Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 757
Filter
1.
Apoptosis ; 29(5-6): 898-919, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38411862

ABSTRACT

The cytosolic sulfotransferases (SULTs) are phase II conjugating enzymes, which are widely expressed in the liver and mainly mediate the sulfation of numerous xenobiotics and endogenous compounds. However, the role of various SULTs genes has not been reported in hepatocellular carcinoma (HCC). This study aims to analyze the expression and potential functional roles of SULTs genes in HCC and to identify the role of SULT2A1 in HCC stemness as well as the possible mechanism. We found that all of the 12 SULTs genes were differentially expressed in HCC. Moreover, clinicopathological features and survival rates were also investigated. Multivariate regression analysis showed that SULT2A1 and SULT1C2 could be used as independent prognostic factors in HCC. SULT1C4, SULT1E1, and SULT2A1 were significantly associated with immune infiltration. SULT2A1 deficiency in HCC promoted chemotherapy resistance and stemness maintenance. Mechanistically, silencing of SULT2A1 activated the AKT signaling pathway, on the one hand, promoted the expression of downstream stemness gene c-Myc, on the other hand, facilitated the NRF2 expression to reduce the accumulation of ROS, and jointly increased HCC stemness. Moreover, knockdown NR1I3 was involved in the transcriptional regulation of SULT2A1 in stemness maintenance. In addition, SULT2A1 knockdown HCC cells promoted the proliferation and activation of hepatic stellate cells (HSCs), thereby exerting a potential stroma remodeling effect. Our study revealed the expression and role of SULTs genes in HCC and identified the contribution of SULT2A1 to the initiation and progression of HCC.


Subject(s)
Arylsulfotransferase , Carcinoma, Hepatocellular , Liver Neoplasms , Neoplastic Stem Cells , Sulfotransferases , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/enzymology , Sulfotransferases/genetics , Sulfotransferases/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/enzymology , Male , Gene Expression Regulation, Neoplastic , Female , Cell Line, Tumor , Middle Aged , Animals , Mice , Cell Proliferation/genetics , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Drug Resistance, Neoplasm/genetics
2.
Gastroenterology ; 166(5): 886-901.e7, 2024 05.
Article in English | MEDLINE | ID: mdl-38096955

ABSTRACT

BACKGROUND & AIMS: Metabolic and transcriptional programs respond to extracellular matrix-derived cues in complex environments, such as the tumor microenvironment. Here, we demonstrate how lysyl oxidase (LOX), a known factor in collagen crosslinking, contributes to the development and progression of cholangiocarcinoma (CCA). METHODS: Transcriptomes of 209 human CCA tumors, 143 surrounding tissues, and single-cell data from 30 patients were analyzed. The recombinant protein and a small molecule inhibitor of the LOX activity were used on primary patient-derived CCA cultures to establish the role of LOX in migration, proliferation, colony formation, metabolic fitness, and the LOX interactome. The oncogenic role of LOX was further investigated by RNAscope and in vivo using the AKT/NICD genetically engineered murine CCA model. RESULTS: We traced LOX expression to hepatic stellate cells and specifically hepatic stellate cell-derived inflammatory cancer-associated fibroblasts and found that cancer-associated fibroblast-driven LOX increases oxidative phosphorylation and metabolic fitness of CCA, and regulates mitochondrial function through transcription factor A, mitochondrial. Inhibiting LOX activity in vivo impedes CCA development and progression. Our work highlights that LOX alters tumor microenvironment-directed transcriptional reprogramming of CCA cells by facilitating the expression of the oxidative phosphorylation pathway and by increasing stemness and mobility. CONCLUSIONS: Increased LOX is driven by stromal inflammatory cancer-associated fibroblasts and correlates with diminished survival of patients with CCA. Modulating the LOX activity can serve as a novel tumor microenvironment-directed therapeutic strategy in bile duct pathologies.


Subject(s)
Bile Duct Neoplasms , Cancer-Associated Fibroblasts , Cholangiocarcinoma , Hepatic Stellate Cells , Protein-Lysine 6-Oxidase , Tumor Microenvironment , Humans , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/enzymology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/enzymology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cholangiocarcinoma/pathology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/genetics , Cholangiocarcinoma/enzymology , Gene Expression Regulation, Neoplastic , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatic Stellate Cells/enzymology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/enzymology , Oxidative Phosphorylation , Protein-Lysine 6-Oxidase/metabolism , Protein-Lysine 6-Oxidase/genetics , Signal Transduction
3.
Life Sci ; 320: 121541, 2023 May 01.
Article in English | MEDLINE | ID: mdl-36870386

ABSTRACT

Modern cancer chemotherapy originated in the 1940s, and since then, many chemotherapeutic agents have been developed. However, most of these agents show limited response in patients due to innate and acquired resistance to therapy, which leads to the development of multi-drug resistance to different treatment modalities, leading to cancer recurrence and, eventually, patient death. One of the crucial players in inducing chemotherapy resistance is the aldehyde dehydrogenase (ALDH) enzyme. ALDH is overexpressed in chemotherapy-resistant cancer cells, which detoxifies the generated toxic aldehydes from chemotherapy, preventing the formation of reactive oxygen species and, thus, inhibiting the induction of oxidative stress and the stimulation of DNA damage and cell death. This review discusses the mechanisms of chemotherapy resistance in cancer cells promoted by ALDH. In addition, we provide detailed insight into the role of ALDH in cancer stemness, metastasis, metabolism, and cell death. Several studies investigated targeting ALDH in combination with other treatments as a potential therapeutic regimen to overcome resistance. We also highlight novel approaches in ALDH inhibition, including the potential synergistic employment of ALDH inhibitors in combination with chemotherapy or immunotherapy against different cancers, including head and neck, colorectal, breast, lung, and liver.


Subject(s)
Aldehyde Dehydrogenase , Drug Resistance, Neoplasm , Immunotherapy , Neoplasms , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/metabolism , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/radiotherapy , Drug Resistance, Neoplasm/drug effects , Humans , Animals , Neoplasm Metastasis , Cell Death , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects
4.
Proc Natl Acad Sci U S A ; 119(29): e2110348119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858297

ABSTRACT

The dichotomous behavior of superoxide dismutase-2 (SOD2) in cancer biology has long been acknowledged and more recently linked to different posttranslational forms of the enzyme. However, a distinctive activity underlying its tumor-promoting function is yet to be described. Here, we report that acetylation, one of such posttranslational modifications (PTMs), increases SOD2 affinity for iron, effectively changing the biochemical function of this enzyme from that of an antioxidant to a demethylase. Acetylated, iron-bound SOD2 localizes to the nucleus, promoting stem cell gene expression via removal of suppressive epigenetic marks such as H3K9me3 and H3K927me3. Particularly, H3K9me3 was specifically removed from regulatory regions upstream of Nanog and Oct-4, two pluripotency factors involved in cancer stem cell reprogramming. Phenotypically, cells expressing nucleus-targeted SOD2 (NLS-SOD2) have increased clonogenicity and metastatic potential. FeSOD2 operating as H3 demethylase requires H2O2 as substrate, which unlike cofactors of canonical demethylases (i.e., oxygen and 2-oxoglutarate), is more abundant in tumor cells than in normal tissue. Therefore, our results indicate that FeSOD2 is a demethylase with unique activities and functions in the promotion of cancer evolution toward metastatic phenotypes.


Subject(s)
Breast Neoplasms , Cell Nucleus , Histone Demethylases , Iron , Neoplastic Stem Cells , Superoxide Dismutase , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Nucleus/enzymology , Histone Demethylases/genetics , Histone Demethylases/metabolism , Hydrogen Peroxide/metabolism , Iron/metabolism , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Protein Processing, Post-Translational , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
5.
Anticancer Res ; 42(3): 1199-1205, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35220209

ABSTRACT

BACKGROUND/AIM: Aldehyde dehydrogenase (ALDH) 1A1 is a well-known marker for cancer stem cells (CSCs), characterized by self-renewal capacity and multidrug resistance in breast cancer. We developed a near-infrared turn-on fluorescence probe for ALDH1A1, C5S-A, which is suitable for observing and analyzing viable cells. Here, we demonstrated the utility of C5S-A in CSC research using breast cancer cell lines. MATERIALS AND METHODS: To evaluate concordance between C5S-A and conventional stem cell markers, breast cancer cells sorted for ALDEFLUOR-positive cells and for CD44+/CD24- cell populations were stained with C5S-A. Tumorigenicity of C5S-A-positive cells was examined by mammosphere formation assay and subcutaneous transplantation to immunodeficient mice. Additionally, to determine how long fluorescence from a single staining remained observable, we cultured breast cancer cells for 5 days after C5S-A staining. We then evaluated whether C5S-A-positive cells possessed resistance to cytotoxic drugs by chronological imaging. RESULTS: C5S-A staining showed good concordance with conventional breast CSC markers, and good utility for research into CSC characteristics in breast cancer cell lines, including tumorigenesis. Additionally, C5S-A was observable for more than 3 days with a single staining. Using this property, we then confirmed that C5S-A-positive cells possessed resistance to cytotoxic drugs, which is one of the characteristics of CSCs. CONCLUSION: We showed that C5S-A is suitable for CSC research using breast cancer cell lines, and confirmed its utility in observing cells over time.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/enzymology , Fluorescent Dyes , Neoplastic Stem Cells/enzymology , Retinal Dehydrogenase/metabolism , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Separation , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , Flow Cytometry , Humans , Mice, Inbred NOD , Mice, SCID , Microscopy, Fluorescence , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Paclitaxel/pharmacology , Time Factors
6.
Int J Mol Sci ; 23(3)2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35163585

ABSTRACT

BACKGROUND: The treatment of non-small-cell lung cancer (NSCLC) involves platinum-based chemotherapy. It is typically accompanied by chemoresistance resulting from antioxidant properties conferred by cancer stem cells (CSCs). Human epidermal growth factor receptor 2 (HER2) enhances CSCs and antioxidant properties in cancers, including NSCLC. METHODS: Here, we elucidated the role of histamine N-methyltransferase (HNMT), a histamine metabolism enzyme significantly upregulated in NSCLC and coexpressed with HER2. HNMT expression in lung cancer tissues was determined using quantitative reverse transcription PCR (RT-qPCR). A publicly available dataset was used to determine HNMT's potential as an NSCLC target molecule. Immunohistochemistry and coimmunoprecipitation were used to determine HNMT-HER2 correlations and interactions, respectively. HNMT shRNA and overexpression plasmids were used to explore HNMT functions in vitro and in vivo. We also examined miRNAs that may target HNMT and investigated HNMT/HER2's role on NSCLC cells' antioxidant properties. Finally, how HNMT loss affects NSCLC cells' sensitivity to cisplatin was investigated. RESULTS: HNMT was significantly upregulated in human NSCLC tissues, conferred a worse prognosis, and was coexpressed with HER2. HNMT depletion and overexpression respectively decreased and increased cell proliferation, colony formation, tumorsphere formation, and CSCs marker expression. Coimmunoprecipitation analysis indicated that HNMT directly interacts with HER2. TARGETSCAN analysis revealed that HNMT is a miR-223 and miR-3065-5p target. TBHp treatment increased HER2 expression, whereas shHNMT disrupted the Nuclear factor erythroid 2-related factor 2 (Nrf2)/ hemeoxygenase-1 (HO-1)/HER2 axis and increased reactive oxygen species accumulation in NSCLC cells. Finally, shHNMT sensitized H441 cells to cisplatin treatment in vitro and in vivo. CONCLUSIONS: Therefore, HNMT upregulation in NSCLC cells may upregulate HER2 expression, increasing tumorigenicity and chemoresistance through CSCs maintenance and antioxidant properties. This newly discovered regulatory axis may aid in retarding NSCLC progression and chemoresistance.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Histamine N-Methyltransferase/biosynthesis , Lung Neoplasms/enzymology , Neoplastic Stem Cells/enzymology , Oxidative Stress , Receptor, ErbB-2/metabolism , Up-Regulation , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Female , Histamine N-Methyltransferase/genetics , Humans , Lung Neoplasms/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Receptor, ErbB-2/genetics
7.
Sci Rep ; 12(1): 347, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013447

