RBCs), but the physiological function of GGCT in RBCs is still not clear. Here we reported that Ggct deletion in mice leads to splenomegaly and progressive anaemia phenotypes, due to elevated oxidative damage and the shortened life span of Ggct-/- RBCs. Ggct-/- RBCs have increased reactive oxygen species (ROS), and are more sensitive to H2 O2 -induced damage compared to control RBCs. Glutathione (GSH) and GSH synthesis precursor l-cysteine are decreased in Ggct-/- RBCs. Our study suggests a critical function of Ggct in RBC redox balance and life span maintenance through regulating GSH metabolism.


Subject(s)
Erythrocytes/metabolism , Reactive Oxygen Species/metabolism , gamma-Glutamylcyclotransferase/metabolism , Anemia/genetics , Animals , Antioxidants/metabolism , Cysteine/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Erythropoietin/metabolism , Female , Gene Deletion , Glutathione/metabolism , Male , Metabolomics/methods , Mice , Models, Animal , Phenotype , Splenomegaly/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL