1
|
Zhu W, Fu M, Li Q, Chen X, Liu Y, Li X, Luo N, Tang W, Zhang Q, Yang F, Chen Z, Zhang Y, Peng B, Zhang Q, Zhang Y, Peng X, Hu G. Amino acid metabolism-related genes as potential biomarkers and the role of MATN3 in stomach adenocarcinoma: A bioinformatics, mendelian randomization and experimental validation study. Int Immunopharmacol 2024; 143:113253. [PMID: 39353384 DOI: 10.1016/j.intimp.2024.113253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 09/11/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) is a major contributor to cancer-related mortality worldwide. Alterations in amino acid metabolism, which is integral to protein synthesis, have been observed across various tumor types. However, the prognostic significance of amino acid metabolism-related genes in STAD remains underexplored. METHODS Transcriptomic gene expression and clinical data for STAD patients were obtained from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Amino acid metabolism-related gene sets were sourced from the Gene Set Enrichment Analysis (GSEA) database. A prognostic model was built using LASSO Cox regression based on the TCGA cohort and validated with GEO datasets (GSE84433, GSE84437, GSE84426). Kaplan-Meier analysis compared overall survival (OS) between high- and low-risk groups, and ROC curves assessed model accuracy. A nomogram predicted 1-, 3-, and 5-year survival. Copy number variations (CNVs) in model genes were visualized using data from the Xena platform, and mutation profiles were analyzed with "maftools" to create a waterfall plot. KEGG and GO enrichment analyses were performed to explore biological mechanisms. Immune infiltration and related functions were evaluated via ssGSEA, and Spearman correlation analyzed associations between risk scores and immune components. The TIDE database predicted immunotherapy efficacy, while FDA-approved drug sensitivity was assessed through CellMiner database. The role of MATN3 in STAD was further examined in vitro and in vivo, including amino acid-targeted metabolomic sequencing to assess its impact on metabolism. Finally, Mendelian randomization (MR) analysis evaluated the causal relationship between the model genes and gastric cancer. RESULTS In this study, we developed a prognostic risk model for STAD based on three amino acid metabolism-related genes (SERPINE1, NRP1, MATN3) using LASSO regression analysis. CNV amplification was common in SERPINE1 and NRP1, while CNV deletion frequently occurred in MATN3. STAD patients were classified into high- and low-risk groups based on the median risk score, with the high-risk group showing worse prognosis. A nomogram incorporating the risk score and clinical factors was created to estimate 1-, 3-, and 5-year survival rates. Distinct mutation profiles were observed between risk groups, with KEGG pathway analysis showing immune-related pathways enriched in the high-risk group. High-risk scores were significantly associated with the C6 (TGF-β dominant) subtype, while low-risk scores correlated with the C4 (lymphocyte-depleted) subtype. Higher risk scores also indicated increased immune infiltration, enhanced immune functions, lower tumor purity, and poorer immunotherapy response. Model genes were linked to anticancer drug sensitivity. Manipulating MATN3 expression showed that it promoted STAD cell proliferation and migration in vitro and tumor growth in vivo. Metabolomic sequencing revealed that MATN3 knockdown elevated levels of 30 amino acid metabolites, including alpha-aminobutyric acid, glycine, and aspartic acid, while reducing (S)-β-Aminoisobutyric acid and argininosuccinic acid. MR analysis found a significant causal effect of NRP1 on gastric cancer, but no causal relationship for MATN3 or SERPINE1. CONCLUSION In conclusion, the amino acid metabolism-related prognostic model shows promise as a valuable biomarker for predicting the clinical prognosis, selecting immunotherapy and drug treatment for STAD patients. Furthermore, our study has shed light on the potential value of the MATN3 as a promising strategy for combating the progression of STAD.
Collapse
Affiliation(s)
- Wenjun Zhu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Min Fu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qianxia Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xin Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuanhui Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoyu Li
- Department of Oncology, Hubei Cancer Hospital, Wuhan 430000, China
| | - Na Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenhua Tang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qing Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feng Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ziqi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yiling Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bi Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qiang Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuanyuan Zhang
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Xiaohong Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
2
|
Hlozkova K, Vasylkivska M, Boufersaoui A, Marzullo B, Kolarik M, Alquezar-Artieda N, Shaikh M, Alaei NF, Zaliova M, Zwyrtkova M, Bakardijeva-Mihaylova V, Alberich-Jorda M, Trka J, Tennant DA, Starkova J. Rewired glutamate metabolism diminishes cytostatic action of L-asparaginase. Cancer Lett 2024; 605:217242. [PMID: 39270769 DOI: 10.1016/j.canlet.2024.217242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 09/02/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
Tumor cells often adapt to amino acid deprivation through metabolic rewiring, compensating for the loss with alternative amino acids/substrates. We have described such a scenario in leukemic cells treated with L-asparaginase (ASNase). Clinical effect of ASNase is based on nutrient stress achieved by its dual enzymatic action which leads to depletion of asparagine and glutamine and is accompanied with elevated aspartate and glutamate concentrations in serum of acute lymphoblastic leukemia patients. We showed that in these limited conditions glutamate uptake compensates for the loss of glutamine availability. Extracellular glutamate flux detection confirms its integration into the TCA cycle and its participation in nucleotide and glutathione synthesis. Importantly, it is glutamate-driven de novo synthesis of glutathione which is the essential metabolic pathway necessary for glutamate's pro-survival effect. In vivo findings support this effect by showing that inhibition of glutamate transporters enhances the therapeutic effect of ASNase. In summary, ASNase induces elevated extracellular glutamate levels under nutrient stress, which leads to a rewiring of intracellular glutamate metabolism and has a negative impact on ASNase treatment.
Collapse
Affiliation(s)
- Katerina Hlozkova
- Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Second Faculty of Medicine, Department of Pediatric Hematology and Oncology, Charles University, Prague, Czech Republic.
| | - Maryna Vasylkivska
- Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Second Faculty of Medicine, Department of Pediatric Hematology and Oncology, Charles University, Prague, Czech Republic
| | - Adam Boufersaoui
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Bryan Marzullo
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Matus Kolarik
- Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Second Faculty of Medicine, Department of Pediatric Hematology and Oncology, Charles University, Prague, Czech Republic; First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Natividad Alquezar-Artieda
- Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Second Faculty of Medicine, Department of Pediatric Hematology and Oncology, Charles University, Prague, Czech Republic
| | - Mehak Shaikh
- Laboratory of Hemato-Oncology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Nadia Fatemeh Alaei
- Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Second Faculty of Medicine, Department of Pediatric Hematology and Oncology, Charles University, Prague, Czech Republic; Laboratory of Hemato-Oncology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Marketa Zaliova
- Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Second Faculty of Medicine, Department of Pediatric Hematology and Oncology, Charles University, Prague, Czech Republic; University Hospital Motol, Prague, Czech Republic
| | - Martina Zwyrtkova
- Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Second Faculty of Medicine, Department of Pediatric Hematology and Oncology, Charles University, Prague, Czech Republic
| | - Violeta Bakardijeva-Mihaylova
- Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Second Faculty of Medicine, Department of Pediatric Hematology and Oncology, Charles University, Prague, Czech Republic
| | - Meritxell Alberich-Jorda
- Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Second Faculty of Medicine, Department of Pediatric Hematology and Oncology, Charles University, Prague, Czech Republic; Laboratory of Hemato-Oncology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Trka
- Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Second Faculty of Medicine, Department of Pediatric Hematology and Oncology, Charles University, Prague, Czech Republic; University Hospital Motol, Prague, Czech Republic
| | - Daniel A Tennant
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Julia Starkova
- Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Second Faculty of Medicine, Department of Pediatric Hematology and Oncology, Charles University, Prague, Czech Republic; University Hospital Motol, Prague, Czech Republic.
| |
Collapse
|
3
|
Ochi N, Miyake N, Takeyama M, Yamane H, Fukazawa T, Nagasaki Y, Kawahara T, Ichiyama N, Kosaka Y, Mimura A, Nakanishi H, Hiraki A, Kiura K, Takigawa N. The combined inhibition of SLC1A3 and glutaminase in osimertinib-resistant EGFR mutant cells. Biochim Biophys Acta Gen Subj 2024; 1868:130675. [PMID: 39059510 DOI: 10.1016/j.bbagen.2024.130675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/10/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND We investigated the unknown mechanisms of osimertinib-resistant EGFR-mutant lung cancer. METHODS An osimertinib-resistant cell line (PC-9/OsmR2) was established through continuous exposure to osimertinib using an EGFR exon 19 deletion (19Del) lung adenocarcinoma cell line (PC-9). EGFR 19Del (M1), L858R/T790M/C797S (M6), and L858R/C797S (M8) expression vectors were introduced into Ba/F3 cells. A second osimertinib-resistant line (M1/OsmR) was established through continuous exposure to osimertinib using M1 cells. RESULTS SLC1A3 had the highest mRNA expression level in PC-9/OsmR2 compared to PC-9 cells by microarray analysis and SLC1A3 was increased by flow cytometry. In PC-9/OsmR2 cells, osimertinib sensitivity was significantly increased in combination with siSLC1A3. Because SLC1A3 functions in glutamic acid transport, osimertinib with a glutaminase inhibitor (CB-839) or an SLC1A3 inhibitor (TFB-TBOA) increased the sensitivity. Also, CB-839 plus TFB-TBOA without osimertinib resulted in greater susceptibility than did CB-839 or TFB-TBOA plus osimertinib. Comprehensive metabolome analysis showed that the M1/OsmR cells had significantly more glutamine and glutamic acid than M1 cells. CB-839 plus osimertinib exerted a synergistic effect on M6 cells and an additive effect on M8 cells. CONCLUSION Targeting glutaminase and glutamic acid may overcome the osimertinib-resistant EGFR-mutant lung cancer.
Collapse
Affiliation(s)
- Nobuaki Ochi
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Noriko Miyake
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan; Kajiki Hospital, Okayama, Japan
| | - Masami Takeyama
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Hiromichi Yamane
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Takuya Fukazawa
- Department of General Surgery, Kawasaki Medical School, Okayama, Japan
| | - Yasunari Nagasaki
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Tatsuyuki Kawahara
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Naruhiko Ichiyama
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Youko Kosaka
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Ayaka Mimura
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Hidekazu Nakanishi
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | | | | | - Nagio Takigawa
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan.
| |
Collapse
|
4
|
Huang YK, Wang TM, Chen CY, Li CY, Wang SC, Irshad K, Pan Y, Chang KC. The role of ALDH1A1 in glioblastoma proliferation and invasion. Chem Biol Interact 2024; 402:111202. [PMID: 39128802 DOI: 10.1016/j.cbi.2024.111202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 07/07/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
High-grade gliomas, including glioblastoma multiforme (GBM), continue to be a leading aggressive brain tumor in adults, marked by its rapid growth and invasive nature. Aldehyde dehydrogenase 1 family, member A1 (ALDH1A1), an enzyme, plays a significant role in tumor progression, yet its function in high-grade gliomas is still poorly investigated. In this study, we evaluated ALDH1A1 levels in clinical samples of GBM. We also assessed the prognostic significance of ALDH1A1 expression in GBM and LGG (low grade glioma) patients using TCGA (The Cancer Genome Atlas) database analysis. The MTT and transwell assays were utilized to examine cell growth and the invasive capability of U87 cells, respectively. We quantitatively examined markers for cell proliferation (Ki-67 and cyclin D1) and invasion (MMP2 and 9). A Western blot test was conducted to determine the downstream signaling of ALDH1A1. We found a notable increase in ALDH1A1 expression in high-grade gliomas compared to their low-grade counterparts. U87 cells that overexpressed ALDH1A1 showed increased cell growth and invasion. We found that ALDH1A1 promotes the phosphorylation of AKT, and inhibiting AKT phosphorylation mitigates the ALDH1A1's effects on tumor growth and migration. In summary, our findings suggest ALDH1A1 as a potential therapeutic target for GBM treatment.
Collapse
Affiliation(s)
- Yu-Kai Huang
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Tzu-Ming Wang
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
| | - Chi-Yu Chen
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Shu-Chi Wang
- Department of Medical Laboratory Science and Biotechnology, Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Khushboo Irshad
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yuan Pan
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kun-Che Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA; Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
5
|
Wei X, Li Z, Zheng H, Li X, Lin Y, Yang H, Shen Y. Long non-coding RNA MAGEA4-AS1 binding to p53 enhances MK2 signaling pathway and promotes the proliferation and metastasis of oral squamous cell carcinoma. Funct Integr Genomics 2024; 24:158. [PMID: 39249547 PMCID: PMC11384635 DOI: 10.1007/s10142-024-01436-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/10/2024]
Abstract
Long non-coding RNAs (lncRNAs) regulate the occurrence, development and progression of oral squamous cell carcinoma (OSCC). We elucidated the expression features of MAGEA4-AS1 in patients with OSCC and its activity as an OSCC biomarker. Furthermore, the impact of up-regulation of MAGEA4-AS1 on the cellular behaviors (proliferation, migration and invasion) of OSCC cells and intrinsic signal mechanisms were evaluated. Firstly, we analyzed MAGEA4-AS1 expression data in The Cancer Genome Atlas (TCGA) OSCC using a bioinformatics approach and in 45 pairs of OSCC tissues using qPCR. Then CCK-8, ethynyl deoxyuridine, colony formation, transwell and wound healing assays were conducted to assess changes in the cell proliferation, migration and invasion protential of shMAGEA4-AS1 HSC3 and CAL27 cells. The RNA sequence of MAGEA4-AS1 was identified using the rapid amplification of cDNA ends (RACE) assay. And whole-transcriptome sequencing was used to identify MAGEA4-AS1 affected genes. Additionally, dual-luciferase reporter system, RNA-binding protein immunoprecipitation (RIP), and rescue experiments were performed to clarify the role of the MAGEA4-AS1-p53-MK2 signaling pathway. As results, we found MAGEA4-AS1 was up-regulated in OSCC tissues. We identified a 418 nucleotides length of the MAGEA4-AS1 transcript and it primarily located in the cell nucleus. MAGEA4-AS1 stable knockdown weakened the proliferation, migration and invasion abilities of OSCC cells. Mechanistically, p53 protein was capable to activate MK2 gene transcription. RIP assay revealed an interaction between p53 and MAGEA4-AS1. MK2 up-regulation in MAGEA4-AS1 down-regulated OSCC cells restored MK2 and epithelial-to-mesenchymal transition related proteins' expression levels. In conclusion, MAGEA4-AS1-p53 complexes bind to MK2 promoter, enhancing the transcription of MK2 and activating the downstream signaling pathways, consequently promoting the proliferation and metastasis of OSCC cells. MAGEA4-AS1 may serve as a diagnostic marker and therapeutic target for OSCC patients.
Collapse
Affiliation(s)
- Xiaoxiao Wei
- Peking University Shenzhen Hospital Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, The Institute of Stomatology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| | - Zhangfu Li
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, The Institute of Stomatology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| | - Heng Zheng
- Peking University Shenzhen Hospital Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, The Institute of Stomatology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| | - Xiaolian Li
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, The Institute of Stomatology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| | - Yuntao Lin
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, The Institute of Stomatology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| | - Hongyu Yang
- Peking University Shenzhen Hospital Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, China.
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, The Institute of Stomatology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China.
| | - Yuehong Shen
- Peking University Shenzhen Hospital Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, China.
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, The Institute of Stomatology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China.
| |
Collapse
|
6
|
Hu S, Zhang X, Xin H, Guo M, Xiao Y, Chang Z, Luo Q, Li Y, Zhu C. Identification of PI3K-AKT Pathway-Related Genes and Construction of Prognostic Prediction Model for ccRCC. Cancer Rep (Hoboken) 2024; 7:e70010. [PMID: 39233640 PMCID: PMC11375326 DOI: 10.1002/cnr2.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 07/23/2024] [Accepted: 08/10/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC), the predominate histological type of renal cell carcinoma (RCC), has been extensively studied, with poor prognosis as the stage increases. Research findings consistently indicated that the PI3K-Akt pathway is commonly dysregulated across various cancer types, including ccRCC. Targeting the PI3K-Akt pathway held promise as a potential therapeutic approach for treating ccRCC. Development and validation of PI3K-Akt pathway-related genes related biomarkers can enhance healthcare management of patients with ccRCC. PURPOSE This study aimed to identify the key genes in the PI3K-Akt pathway associated with the diagnosis and prognosis of CCRCC using data mining from the Cancer Genome Atlas (TCGA) and Gene Expression Synthesis (GEO) datasets. METHODS The purpose of this study is to use bioinformatics methods to screen data sets and clinicopathological characteristics associated with ccRCC patients. The exhibited significantly differential expressed genes (DEGs) associated with the PI3K-Akt pathway were examined by KEGG. In addition, Kaplan-Meier (KM) analysis used to estimate the survival function of the differential genes by using the UALCAN database and graphPad Prism 9.0. And exploring the association between the expression levels of the selected genes and the survival status and time of patients with ccRCC based on SPSS22.0. Finally, a multigene prognostic model was constructed to assess the prognostic risk of ccRCC patients. RESULTS A total of 911 genes with common highly expressed were selected based on the GEO and TCGA databases. According to the KEGG pathway analysis, there were 42 genes enriched in PI3K-Akt signalling pathway. And seven of highly expressed genes were linked to a poor prognosis in ccRCC. And a multigene prognostic model was established based on IL2RG, EFNA3, and MTCP1 synergistic expression might be utilized to predict the survival of ccRCC patients. CONCLUSIONS Three PI3K-Akt pathway-related genes may be helpful to identify the prognosis and molecular characteristics of ccRCC patients and to improve therapeutic regimens, and these risk characteristics might be further applied in the clinic.
Collapse
Affiliation(s)
- Shaowen Hu
- Department of Urinary SurgeryHuaihe Hospital of Henan UniversityKaifengChina
| | - Xiaoli Zhang
- Department of Urinary SurgeryHuaihe Hospital of Henan UniversityKaifengChina
| | - Huiru Xin
- Department of Thoracic and Cardiovascular SurgeryHuaihe Hospital of Henan UniversityKaifengChina
| | - Mingjie Guo
- Department of Thoracic and Cardiovascular SurgeryThe First Affiliated Hospital of Henan UniversityKaifengChina
| | - Yafei Xiao
- Gastrointestinal Surgery, Huaihe Hospital of Henan UniversityKaifengChina
| | - Zhongwei Chang
- Gastrointestinal Surgery, Huaihe Hospital of Henan UniversityKaifengChina
| | - Qingyang Luo
- Department of Urinary SurgeryHuaihe Hospital of Henan UniversityKaifengChina
| | - Yang Li
- Department of Urinary SurgeryHuaihe Hospital of Henan UniversityKaifengChina
| | - Chaoyang Zhu
- Department of Urinary SurgeryHuaihe Hospital of Henan UniversityKaifengChina
| |
Collapse
|
7
|
Gao M, Shen Y, Yang P, Yuan C, Sun Y, Li Z. Transcriptomics integrated with metabolomics reveals partial molecular mechanisms of nutritional risk and neurodevelopment in children with congenital heart disease. Front Cardiovasc Med 2024; 11:1414089. [PMID: 39185136 PMCID: PMC11341388 DOI: 10.3389/fcvm.2024.1414089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/30/2024] [Indexed: 08/27/2024] Open
Abstract
Purpose To explore molecular mechanisms affecting nutritional risk and neurodevelopment in children with congenital heart disease (CHD) by combining transcriptome and metabolome analysis. Methods A total of 26 blood and serum samples from 3 groups of children with CHD low nutritional risk combined with normal neurodevelopment (group A), low nutritional risk combined with neurodevelopmental disorders (group B) and high nutritional risk combined with normal neurodevelopment (group C) were analyzed by transcriptome and metabolomics to search for differentially expressed genes (DEGs) and metabolites (DEMs). Functional analysis was conducted for DEGs and DEMs. Further, the joint pathway analysis and correlation analysis of DEGs and DEMs were performed. Results A total of 362 and 1,351 DEGs were detected in group B and C compared to A, respectively. A total of 6 and 7 DEMs were detected in group B and C compared to A in positive mode, respectively. There were 39 and 31 DEMs in group B and C compared to A in negative mode. Transcriptomic analysis indicated that neurodevelopment may be regulated by some genes such as NSUN7, SLC6A8, CXCL1 and LCN8, nutritional risk may be regulated by SLC1A3 and LCN8. Metabolome analysis and joint pathway analysis showed that tryptophan metabolism, linoleic and metabolism and glycerophospholipid metabolism may be related to neurodevelopment, and glycerophospholipid metabolism pathway may be related to nutritional risk. Conclusion By integrating transcriptome and metabolome analyses, this study revealed key genes and metabolites associated with nutritional risk and neurodevelopment in children with CHD, as well as significantly altered pathways. It has important clinical translational significance.
Collapse
Affiliation(s)
- Minglei Gao
- Heart Center, Qingdao Women and Children’s Hospital, Shandong University, Qingdao, China
- Heart Center, Dalian Women and Children’s Medical Group, Dalian, China
| | - Yang Shen
- Clinical Laboratory, Dalian Women and Children’s Medical Group, Dalian, China
| | - Ping Yang
- Heart Center, Dalian Women and Children’s Medical Group, Dalian, China
| | - Chang Yuan
- Heart Center, Dalian Women and Children’s Medical Group, Dalian, China
| | - Yanan Sun
- Heart Center, Dalian Women and Children’s Medical Group, Dalian, China
| | - Zipu Li
- Heart Center, Qingdao Women and Children’s Hospital, Shandong University, Qingdao, China
| |
Collapse
|
8
|
Zong Z, Tang G, Guo Y, Kong F. Down-regulated expression of TIPE3 inhibits malignant progression of non-small cell lung cancer via Wnt signaling. Exp Cell Res 2024; 439:114093. [PMID: 38759744 DOI: 10.1016/j.yexcr.2024.114093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/12/2024] [Accepted: 05/12/2024] [Indexed: 05/19/2024]
Abstract
Non-small cell lung cancer (NSCLC) accounts for approximately 80 % of all lung cancers with a low five-year survival rate. Therefore, the mechanistic pathways and biomarkers of NSCLC must be explored to elucidate its pathogenesis. In this study, we examined TIPE3 expression in NSCLC cells and investigated the molecular mechanisms underlying NSCLC regulation in vivo and in vitro. We collected tissue samples from patients with NSCLC to examine TIPE3 expression and its association with patient metastasis and prognosis. Furthermore, we evaluated the expression level of TIPE3 in NSCLC cells. Cell lines with the highest expression were selected for molecular mechanism experiments, and animal models were established for in vivo verification. The results showed that TIPE3 was significantly increased in patients with NSCLC, and this increased expression was associated with tumor metastasis and patient prognosis. TIPE3 knockdown inhibited proliferation, migration, invasion, EMT, angiogenesis, and tumorsphere formation in NSCLC cells. Moreover, it reduced the metabolic levels of tumor cells. However, overexpression of TIPE3 has the opposite effect. The in vivo results showed that TIPE3 knockdown reduced tumor volume, weight, and metastasis. Furthermore, the results showed that TIPE3 may inhibit malignant progression of NSCLC via the regulation of Wnt/β-catenin expression. These findings suggest that TIPE3 is significantly elevated in patients with NSCLC and that downregulation of TIPE3 can suppress the malignant progression of NSCLC, which could serve as a potential diagnostic and treatment strategy for NSCLC.
Collapse
Affiliation(s)
- Zhenfeng Zong
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei, 061000, China.
| | - Guojie Tang
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei, 061000, China
| | - Yu Guo
- Department of Respiratory Medicine, Hejian People's Hospital, Cangzhou, Hebei, 061000, China
| | - Fanyi Kong
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei, 061000, China
| |
Collapse
|
9
|
Hushmandi K, Einollahi B, Saadat SH, Lee EHC, Farani MR, Okina E, Huh YS, Nabavi N, Salimimoghadam S, Kumar AP. Amino acid transporters within the solute carrier superfamily: Underappreciated proteins and novel opportunities for cancer therapy. Mol Metab 2024; 84:101952. [PMID: 38705513 PMCID: PMC11112377 DOI: 10.1016/j.molmet.2024.101952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/24/2024] [Accepted: 04/27/2024] [Indexed: 05/07/2024] Open
Abstract
BACKGROUND Solute carrier (SLC) transporters, a diverse family of membrane proteins, are instrumental in orchestrating the intake and efflux of nutrients including amino acids, vitamins, ions, nutrients, etc, across cell membranes. This dynamic process is critical for sustaining the metabolic demands of cancer cells, promoting their survival, proliferation, and adaptation to the tumor microenvironment (TME). Amino acids are fundamental building blocks of cells and play essential roles in protein synthesis, nutrient sensing, and oncogenic signaling pathways. As key transporters of amino acids, SLCs have emerged as crucial players in maintaining cellular amino acid homeostasis, and their dysregulation is implicated in various cancer types. Thus, understanding the intricate connections between amino acids, SLCs, and cancer is pivotal for unraveling novel therapeutic targets and strategies. SCOPE OF REVIEW In this review, we delve into the significant impact of amino acid carriers of the SLCs family on the growth and progression of cancer and explore the current state of knowledge in this field, shedding light on the molecular mechanisms that underlie these relationships and highlighting potential avenues for future research and clinical interventions. MAJOR CONCLUSIONS Amino acids transportation by SLCs plays a critical role in tumor progression. However, some studies revealed the tumor suppressor function of SLCs. Although several studies evaluated the function of SLC7A11 and SLC1A5, the role of some SLC proteins in cancer is not studied well. To exert their functions, SLCs mediate metabolic rewiring, regulate the maintenance of redox balance, affect main oncogenic pathways, regulate amino acids bioavailability within the TME, and alter the sensitivity of cancer cells to therapeutics. However, different therapeutic methods that prevent the function of SLCs were able to inhibit tumor progression. This comprehensive review provides insights into a rapidly evolving area of cancer biology by focusing on amino acids and their transporters within the SLC superfamily.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
10
|
Fontana F, Giannitti G, Marchesi S, Limonta P. The PI3K/Akt Pathway and Glucose Metabolism: A Dangerous Liaison in Cancer. Int J Biol Sci 2024; 20:3113-3125. [PMID: 38904014 PMCID: PMC11186371 DOI: 10.7150/ijbs.89942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 04/11/2024] [Indexed: 06/22/2024] Open
Abstract
Aberrant activation of the PI3K/Akt pathway commonly occurs in cancers and correlates with multiple aspects of malignant progression. In particular, recent evidence suggests that the PI3K/Akt signaling plays a fundamental role in promoting the so-called aerobic glycolysis or Warburg effect, by phosphorylating different nutrient transporters and metabolic enzymes, such as GLUT1, HK2, PFKB3/4 and PKM2, and by regulating various molecular networks and proteins, including mTORC1, GSK3, FOXO transcription factors, MYC and HIF-1α. This leads to a profound reprogramming of cancer metabolism, also impacting on pentose phosphate pathway, mitochondrial oxidative phosphorylation, de novo lipid synthesis and redox homeostasis and thereby allowing the fulfillment of both the catabolic and anabolic demands of tumor cells. The present review discusses the interactions between the PI3K/Akt cascade and its metabolic targets, focusing on their possible therapeutic implications.
Collapse
Affiliation(s)
- Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | | | | | | |
Collapse
|
11
|
Song N, Ma C, Guo Y, Cui S, Chen S, Chen Z, Ling Y, Zhang Y, Liu H. Identifying differentially expressed genes in goat mammary epithelial cells induced by overexpression of SOCS3 gene using RNA sequencing. Front Vet Sci 2024; 11:1392152. [PMID: 38835896 PMCID: PMC11148363 DOI: 10.3389/fvets.2024.1392152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
The suppressor of cytokine signaling 3 (SOCS3) is a key signaling molecule that regulates milk synthesis in dairy livestock. However, the molecular mechanism by which SOCS3 regulates lipid synthesis in goat milk remains unclear. This study aimed to screen for key downstream genes associated with lipid synthesis regulated by SOCS3 in goat mammary epithelial cells (GMECs) using RNA sequencing (RNA-seq). Goat SOCS3 overexpression vector (PC-SOCS3) and negative control (PCDNA3.1) were transfected into GMECs. Total RNA from cells after SOCS3 overexpression was used for RNA-seq, followed by differentially expressed gene (DEG) analysis, functional enrichment analysis, and network prediction. SOCS3 overexpression significantly inhibited the synthesis of triacylglycerol, total cholesterol, non-esterified fatty acids, and accumulated lipid droplets. In total, 430 DEGs were identified, including 226 downregulated and 204 upregulated genes, following SOCS3 overexpression. Functional annotation revealed that the DEGs were mainly associated with lipid metabolism, cell proliferation, and apoptosis. We found that the lipid synthesis-related genes, STAT2 and FOXO6, were downregulated. In addition, the proliferation-related genes BCL2, MMP11, and MMP13 were upregulated, and the apoptosis-related gene CD40 was downregulated. In conclusion, six DEGs were identified as key regulators of milk lipid synthesis following SOCS3 overexpression in GMECs. Our results provide new candidate genes and insights into the molecular mechanisms involved in milk lipid synthesis regulated by SOCS3 in goats.
Collapse
Affiliation(s)
- Ning Song
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Cunxia Ma
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Yuzhu Guo
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Shuangshuang Cui
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Shihao Chen
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zhi Chen
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yinghui Ling
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Yunhai Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Hongyu Liu
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| |
Collapse
|
12
|
Bakinowska E, Kiełbowski K, Skórka P, Dach A, Olejnik-Wojciechowska J, Szwedkowicz A, Pawlik A. Non-Coding RNA as Biomarkers and Their Role in the Pathogenesis of Gastric Cancer-A Narrative Review. Int J Mol Sci 2024; 25:5144. [PMID: 38791187 PMCID: PMC11121563 DOI: 10.3390/ijms25105144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Non-coding RNAs (ncRNAs) represent a broad family of molecules that regulate gene expression, including microRNAs, long non-coding RNAs and circular RNAs, amongst others. Dysregulated expression of ncRNAs alters gene expression, which is implicated in the pathogenesis of several malignancies and inflammatory diseases. Gastric cancer is the fifth most frequently diagnosed cancer and the fourth most common cause of cancer-related death. Studies have found that altered expression of ncRNAs may contribute to tumourigenesis through regulating proliferation, apoptosis, drug resistance and metastasis. This review describes the potential use of ncRNAs as diagnostic and prognostic biomarkers. Moreover, we discuss the involvement of ncRNAs in the pathogenesis of gastric cancer, including their interactions with the members of major signalling pathways.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (K.K.); (P.S.); (A.D.); (J.O.-W.); (A.S.)
| |
Collapse
|
13
|
Ma G, Liu S, Cai F, Liang H, Deng J, Zhang R, Cai M. Ketohexokinase-A deficiency attenuates the proliferation via reducing β-catenin in gastric cancer cells. Exp Cell Res 2024; 438:114038. [PMID: 38614422 DOI: 10.1016/j.yexcr.2024.114038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 02/20/2024] [Accepted: 04/08/2024] [Indexed: 04/15/2024]
Abstract
Overconsumption of fructose is closely related to cancer. Ketohexokinase (KHK) catalyzes the conversion from fructose to fructose-1-phosphate (F1P), which is the first and committed step of fructose metabolism. Recently, aberrant KHK activation has been identified in multiple malignancies. However, the roles of KHK in gastric cancer (GC) cells are largely unclear. Herein, we reveal that the expression of ketohexokinase-A (KHK-A), one alternatively spliced KHK isoform that possesses low affinity for fructose, was markedly increased in GC cells. Depletion of endogenous KHK-A expression using lentiviruses encoding short hairpin RNAs (shRNAs) or pharmaceutical disruption of KHK-A activity using KHK-IN-1 hydrochloride in GC NCI-N87 and HGC-27 cells inhibited the proliferation in vitro and in vivo. Additionally, the mitochondrial respiration in the GC cells with KHK-A deficiency compared with the control cells was significantly impaired. One commercially-available antibody array was used to explore the effects of KHK-A knockdown on signaling pathways, showing that β-catenin was remarkably reduced in the KHK-A deficient GC cells compared with the control ones. Pharmaceutical reduction in β-catenin levels slowed down the proliferation of GC cells. These data uncover that KHK-A promotes the proliferation in GC cells, indicating that this enzyme might be a promising therapeutical target for GC treatment.
Collapse
Affiliation(s)
- Gang Ma
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, China; Tianjin Key Laboratory of Digestive Cancer, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Siya Liu
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, China; Tianjin Key Laboratory of Digestive Cancer, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Fenglin Cai
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, China; Tianjin Key Laboratory of Digestive Cancer, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Han Liang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, China; Tianjin Key Laboratory of Digestive Cancer, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jingyu Deng
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, China; Tianjin Key Laboratory of Digestive Cancer, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Rupeng Zhang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, China; Tianjin Key Laboratory of Digestive Cancer, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Mingzhi Cai
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, China; Tianjin Key Laboratory of Digestive Cancer, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
14
|
Orbach SM, DeVaull CY, Bealer EJ, Ross BC, Jeruss JS, Shea LD. An engineered niche delineates metastatic potential of breast cancer. Bioeng Transl Med 2024; 9:e10606. [PMID: 38193115 PMCID: PMC10771563 DOI: 10.1002/btm2.10606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/29/2023] [Accepted: 09/20/2023] [Indexed: 01/10/2024] Open
Abstract
Metastatic breast cancer is often not diagnosed until secondary tumors have become macroscopically visible and millions of tumor cells have invaded distant tissues. Yet, metastasis is initiated by a cascade of events leading to formation of the pre-metastatic niche, which can precede tumor formation by a matter of years. We aimed to distinguish the potential for metastatic disease from nonmetastatic disease at early times in triple-negative breast cancer using sister cell lines 4T1 (metastatic), 4T07 (invasive, nonmetastatic), and 67NR (nonmetastatic). We used a porous, polycaprolactone scaffold, that serves as an engineered metastatic niche, to identify metastatic disease through the characteristics of the microenvironment. Analysis of the immune cell composition at the scaffold was able to distinguish noninvasive 67NR tumor-bearing mice from 4T07 and 4T1 tumor-bearing mice but could not delineate metastatic potential between the two invasive cell lines. Gene expression in the scaffolds correlated with the up-regulation of cancer hallmarks (e.g., angiogenesis, hypoxia) in the 4T1 mice relative to 4T07 mice. We developed a 9-gene signature (Dhx9, Dusp12, Fth1, Ifitm1, Ndufs1, Pja2, Slc1a3, Soga1, Spon2) that successfully distinguished 4T1 disease from 67NR or 4T07 disease throughout metastatic progression. Furthermore, this signature proved highly effective at distinguishing diseased lungs in publicly available datasets of mouse models of metastatic breast cancer and in human models of lung cancer. The early and accurate detection of metastatic disease that could lead to early treatment has the potential to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Sophia M. Orbach
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | | | - Elizabeth J. Bealer
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Brian C. Ross
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Jacqueline S. Jeruss
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
- Department of SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Lonnie D. Shea
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of Chemical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
15
|
Ding Y, Zhang M, Hu S, Zhang C, Zhou Y, Han M, Li J, Li F, Ni H, Fang S, Chen Q. MiRNA-766-3p inhibits gastric cancer via targeting COL1A1 and regulating PI3K/AKT signaling pathway. J Cancer 2024; 15:990-998. [PMID: 38230216 PMCID: PMC10788715 DOI: 10.7150/jca.90321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/17/2023] [Indexed: 01/18/2024] Open
Abstract
Objective MiRNA-766-3p has been shown to be associated with a variety of cancers. However, few studies have been done in gastric cancer (GC). This study explores the mechanism of miR-766-3p in GC. Methods The potential targets of microRNA (miRNA) were predicted using Tarbase and Targetscan databases. The results are intersected with differential genes (DEGs) (fold change > 1.5, P < 0.05) in gastric cancer to obtain potential core targets. The hub targets screened by constructing PPI networks (degree > 5, expression > 0.5). Validating the differential expression and expression in immunohistochemistry of these targets through the database. And the binding sites between miRNAs and mRNAs were verified using dual-luciferase Assay. Finally, qRT-PCR and Western Blot experiments were conducted to validate the hub targets and signal pathways. Results The potential hub targets from the PPI network were THBS2, COL1A1, FGG, FGB, and PLAU. Combining database, luciferase Assay and experimental validation, miR-766-3p can sponge COL1A1 and it plays the most important role in gastric cancer progression. In GC, COL1A1 was upregulated and the enrichment analysis revealed that COL1A1 regulates PI3K/AKT signal pathway, and AKT is also highly expressed in gastric cancer. Conclusion The miR-766-3p can inhibit the progression of gastric cancer by targeting COL1A1 and regulating the PI3K/AKT signal pathway. It could be a potential therapy option for the GC.
Collapse
Affiliation(s)
- Yujie Ding
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mengyuan Zhang
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Sheng Hu
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Caiyun Zhang
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Yue Zhou
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Ming Han
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Jingjing Li
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Fulong Li
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Hongmei Ni
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shengquan Fang
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Qilong Chen
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, China
| |
Collapse
|
16
|
Wang L, Li Z, Li Z, Ren Y, Qian L, Yu Y, Shi W, Xiong Y. Identification of A Novel Gene Signature Combining Ferroptosis- and Immunity-Related Genes for Prognostic Prediction, Immunotherapy and Potential Therapeutic Targets in Gastric Cancer. J Cancer 2023; 14:3457-3476. [PMID: 38021154 PMCID: PMC10647194 DOI: 10.7150/jca.87223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/17/2023] [Indexed: 12/01/2023] Open
Abstract
Gastric cancer (GC) is one of the most prevalent cancers worldwide. Ferroptosis and the immune status of tumor tissue play vital roles in the initiation and progression of GC. However, the role and functional mechanisms of ferroptosis- and immunity-related genes (FIRGs) in GC pathogenesis and their correlations with GC prognosis have not been elucidated. We aim to establish a prognostic prediction model based on the FIRGs signature for GC patients. Differentially expressed genes were screened from the Cancer Genome Atlas (TCGA) GC cohorts. The least absolute shrinkage and selection operator (LASSO) regression was performed to establish a FIRGs-based risk model. This gene signature with 7 FIRGs was identified as an independent prognostic factor. A nomogram incorporating clinical parameters and the FIRG signature was constructed to individualize outcome predictions. Finally, we provided in vivo and in vitro evidence to verify the reliability of FIRG signature for GC prognosis, and validate the expression and function of FIRGs contributing to the development and progression of GC. Herein, our work represents great therapeutic and prognostic potentials for GC.
Collapse
Affiliation(s)
- Liwei Wang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Zhuozhuo Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Zi Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Yuanyuan Ren
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Department of Endocrinology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Wenzhen Shi
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Medical Research Center, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
| | - Yuyan Xiong
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| |
Collapse
|
17
|
Wan M, Dai J, Gan A, Wang J, Lin F, Zhang X, Lv X, Wu B, Yan T, Jia Y. A network pharmacology approach to investigate dehydrocostus lactone inhibits the proliferation and epithelial-mesenchymal transition of human gastric cancer cells via regulating the PI3K/Akt and extracellular signal-regulated kinases/mitogen-activated protein kinase signalling pathways. J Pharm Pharmacol 2023; 75:1344-1356. [PMID: 37403268 DOI: 10.1093/jpp/rgad065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/22/2023] [Indexed: 07/06/2023]
Abstract
OBJECTIVES Dehydrocostus lactone (DHE), a sesquiterpene lactone, has been proven the significant inhibition of multiple cancer cells. However, there are limited reports on the activity of DHE in gastric cancer (GC). In this research, Network pharmacology predicted the anti-GC mechanism of DHE, and the prediction was verified by in-vitro experiments. METHODS Network pharmacology confirmed the major effect signalling pathway of DHE in treating GC. Cell viability assay, colony formation assay, wound healing assay, cell migration and invasion assay, apoptosis assay, western blot and real-time quantitative polymerase chain reaction verified the mechanism of DHE in GC cell lines. KEY FINDINGS The results showed that DHE inhibited the growth and metastasis of MGC803 and AGS GC cells. Mechanistically, the analysis results indicated that DHE significantly induced the apoptosis process by suppressing the PI3K/protein kinase B (Akt) signalling pathway, and inhibited epithelial-mesenchymal transition by suppressing the extracellular signal-regulated kinases (ERK)/MAPK signalling pathway. The Akt activator (SC79) inhibited DHE induced apoptosis, and DHE had similar effects with the ERK inhibitor (FR180204). CONCLUSIONS All results suggested that DHE was a potential natural chemotherapeutic drug in GC treatment.
Collapse
Affiliation(s)
- Meiqi Wan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Jun Dai
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Anna Gan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Jinyu Wang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Fei Lin
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaoying Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Xinyan Lv
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Bo Wu
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Tingxu Yan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Ying Jia
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
18
|
Chen C, Ye L, Yi J, Liu T, Li Z. FN1 mediated activation of aspartate metabolism promotes the progression of triple-negative and luminal a breast cancer. Breast Cancer Res Treat 2023; 201:515-533. [PMID: 37458908 DOI: 10.1007/s10549-023-07032-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/28/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Breast cancer (BC) is regarded as one of the most common cancers diagnosed among the female population and has an extremely high mortality rate. It is known that Fibronectin 1 (FN1) drives the occurrence and development of a variety of cancers through metabolic reprogramming. Aspartic acid is considered to be an important substrate for nucleotide synthesis. However, the regulatory mechanism between FN1 and aspartate metabolism is currently unclear. METHODS We used RNA sequencing (RNA seq) and liquid chromatography-mass spectrometry to analyze the tumor tissues and paracancerous tissues of patients. MCF7 and MDA-MB-231 cells were used to explore the effects of FN1-regulated aspartic acid metabolism on cell survival, invasion, migration and tumor growth. We used PCR, Western blot, immunocytochemistry and immunofluorescence techniques to study it. RESULTS We found that FN1 was highly expressed in tumor tissues, especially in Lumina A and TNBC subtypes, and was associated with poor prognosis. In vivo and in vitro experiments showed that silencing FN1 inhibits the activation of the YAP1/Hippo pathway by enhancing YAP1 phosphorylation, down-regulates SLC1A3-mediated aspartate uptake and utilization by tumor cells, inhibits BC cell proliferation, invasion and migration, and promotes apoptosis. In addition, inhibition of FN1 combined with the YAP1 inhibitor or SLC1A3 inhibitor can effectively inhibit tumor growth, of which inhibition of FN1 combined with the YAP1 inhibitor is more effective. CONCLUSION Targeting the "FN1/YAP1/SLC1A3/Aspartate metabolism" regulatory axis provides a new target for BC diagnosis and treatment. This study also revealed that intratumoral metabolic heterogeneity plays an important role in the progression of different subtypes of breast cancer.
Collapse
Affiliation(s)
- Chen Chen
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Leiguang Ye
- Department of Respiratory Medicine, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Jinfeng Yi
- Department of Pathology, Harbin Medical University, Harbin, 150081, China
| | - Tang Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| | - Zhigao Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
19
|
Byerly CD, Patterson LL, Pittner NA, Solomon RN, Patel JG, Rogan MR, McBride JW. Ehrlichia Wnt SLiM ligand mimic deactivates the Hippo pathway to engage the anti-apoptotic Yap-GLUT1-BCL-xL axis. Infect Immun 2023; 91:e0008523. [PMID: 37530530 PMCID: PMC10501218 DOI: 10.1128/iai.00085-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/03/2023] [Indexed: 08/03/2023] Open
Abstract
Ehrlichia chaffeensis TRP120 effector has evolved short linear motif (SLiM) ligand mimicry to repurpose multiple evolutionarily conserved cellular signaling pathways, including Wnt, Notch, and Hedgehog. In this investigation, we demonstrate that E. chaffeensis and recombinant TRP120 deactivate Hippo signaling, resulting in the activation of Hippo transcription coactivator Yes-associated protein (Yap). Moreover, a homologous 6 amino acid (QDVASH) SLiM shared by TRP120 and Wnt3a/5a ligands phenocopied Yap and β-catenin activation induced by E. chaffeensis, rTRP120, and Wnt5a. Similar Hippo gene expression profiles were also stimulated by E. chaffeensis, rTRP120, SLiM, and Wnt5a. Single siRNA knockdown of Hippo transcription co-activator/factors, Yap, and transcriptional enhanced associate domain (TEAD) significantly decreased E. chaffeensis infection. Yap activation was abolished in THP-1 Wnt Frizzled-5 (Fzd5) receptor knockout cells (KO), demonstrating Fzd5 receptor dependence. In addition, the TRP120-Wnt-SLiM antibody blocked Hippo deactivation (Yap activation). Expression of anti-apoptotic Hippo target gene SLC2A1 (encodes glucose transporter 1; GLUT1) was upregulated by E. chaffeensis and corresponded to increased levels of GLUT1. Conversely, siRNA knockdown of SLC2A1 significantly inhibited infection. Higher GLUT1 levels correlated with increased B cell lymphoma-extra large (BCL-xL) and decreased BCL2-associated X, apoptosis regulator (Bax) levels. Moreover, blocking Yap activation with the inhibitor Verteporfin induced apoptosis that corresponded to significant reductions in GLUT1 and BCL-xL levels and activation of Bax and Caspase-3 and -9. This study identifies a novel shared Wnt/Hippo SLiM ligand mimic and demonstrates that E. chaffeensis deactivates the Hippo pathway to engage the anti-apoptotic Yap-GLUT1-BCL-xL axis.
Collapse
Affiliation(s)
- Caitlan D. Byerly
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - LaNisha L. Patterson
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Nicholas A. Pittner
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Regina N. Solomon
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jignesh G. Patel
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Madison R. Rogan
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jere W. McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
20
|
Ling ZN, Jiang YF, Ru JN, Lu JH, Ding B, Wu J. Amino acid metabolism in health and disease. Signal Transduct Target Ther 2023; 8:345. [PMID: 37699892 PMCID: PMC10497558 DOI: 10.1038/s41392-023-01569-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/12/2023] [Accepted: 07/13/2023] [Indexed: 09/14/2023] Open
Abstract
Amino acids are the building blocks of protein synthesis. They are structural elements and energy sources of cells necessary for normal cell growth, differentiation and function. Amino acid metabolism disorders have been linked with a number of pathological conditions, including metabolic diseases, cardiovascular diseases, immune diseases, and cancer. In the case of tumors, alterations in amino acid metabolism can be used not only as clinical indicators of cancer progression but also as therapeutic strategies. Since the growth and development of tumors depend on the intake of foreign amino acids, more and more studies have targeted the metabolism of tumor-related amino acids to selectively kill tumor cells. Furthermore, immune-related studies have confirmed that amino acid metabolism regulates the function of effector T cells and regulatory T cells, affecting the function of immune cells. Therefore, studying amino acid metabolism associated with disease and identifying targets in amino acid metabolic pathways may be helpful for disease treatment. This article mainly focuses on the research of amino acid metabolism in tumor-oriented diseases, and reviews the research and clinical research progress of metabolic diseases, cardiovascular diseases and immune-related diseases related to amino acid metabolism, in order to provide theoretical basis for targeted therapy of amino acid metabolism.
Collapse
Affiliation(s)
- Zhe-Nan Ling
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, P.R. China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang Province, P.R. China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang Province, P.R. China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, P.R. China
| | - Yi-Fan Jiang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, P.R. China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang Province, P.R. China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang Province, P.R. China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, P.R. China
| | - Jun-Nan Ru
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, P.R. China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang Province, P.R. China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang Province, P.R. China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, P.R. China
| | - Jia-Hua Lu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, P.R. China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang Province, P.R. China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang Province, P.R. China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, P.R. China
| | - Bo Ding
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, P.R. China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang Province, P.R. China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang Province, P.R. China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, P.R. China
| | - Jian Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, P.R. China.
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang Province, P.R. China.
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang Province, P.R. China.
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, P.R. China.
| |
Collapse
|
21
|
Wang HY, Chen Y, Zhu SM. Regulatory effect of tetramethylpyrazine on cell proliferation, migration, invasion, and glycolysis in gastric cancer cells. Shijie Huaren Xiaohua Zazhi 2023; 31:485-491. [DOI: 10.11569/wcjd.v31.i12.485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/26/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Tetramethylpyrazine (TMP) has been reported to have antitumor effects, but its effect on gastric cancer and the underlying mechanism are not fully understood.
AIM To investigate the regulatory effect and potential mechanism of TMP on the proliferation, migration, invasion, and glycolysis of gastric cancer cells.
METHODS Gastric cancer cells cultured in vitro were randomly divided into four groups: Control group (Ctrl), low-dose TMP group (TMP-L, 10 μM), medium-dose TMP group (TMP-M, 20 μM), and high-dose TMP group (TMP-H, 40 μM). After treatment, cell viability was detected by CCK-8 assay, and cell proliferation, migration, and invasion were detected by colony formation assay and transwell assay. Glucose metabolism was determined by measurements of glucose uptake, lactate production, oxygen consumption rate (OCR), and extracellular acidification rate (ECAR). The activity of hexokinase (HK) and lactate dehydrogenase (LDH) was determined. The expression of glycolysis-related proteins and activation of the protein kinase B (AKT)/glucose trans-porter-1 (GLUT1) axis were assessed by Western blot assay.
RESULTS TMP reduced the viability of gastric cancer cells in a dose-dependent manner (P < 0.05). Compared to the Ctrl group, medium- and high-dose TMP inhibited cell proliferation, migration, and invasion (P < 0.05). After treatment with TMP, glucose uptake, lactate production, OCR, and ECAR were decreased significantly (P < 0.05), and the activity of HK and LDH was reduced significantly in the TMP-H group (P < 0.05). Western blot analysis showed that the expression of p-AKT/AKT, GLUT1, HK2, and LDHA in gastric cancer cells treated with TMP was downregulated (P < 0.05).
CONCLUSION TMP reduces glycolysis and inhibits cell proliferation and migration in gartric cancer cells.
Collapse
Affiliation(s)
- Han-Ying Wang
- Department of Tumor Chemoradiotherapy, Lishui People's Hospital, Lishui 323000, Zhejiang Province, China
| | - Yong Chen
- Department of Tumor Chemoradiotherapy, Lishui People's Hospital, Lishui 323000, Zhejiang Province, China
| | - Shuang-Mei Zhu
- Department of Tumor Chemoradiotherapy, Lishui People's Hospital, Lishui 323000, Zhejiang Province, China
| |
Collapse
|
22
|
Niu P, Liu F, Lei F, Peng J, Wang Y, Zhao J, Gao Z, Gao Q, Gu J. Breviscapine regulates the proliferation, migration, invasion, and apoptosis of colorectal cancer cells via the PI3K/AKT pathway. Sci Rep 2023; 13:9674. [PMID: 37316553 DOI: 10.1038/s41598-023-33792-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 04/19/2023] [Indexed: 06/16/2023] Open
Abstract
Colorectal cancer (CRC) is ranked as one of the most common malignancies with a high death rate. It has been discovered that breviscapine can alter the progression and development of various cancers. Nevertheless, the function and mechanisms of breviscapine in CRC progression have not yet been described. The cell proliferation capacity of HCT116 and SW480 cells was assessed using the CCK-8 and EdU assays. Cell apoptosis was tested through flow cytometry, and cell migration and invasion were examined using the transwell assay. Moreover, protein expression was examined through a western blot. Tumor weight and volume were assessed using the nude mice in vivo assay, and the Ki-67 protein expression was verified through the IHC assay. This study discovered that an increased dose of breviscapine (0, 12.5, 25, 50, 100, 200, and 400 μM) gradually reduced cell proliferation and increased apoptosis in CRC. Additionally, breviscapine restricted the migration and invasion CRC cells. Moreover, it was revealed that breviscapine inactivated the PI3K/AKT pathway and inhibited CRC progression. Finally, an in vivo assay demonstrated that breviscapine restrained tumor growth in vivo. It affected the CRC cells' proliferation, migration, invasion, and apoptosis through the PI3K/AKT pathway. This discovery may offer new insights into CRC treatment.
Collapse
Affiliation(s)
- Pengfei Niu
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No. 9, Jinyuanzhuang Road, The Shijingshan District, Beijing, 100144, China
| | - Feng Liu
- Beijing Viewsolid Biotechnology Co., Ltd., Beijing, 100195, China
| | - Fuming Lei
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No. 9, Jinyuanzhuang Road, The Shijingshan District, Beijing, 100144, China
| | - Jisheng Peng
- Department of Traditional Chinese Medicine, Peking University Shougang Hospital, Beijing, 100144, China
| | - Yanzhao Wang
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No. 9, Jinyuanzhuang Road, The Shijingshan District, Beijing, 100144, China
| | - Jun Zhao
- Department of Traditional Chinese Medicine, Peking University Shougang Hospital, Beijing, 100144, China
| | - Zhaoya Gao
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No. 9, Jinyuanzhuang Road, The Shijingshan District, Beijing, 100144, China
| | - Qingkun Gao
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No. 9, Jinyuanzhuang Road, The Shijingshan District, Beijing, 100144, China
| | - Jin Gu
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No. 9, Jinyuanzhuang Road, The Shijingshan District, Beijing, 100144, China.
| |
Collapse
|
23
|
Quiroga J, Alarcón P, Ramírez MF, Manosalva C, Teuber S, Carretta MD, Burgos RA. d-lactate-induced ETosis in cattle polymorphonuclear leucocytes is dependent on the release of mitochondrial reactive oxygen species and the PI3K/Akt/HIF-1 and GSK-3β pathways. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 145:104728. [PMID: 37164278 DOI: 10.1016/j.dci.2023.104728] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/12/2023]
Abstract
d-lactate is a metabolite originating from bacterial metabolism that accumulates as a result of dietary disturbances in cattle, leading to ruminal acidosis. d-lactate exerts functions as a metabolic signal inducing metabolic reprogramming and extracellular trap (ET) release in polymorphonuclear leucocytes (PMNs). We previously demonstrated that d-lactate induces metabolic reprogramming via hypoxia-induced factor 1 alpha (HIF-1α) stabilization in bovine fibroblast-like synoviocytes (FLSs). In the present study, the role of HIF-1 in ET formation induced by d-lactate was assessed. HIF-1α stabilization in PMNs was controlled by mitochondrial reactive oxygen species (mtROS) release. Furthermore, inhibition of mitochondrial complex I and scavenging of mtROS decreased d-lactate-triggered ETosis. d-lactate-enhanced HIF-1α accumulation was dependent on the PI3K/Akt pathway but independent of GSK-3β activity. Pharmacological blockade of the PI3K/Akt/HIF-1 and GSK-3β axes inhibited d-lactate-triggered ETosis and downregulated PDK1 and LDHA expression. However, only GSK-3β inhibition decreased the expression of glycogen metabolism enzymes and prevented the decline in glycogen stores induced by d-lactate exposure. The results of this study suggest that mtROS, PI3K/Akt/HIF-1 and GSK-3β axes regulate carbohydrate metabolism adaptations that support d-lactate-induced ET formation in cattle.
Collapse
Affiliation(s)
- John Quiroga
- Laboratory of Inflammation Pharmacology and Immunometabolism, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Pablo Alarcón
- Laboratory of Inflammation Pharmacology and Immunometabolism, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - María Fernanda Ramírez
- Laboratory of Inflammation Pharmacology and Immunometabolism, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Carolina Manosalva
- Institute of Pharmacy, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Stefanie Teuber
- Laboratory of Inflammation Pharmacology and Immunometabolism, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - María Daniella Carretta
- Laboratory of Inflammation Pharmacology and Immunometabolism, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Rafael Agustín Burgos
- Laboratory of Inflammation Pharmacology and Immunometabolism, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
24
|
Byerly CD, Patterson LL, Pittner NA, Solomon RN, Patel JG, Rogan MR, McBride JW. Ehrlichia Wnt short linear motif ligand mimetic deactivates the Hippo pathway to engage the anti-apoptotic Yap-GLUT1-BCL-xL axis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531456. [PMID: 36945589 PMCID: PMC10028901 DOI: 10.1101/2023.03.06.531456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Ehrlichia chaffeensis TRP120 effector has evolved short linear motif (SLiM) ligand mimicry to repurpose multiple evolutionarily conserved cellular signaling pathways including Wnt, Notch and Hedgehog. In this investigation, we demonstrate that E. chaffeensis and recombinant TRP120 deactivate Hippo signaling resulting in activation of Hippo transcription coactivator Yap and target gene expression. Moreover, a homologous 6 amino acid (QDVASH) SLiM shared by TRP120 and Wnt3a/5a ligands phenocopied Yap and β-catenin activation induced by E. chaffeensis, rTRP120 and Wnt5a. Similar Hippo gene expression profiles were also stimulated by E. chaffeensis, rTRP120, SLiM and Wnt5a. Single siRNA knockdown of Hippo transcription co-activator/factors (Yap and TEAD) significantly decreased E. chaffeensis infection. Yap activation was abolished in THP-1 Wnt Frizzled-5 (Fzd5) receptor knockout cells (KO), demonstrating Fzd5 receptor dependence. In addition, TRP120 Wnt-SLiM antibody blocked Hippo deactivation (Yap activation). Expression of anti-apoptotic Hippo target gene SLC2A1 (encodes glucose transporter 1; GLUT1) was upregulated by E. chaffeensis and corresponded to increased levels of GLUT1. Conversely, siRNA knockdown of SLC2A1 significantly inhibited infection. Higher GLUT1 levels correlated with increased BCL-xL and decreased Bax levels. Moreover, blocking Yap activation with the inhibitor Verteporfin induced apoptosis that corresponded to significant reductions in levels of GLUT1 and BCL-xL, and activation of Bax and Caspase-3 and -9. This study identifies a novel shared Wnt/Hippo SLiM ligand mimetic and demonstrates that E. chaffeensis deactivates the Hippo pathway to engage the anti-apoptotic Yap-GLUT1-BCL-xL axis.
Collapse
Affiliation(s)
- Caitlan D. Byerly
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - LaNisha L. Patterson
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Nicholas A. Pittner
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Regina N. Solomon
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jignesh G. Patel
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Madison R. Rogan
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jere W. McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Department Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
25
|
Shen J, Yang Z, Wu X, Yao G, Hou M. Baicalein facilitates gastric cancer cell apoptosis by triggering endoplasmic reticulum stress via repression of the PI3K/AKT pathway. APPLIED BIOLOGICAL CHEMISTRY 2023; 66:10. [PMID: 36815904 PMCID: PMC9924871 DOI: 10.1186/s13765-022-00759-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/19/2022] [Indexed: 06/18/2023]
Abstract
OBJECTIVE Gastric cancer (GC) remains a prevailing threat to life. Baicalein exhibits anti-cancer properties. This study estimated the mechanism of baicalein in GC cell apoptosis by mediating endoplasmic reticulum stress (ERS) through the PI3K/AKT pathway. METHODS After treatment with different concentrations of baicalein, GC cell (HGC-27 and AGS) viability was detected by MTT assay. AGS cells more sensitive to baicalein treatment were selected as study subjects. The IC50 of baicalein on AGS cells was determined. Colony formation, cell cycle, and apoptosis were detected using crystal violet staining and flow cytometry. Levels of ERS-related and BTG3/PI3K/AKT pathway-related proteins were determined by Western blot. Intracellular Ca2+ level was measured using Fluo-3 AM fluorescence working solution. GC mouse models were established by subcutaneously injecting AGS cells into the right rib and were intragastrically administrated with baicalein. Tumor volume and weight were recorded. Expression of Ki67 in tumor tissues and positive expression of apoptotic cells were detected by immunohistochemistry and TUNEL staining. RESULTS Baicalein inhibited cell proliferation and induced G0/G1 arrest and apoptosis by regulating the cell cycle, and triggered ERS in GC cells. Baicalein impeded the PI3K/AKT pathway by activating BTG3, thereby triggering ERS and inducing apoptosis. BTG3 inhibition reversed baicalein-induced apoptosis and ERS. Baicalein regulated GC cells in a concentration-dependent manner. Moreover, in xenograft mice, baicalein prevented tumor growth, decreased Ki67-positive cells, activated BTG3, and inhibited the PI3K/AKT pathway, thus activating ERS and increasing apoptotic cells. CONCLUSION Baicalein facilitates GC cell apoptosis by triggering ERS via repression of the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Junjie Shen
- Nanjing University of Chinese Medicine, Nanjing, 210029 Jiangsu province China
- Gastrointestinal Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050 Inner Mongolia China
| | - Zhiwen Yang
- Gastrointestinal Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050 Inner Mongolia China
| | - Xinlin Wu
- Gastrointestinal Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050 Inner Mongolia China
| | - Guodong Yao
- Gastrointestinal Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050 Inner Mongolia China
| | - Mingxing Hou
- Nanjing University of Chinese Medicine, Nanjing, 210029 Jiangsu province China
- Gastrointestinal Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050 Inner Mongolia China
- The Affiliated Hospital of Inner Mongolia Medical University, No. 1, Datong North Street, Huimin District, 010050 Hohhot, Inner Mongolia China
| |
Collapse
|
26
|
Haq MFU, Hussain MZ, Mahjabeen I, Akram Z, Saeed N, Shafique R, Abbasi SF, Kayani MA. Oncometabolic role of mitochondrial sirtuins in glioma patients. PLoS One 2023; 18:e0281840. [PMID: 36809279 PMCID: PMC9943017 DOI: 10.1371/journal.pone.0281840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/31/2023] [Indexed: 02/23/2023] Open
Abstract
Mitochondrial sirtuins have diverse role specifically in aging, metabolism and cancer. In cancer, these sirtuins play dichotomous role as tumor suppressor and promoter. Previous studies have reported the involvement of sirtuins in different cancers. However, till now no study has been published with respect to mitochondrial sirtuins and glioma risks. Present study was purposed to figure out the expression level of mitochondrial sirtuins (SIRT3, SIRT4, SIRT5) and related genes (GDH, OGG1-2α, SOD1, SOD2, HIF1α and PARP1) in 153 glioma tissue samples and 200 brain tissue samples from epilepsy patients (taken as controls). To understand the role of selected situins in gliomagenesis, DNA damage was measured using the comet assay and oncometabolic role (oxidative stress level, ATP level and NAD level) was measured using the ELISA and quantitative PCR. Results analysis showed significant down-regulation of SIRT4 (p = 0.0337), SIRT5 (p<0.0001), GDH (p = 0.0305), OGG1-2α (p = 0.0001), SOD1 (p<0.0001) and SOD2 (p<0.0001) in glioma patients compared to controls. In case of SIRT3 (p = 0.0322), HIF1α (p = 0.0385) and PARP1 (p = 0.0203), significant up-regulation was observed. ROC curve analysis and cox regression analysis showed the good diagnostic and prognostic value of mitochondrial sirtuins in glioma patients. Oncometabolic rate assessment analysis showed significant increased ATP level (p<0.0001), NAD+ level [(NMNAT1 (p<0.0001), NMNAT3 (p<0.0001) and NAMPT (p<0.04)] and glutathione level (p<0.0001) in glioma patients compared to controls. Significant increased level of damage ((p<0.04) and decrease level of antioxidant enzymes include superoxide dismutase (SOD, p<0.0001), catalase (CAT, p<0.0001) and glutathione peroxidase (GPx, p<0.0001) was observed in patients compared to controls. Present study data suggest that variation in expression pattern of mitochondrial sirtuins and increased metabolic rate may have diagnostic and prognostic significance in glioma patients.