ABSTRACT

Cancer stem cells (CSCs) are capable of continuous proliferation, self-renewal and are proposed to play significant roles in oncogenesis, tumor growth, metastasis and cancer recurrence. We have established a model of CSCs that was originally developed from mouse induced pluripotent stem cells (miPSCs) by proposing miPSCs to the conditioned medium (CM) of cancer derived cells, which is a mimic of carcinoma microenvironment. Further research found that not only PI3K-Akt but also EGFR signaling pathway was activated during converting miPSCs into CSCs. In this study, we tried to observe both of PI3Kγ inhibitor Eganelisib and EGFR inhibitor Gefitinib antitumor effects on the models of CSCs derived from miPSCs (miPS-CSC) in vitro and in vivo. As the results, targeting these two pathways exhibited significant inhibition of cell proliferation, self-renewal, migration and invasion abilities in vitro. Both Eganelisib and Gefitinib showed antitumor effects in vivo while Eganelisib displayed more significant therapeutic efficacy and less side effects than Gefitinib on all miPS-CSC models. Thus, these data suggest that the inhibitiors of PI3K and EGFR, especially PI3Kγ, might be a promising therapeutic strategy against CSCs defeating cancer in the near future.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/metabolism , ErbB Receptors/antagonists & inhibitors , Gefitinib/pharmacology , Induced Pluripotent Stem Cells/drug effects , Neoplastic Stem Cells/drug effects , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , ErbB Receptors/metabolism , Female , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Signal Transduction , Tumor Burden/drug effects
8.
J Pathol ; 256(3): 349-362, 2022 03.
Article in English | MEDLINE | ID: mdl-34897678

ABSTRACT

Alveolar rhabdomyosarcoma (ARMS) is an aggressive pediatric cancer with poor prognosis. Cancer stem cells (CSCs) are seeds for tumor relapse and metastasis. However, pathways that maintain stemness genes are not fully understood. Here, we report that the enzyme euchromatic histone lysine methyltransferase 1 (EHMT1) is expressed in primary and relapse ARMS tumors. EHMT1 suppression impaired motility and induced differentiation in ARMS cell lines and reduced tumor progression in a mouse xenograft model in vivo. RNA sequencing of EHMT1-depleted cells revealed downregulation of ALDH1A1 that is associated with CSCs. Consistent with this, inhibition of ALDH1A1 expression and activity mimicked EHMT1 depletion phenotypes and reduced tumorsphere formation. Mechanistically, we demonstrate that EHMT1 does not bind to the ALDH1A1 promoter but activates it by stabilizing C/EBPß, a known regulator of ALDH1A1 expression. Our findings identify a role for EHMT1 in maintenance of stemness by regulating ALDH1A1 expression and suggest that targeting ALDH+ cells is a promising strategy in ARMS. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Neoplastic Stem Cells/enzymology , Retinal Dehydrogenase/metabolism , Rhabdomyosarcoma, Alveolar/enzymology , Aldehyde Dehydrogenase 1 Family/genetics , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/genetics , Humans , Mice, Nude , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Phenotype , Retinal Dehydrogenase/genetics , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/pathology , Signal Transduction , Tumor Burden
9.
J Appl Biomed ; 19(4): 181-189, 2021 12.
Article in English | MEDLINE | ID: mdl-34907737

ABSTRACT

INTRODUCTION: Cancer stem cells (CSCs) are a theorized subset of cells within the tumor that is thought to drive disease recurrence and metastatic spread. The aim of this study is to investigate mRNA and protein levels of ganglioside GD2 synthase (GD2S), in breast cancer (BC) patients. METHODS: 65 PBMCs of preoperative BC patients without chemotherapy were compared to PBMCs after chemotherapy and controls. RESULTS: GD2S were significantly higher in BC patients after chemotherapy compared to pre-chemotherapy at both mRNA and protein. GD2S was higher in pre-chemotherapy blood samples compared to control samples. CONCLUSIONS: Higher expression of GD2S in BC samples compared to healthy control indicates the potential utility of GD2S as a marker of malignancy.


Subject(s)
Breast Neoplasms , N-Acetylgalactosaminyltransferases , Neoplasm Recurrence, Local , Neoplastic Stem Cells , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Female , Humans , N-Acetylgalactosaminyltransferases/blood , Neoplasm Recurrence, Local/metabolism , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/metabolism , RNA, Messenger/metabolism
10.
Anticancer Res ; 41(11): 5481-5488, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34732418

ABSTRACT

BACKGROUND/AIM: Aldehyde dehydrogenases (ALDHs) are considered as markers for normal and cancer stem cells (CSC) and are involved in cell metabolism, proliferation, differentiation, stemness, and retinoic acid (RA) biosynthesis. The aim of the present study was to identify the ALDH isoforms that are associated with the CSC phenotype in non-small cell lung and hepatocellular carcinomas. MATERIALS AND METHODS: We utilized lung (A549) and hepatocellular (HepG2) cancer cells and generated tumor spheres to isolate the CSC sub-population. RESULTS: The CSC enrichment was confirmed by the up-regulation of various CSC-related genes. Comparative qPCR analysis indicated the up-regulation of several ALDH isoforms in A549 and HepG2 spheres. Interestingly, cyclin D1 and Akt, down-stream targets of the RA signaling pathway, were also shown to be significantly up-regulated in both sphere populations. CONCLUSION: Specific ALDH isoforms appear to be important mediators for the acquisition of an CSC phenotype and thus, are potential promising targets for CSC-based therapeutic approaches in lung and hepatocellular carcinomas.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Lung Neoplasms/enzymology , Neoplastic Stem Cells/enzymology , A549 Cells , Aldehyde Dehydrogenase/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cyclin D1/genetics , Cyclin D1/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Isoenzymes , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplastic Stem Cells/pathology , Phenotype , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Spheroids, Cellular
11.
Biomed Pharmacother ; 144: 112339, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34656057

ABSTRACT

Colorectal cancer is the second deadly cancer in the world. Trametes versicolor is a traditional Chinese medicinal mushroom with a long history of being used to regulate immunity and prevent cancer. Trametes versicolor mushroom extract demonstrates strongly cell growth inhibitory activity on human colorectal tumor cells. In this study, we characterized a novel 12-kDa protein that named musarin, which was purified from Trametes versicolor mushroom extract and showed significant growth inhibition on multiple human colorectal cancer cell lines in vitro. The protein sequence of musarin was determined through enzyme digestion and MS/MS analysis. Furthermore, Musarin, in particular, strongly inhibits aggressive human colorectal cancer stem cell-like CD24+CD44+ HT29 proliferation in vitro and in a NOD/SCID murine xenograft model. Through whole transcription profile and gene enrichment analysis of musarin-treated CSCs-like cells, major signaling pathways and network modulated by musarin have been enriched, including the bioprocess of the Epithelial-Mesenchymal Transition, the EGFR-Ras signaling pathway and enzyme inhibitor activity. Musarin demonstrated tyrosine kinase inhibitory activity in vitro. Musarin strongly attenuated EGFR expression and down-regulated phosphorylation level, thereby slowing cancer cells proliferation. In addition, oral ingestion of musarin significantly inhibited CD24+CD44+ HT29 generated tumor development in SCID/NOD mice with less side effects in microgram doses. Targeting self-renewal aggressive stem-cell like cancer cell proliferation, with higher water solubility and lower cytotoxicity, musarin has shown strong potence to be developed as a promising novel therapeutic drug candidate against colorectal cancers, especially those that acquire chemo-resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Fungal Proteins/pharmacology , Neoplastic Stem Cells/drug effects , Polyporaceae , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/toxicity , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fungal Proteins/isolation & purification , Fungal Proteins/toxicity , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , HT29 Cells , Humans , Male , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Polyporaceae/chemistry , Protein Kinase Inhibitors/isolation & purification , Protein Kinase Inhibitors/toxicity , Signal Transduction , Transcriptome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
Life Sci Alliance ; 4(12)2021 12.
Article in English | MEDLINE | ID: mdl-34645618

ABSTRACT

Better understanding of GBM signalling networks in-vivo would help develop more physiologically relevant ex vivo models to support therapeutic discovery. A "functional proteomics" screen was undertaken to measure the specific activity of a set of protein kinases in a two-step cell-free biochemical assay to define dominant kinase activities to identify potentially novel drug targets that may have been overlooked in studies interrogating GBM-derived cell lines. A dominant kinase activity derived from the tumour tissue, but not patient-derived GBM stem-like cell lines, was Bruton tyrosine kinase (BTK). We demonstrate that BTK is expressed in more than one cell type within GBM tissue; SOX2-positive cells, CD163-positive cells, CD68-positive cells, and an unidentified cell population which is SOX2-negative CD163-negative and/or CD68-negative. The data provide a strategy to better mimic GBM tissue ex vivo by reconstituting more physiologically heterogeneous cell co-culture models including BTK-positive/negative cancer and immune cells. These data also have implications for the design and/or interpretation of emerging clinical trials using BTK inhibitors because BTK expression within GBM tissue was linked to longer patient survival.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/mortality , Glioblastoma/enzymology , Glioblastoma/mortality , Proteome/metabolism , Signal Transduction , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Coculture Techniques/methods , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/enzymology , Proteomics/methods , SOXB1 Transcription Factors/metabolism , Survival Rate
14.
Toxicol Appl Pharmacol ; 429: 115700, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34464674

ABSTRACT

3,3'-diindolylmethane (DIM) is a dimer compound converted from Indoly-3-carbinol that had been studied as promising chemo-preventive agent against breast cancer. In this study, we observed that proportion of CD133+Nanog+ subpopulation in MCF-7 cells was significantly increased after DIM administration with up-regulated AKT activity by using CyTOF assay. SPADE analysis revealed this stem-like subpopulation exhibited apoptosis-resistance property against DIM treatment. By combining with AKT inhibitor AZD5363, DIM induced CD133 expression could be suppressed. In addition, a combination treatment of MCF-7 and MDA-MB-231 breast cancer cells with DIM and AZD5363 showed synergistic decreases in cell proliferation and induced apoptosis. Furthermore, results from imaging flow cytometry suggested that FoxO3a nuclear localization and PUMA expression could be improved by combination of AZD5363 with DIM. Taken together, the above observations suggested that the combination of AZD5363 with DIM could be developed as potential therapy for breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Indoles/pharmacology , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrroles/pharmacology , AC133 Antigen/metabolism , Active Transport, Cell Nucleus , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Female , Forkhead Box Protein O3/metabolism , Humans , MCF-7 Cells , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
15.
Int J Mol Sci ; 22(13)2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34209376

ABSTRACT

Chronic myeloid leukemia (CML) is a classical example of stem cell cancer since it arises in a multipotent hematopoietic stem cell upon the acquisition of the t(9;22) chromosomal translocation, that converts it into a leukemic stem cell (LSC). The resulting BCR-ABL1 fusion gene encodes a deregulated tyrosine kinase that is recognized as the disease driver. Therapy with tyrosine kinase inhibitors (TKIs) eliminates progenitor and more differentiated cells but fails to eradicate quiescent LSCs. Thus, although many patients obtain excellent responses and a proportion of them can even attempt treatment discontinuation (treatment free remission [TFR]) after some years of therapy, LSCs persist, and represent a potentially dangerous reservoir feeding relapse and hampering TFR. Over the past two decades, intensive efforts have been devoted to the characterization of CML LSCs and to the dissection of the cell-intrinsic and -extrinsic mechanisms sustaining their persistence, in an attempt to find druggable targets enabling LSC eradication. Here we provide an overview and an update on these mechanisms, focusing in particular on the most recent acquisitions. Moreover, we provide a critical appraisal of the clinical relevance and feasibility of LSC targeting in CML.


Subject(s)
Drug Delivery Systems , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Neoplastic Stem Cells/enzymology , Protein Kinase Inhibitors/therapeutic use , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
16.
Cell Cycle ; 20(16): 1589-1602, 2021 08.
Article in English | MEDLINE | ID: mdl-34288821

ABSTRACT

Bladder cancer (BLCA) is a common malignant urothelial cancer in the world. Although circular RNAs (circRNAs) involve in regulating BLCA progression, the role of a novel circular RNA circSETD3 in regulating BLCA pathogenesis has not been studied. The expression of circSETD3, miR-641, PTEN mRNA in BLCA tissues and cell lines were measured using RT-qPCR. The gain-of-function experiments were performed in vitro and in vivo to detect the effects of circSETD3 on cell proliferation, migration, EMT, and stemness maintenance. Besides, rescue experiments were performed to demonstrate the regulatory mechanism of circSETD3/miR-641/PTEN in BLCA cell malignant phenotypes in vitro. CircSETD3 was remarkably downregulated in the cancerous clinical tissues and cell lines, in contrast with their normal counterparts, and circSETD3 tended to be deficient in BLCA patients with larger tumor size, advanced clinical stages, positive lymph metastasis and worse prognosis. In addition, circular isoforms of circSETD3 were more resistant to RNase R+ and actinomycetes D treatment compared to their linear isoforms, and circSETD3 mainly distributed in the cytoplasm of the BLCA cells. Further gain-of-function experiments showed that circSETD3 acted as a tumor suppressor to suppress BLCA cell proliferation, migration, EMT and stemness, and the underlying mechanisms had also been elucidated. Mechanistically, circSETD3 sponged miR-641 to upregulate PTEN, resulting in the blockage of BLCA progression. Our findings indicated that circSETD3 acted as a vital tumor suppressor in BLCA via regulating the miR-641/PTEN axis.


Subject(s)
Cell Movement , Cell Proliferation , MicroRNAs/metabolism , Neoplastic Stem Cells/enzymology , PTEN Phosphohydrolase/metabolism , RNA, Circular/metabolism , Urinary Bladder Neoplasms/enzymology , Cell Line, Tumor , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , PTEN Phosphohydrolase/genetics , Phenotype , RNA, Circular/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
17.
Biochem J ; 478(14): 2899-2920, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34319405

ABSTRACT

Cancer stem cells (CSCs) are involved in the initiation and progression of human malignancies by enabling cancer tissue self-renewal capacity and constituting the therapy-resistant population of tumor cells. However, despite the exhausting characterization of CSC genetics, epigenetics, and kinase signaling, eradication of CSCs remains an unattainable goal in most human malignancies. While phosphatases contribute equally with kinases to cellular phosphoregulation, our understanding of phosphatases in CSCs lags severely behind our knowledge about other CSC signaling mechanisms. Many cancer-relevant phosphatases have recently become druggable, indicating that further understanding of the CSC phosphatases might provide novel therapeutic opportunities. This review summarizes the current knowledge about fundamental, but yet poorly understood involvement of phosphatases in the regulation of major CSC signaling pathways. We also review the functional roles of phosphatases in CSC self-renewal, cancer progression, and therapy resistance; focusing particularly on hematological cancers and glioblastoma. We further discuss the small molecule targeting of CSC phosphatases and their therapeutic potential in cancer combination therapies.


Subject(s)
Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Phosphoric Monoester Hydrolases/metabolism , Signal Transduction , Cell Self Renewal/genetics , Disease Progression , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic/genetics , Humans , Models, Biological , Neoplasms/genetics , Neoplasms/therapy , Neoplastic Stem Cells/enzymology , Phosphoric Monoester Hydrolases/classification
18.
Cancer Rep (Hoboken) ; 4(6): e1417, 2021 12.
Article in English | MEDLINE | ID: mdl-34101386

ABSTRACT

BACKGROUND: Bladder cancer (BC) is the 10th most common cancer in the world. BC with muscle invasion results in a poor prognosis and is usually fatal. Cancer cell metabolism has an essential role in the development and progression of tumors. Expression of tryptophan 2,3-dioxygenase (TDO2) is associated with tumor progression and worse survival in some other cancers. However, no studies have been performed to uncover the biofunctional roles of TDO2 in BC. AIM: This study aim to investigate the clinicopathologic significance of TDO2 in BC. METHODS AND RESULTS: TDO2 expression was evaluated by qRT-PCR and immunohistochemistry in an integrated analysis with the Cancer Genome Atlas (TCGA) and other published datasets. TDO2 overexpression was significantly associated with T classification, N classification, and M classification, tumor stage, recurrence, and basal type, and with the expression of CD44 and aldehyde dehydrogenase 1 (ALDH1) in BC. High TDO2 expression correlated with poor outcome of BC patients. Using BC cell lines with knockdown and forced expression of TDO2, we found that TDO2 was involved in the growth, migration, and invasiveness of BC cells. Moreover, TDO2 was found to be crucial for spheroid formation in BC cells. Importantly, TDO2 promoted BC cells resistance to cetuximab through integration of the EGFR pathway. CONCLUSION: Our results indicate that TDO2 might take an essential part in BC progression and could be a potential marker for targeted therapy in BC.


Subject(s)
Cetuximab/pharmacology , Drug Resistance, Neoplasm , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Tryptophan Oxygenase/metabolism , Urinary Bladder Neoplasms/pathology , Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor , Case-Control Studies , Follow-Up Studies , Humans , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/enzymology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Prognosis , Survival Rate , Tryptophan Oxygenase/genetics , Tumor Cells, Cultured , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/enzymology
19.
Int J Mol Sci ; 22(11)2021 May 26.
Article in English | MEDLINE | ID: mdl-34073600

ABSTRACT

The activity of nicotinamide N-methyltransferase (NNMT) is tightly linked to the maintenance of the nicotinamide adenine dinucleotide (NAD+) level. This enzyme catalyzes methylation of nicotinamide (NAM) into methyl nicotinamide (MNAM), which is either excreted or further metabolized to N1-methyl-2-pyridone-5-carboxamide (2-PY) and H2O2. Enzymatic activity of NNMT is important for the prevention of NAM-mediated inhibition of NAD+-consuming enzymes poly-adenosine -diphosphate (ADP), ribose polymerases (PARPs), and sirtuins (SIRTs). Inappropriately high expression and activity of NNMT, commonly present in various types of cancer, has the potential to disrupt NAD+ homeostasis and cellular methylation potential. Largely overlooked, in the context of cancer, is the inhibitory effect of 2-PY on PARP-1 activity, which abrogates NNMT's positive effect on cellular NAD+ flux by stalling liberation of NAM and reducing NAD+ synthesis in the salvage pathway. This review describes, and discusses, the mechanisms by which NNMT promotes NAD+ depletion and epigenetic reprogramming, leading to the development of metabolic plasticity, evasion of a major tumor suppressive process of cellular senescence, and acquisition of stem cell properties. All these phenomena are related to therapy resistance and worse clinical outcomes.


Subject(s)
NAD/metabolism , Neoplasm Proteins/metabolism , Neoplasms , Neoplastic Stem Cells/enzymology , Niacinamide/metabolism , Nicotinamide N-Methyltransferase/metabolism , Humans , Methylation , Neoplasms/epidemiology , Neoplasms/therapy
20.
Cell Death Dis ; 12(7): 633, 2021 06 19.
Article in English | MEDLINE | ID: mdl-34148069

ABSTRACT

Colorectal cancer (CRC) stem cells are resistant to cancer therapy and are therefore responsible for tumour progression after conventional therapy fails. However, the molecular mechanisms underlying the maintenance of stemness are poorly understood. In this study, we identified PCGF1 as a crucial epigenetic regulator that sustains the stem cell-like phenotype of CRC. PCGF1 expression was increased in CRC and was significantly correlated with cancer progression and poor prognosis in CRC patients. PCGF1 knockdown inhibited CRC stem cell proliferation and CRC stem cell enrichment. Importantly, PCGF1 silencing impaired tumour growth in vivo. Mechanistically, PCGF1 bound to the promoters of CRC stem cell markers and activated their transcription by increasing the H3K4 histone trimethylation (H3K4me3) marks and decreasing the H3K27 histone trimethylation (H3K27me3) marks on their promoters by increasing expression of the H3K4me3 methyltransferase KMT2A and the H3K27me3 demethylase KDM6A. Our findings suggest that PCGF1 is a potential therapeutic target for CRC treatment.


Subject(s)
Colorectal Neoplasms/enzymology , DNA Methylation , Epigenesis, Genetic , Neoplastic Stem Cells/enzymology , Polycomb Repressive Complex 1/metabolism , Animals , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Mice, Inbred BALB C , Mice, Nude , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Polycomb Repressive Complex 1/genetics , Tumor Burden , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...