Collapse
Affiliation(s)
- Maria Fazal Ul Haq
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | | | - Ishrat Mahjabeen
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
- * E-mail:
| | - Zertashia Akram
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Nadia Saeed
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Rabia Shafique
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Sumaira Fida Abbasi
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Mahmood Akhtar Kayani
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| |
Collapse
|
27
|
De Falco P, Lazzarino G, Felice F, Desideri E, Castelli S, Salvatori I, Ciccarone F, Ciriolo MR. Hindering NAT8L expression in hepatocellular carcinoma increases cytosolic aspartate delivery that fosters pentose phosphate pathway and purine biosynthesis promoting cell proliferation. Redox Biol 2022; 59:102585. [PMID: 36580805 PMCID: PMC9813579 DOI: 10.1016/j.redox.2022.102585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/05/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
N-acetylaspartate (NAA) is synthesized by the mitochondrial enzyme NAT8L, which uses acetyl-CoA and aspartate as substrates. These metabolites are fundamental for bioenergetics and anabolic requirements of highly proliferating cells, thus, NAT8L modulation may impinge on the metabolic reprogramming of cancer cells. Specifically, aspartate represents a limiting amino acid for nucleotide synthesis in cancer. Here, the expression of the NAT8L enzyme was modulated to verify how it impacts the metabolic adaptations and proliferative capacity of hepatocellular carcinoma. We demonstrated that NAT8L downregulation is associated with increased proliferation of hepatocellular carcinoma cells and immortalized hepatocytes. The overexpression of NAT8L instead decreased cell growth. The pro-tumoral effect of NAT8L silencing depended on glutamine oxidation and the rewiring of glucose metabolism. Mechanistically, NAT8L downregulation triggers aspartate outflow from mitochondria via the exporter SLC25A13 to promote glucose flux into the pentose phosphate pathway, boosting purine biosynthesis. These results were corroborated by the analyses of human and mouse hepatocellular carcinoma samples revealing a decrease in NAT8L expression compared to adjacent non-tumoral tissues. Overall, this work demonstrates that NAT8L expression in liver cells limits the cytosolic availability of aspartate necessary for enhancing the pentose phosphate pathway and purine biosynthesis, counteracting cell proliferation.
Collapse
Affiliation(s)
- Pamela De Falco
- Department of Biology, University of Rome “Tor Vergata”, Via Della Ricerca Scientifica, 00133, Rome, Italy
| | - Giacomo Lazzarino
- UniCamillus-Saint Camillus International University of Health and Medical Sciences, Via di Sant'Alessandro 8, 00131, Rome, Italy
| | - Federica Felice
- Department of Biology, University of Rome “Tor Vergata”, Via Della Ricerca Scientifica, 00133, Rome, Italy
| | - Enrico Desideri
- IRCCS San Raffaele Roma, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta, 247, 00166, Rome, Italy
| | - Serena Castelli
- IRCCS San Raffaele Roma, Via di Val Cannuta, 247, 00166, Rome, Italy
| | - Illari Salvatori
- IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano 64, Rome, 00143, Italy,Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Fabio Ciccarone
- Department of Biology, University of Rome "Tor Vergata", Via Della Ricerca Scientifica, 00133, Rome, Italy; IRCCS San Raffaele Roma, Via di Val Cannuta, 247, 00166, Rome, Italy.
| | - Maria Rosa Ciriolo
- Department of Biology, University of Rome "Tor Vergata", Via Della Ricerca Scientifica, 00133, Rome, Italy; IRCCS San Raffaele Roma, Via di Val Cannuta, 247, 00166, Rome, Italy.
| |
Collapse
|
28
|
Zhou Q, Liu S, Kou Y, Yang P, Liu H, Hasegawa T, Su R, Zhu G, Li M. ATP Promotes Oral Squamous Cell Carcinoma Cell Invasion and Migration by Activating the PI3K/AKT Pathway via the P2Y2-Src-EGFR Axis. ACS OMEGA 2022; 7:39760-39771. [PMID: 36385800 PMCID: PMC9648055 DOI: 10.1021/acsomega.2c03727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
Oral cancer is one of the most common malignancies of the head and neck, and approximately 90% of oral cancers are oral squamous cell carcinomas (OSCCs). The purinergic P2Y2 receptor is upregulated in breast cancer, pancreatic cancer, colorectal cancer, and liver cancer, but its role in OSCC is still unclear. Here, we examined the effects of P2Y2 on the invasion and migration of oral cancer cells (SCC15 and CAL27). The BALB/c mouse model was used to observe the involvement of P2Y2 with tumors in vivo. P2Y2, Src, and EGFR are highly expressed in OSCC tissues and cell lines. Stimulation with ATP significantly enhanced cell invasion and migration in oral cancer cells, and enhanced the activity of Src and EGFR protein kinases, which is mediated by the PI3K/AKT signaling pathway. P2Y2 knockdown attenuated the above ATP-driven events in vitro and in vivo. The PI3K/AKT signaling pathway was blocked by Src or EGFR inhibitor. Extracellular ATP activates the PI3K/AKT pathway through the P2Y2-Src-EGFR axis to promote OSCC invasion and migration, and thus, P2Y2 may be a potential novel target for antimetastasis therapy.
Collapse
Affiliation(s)
- Qin Zhou
- Department
of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College
of Medicine, Shandong University and Shandong
Key Laboratory of Oral Tissue Regeneration and Shandong Engineering
Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan 250100, China
- Center
of Osteoporosis and Bone Mineral Research, Shandong University, Jinan 250100, China
| | - Shanshan Liu
- Department
of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College
of Medicine, Shandong University and Shandong
Key Laboratory of Oral Tissue Regeneration and Shandong Engineering
Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan 250100, China
- Center
of Osteoporosis and Bone Mineral Research, Shandong University, Jinan 250100, China
| | - Yuying Kou
- Department
of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College
of Medicine, Shandong University and Shandong
Key Laboratory of Oral Tissue Regeneration and Shandong Engineering
Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan 250100, China
- Center
of Osteoporosis and Bone Mineral Research, Shandong University, Jinan 250100, China
| | - Panpan Yang
- Department
of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College
of Medicine, Shandong University and Shandong
Key Laboratory of Oral Tissue Regeneration and Shandong Engineering
Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan 250100, China
- Center
of Osteoporosis and Bone Mineral Research, Shandong University, Jinan 250100, China
| | - Hongrui Liu
- Department
of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College
of Medicine, Shandong University and Shandong
Key Laboratory of Oral Tissue Regeneration and Shandong Engineering
Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan 250100, China
- Center
of Osteoporosis and Bone Mineral Research, Shandong University, Jinan 250100, China
| | - Tomoka Hasegawa
- Department
of Developmental Biology of Hard Tissue, Graduate School of Dental
Medicine, Hokkaido University, Sapporo 060-0808, Japan
| | - Rongjian Su
- College
of Basic Medicine of Jinzhou Medical University, Cell Biology and
Genetic Department of Jinzhou Medical University, Key Lab of Molecular
and Cellular Biology of the Education Department of Liaoning Province, Life Science Institute of Jinzhou Medical University, Jinzhou 121001, China
| | - Guoxiong Zhu
- Department
of Stomatology, No.960 Hospital of PLA, No. 25 Shifan Road, Jinan 250014, China
| | - Minqi Li
- Department
of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College
of Medicine, Shandong University and Shandong
Key Laboratory of Oral Tissue Regeneration and Shandong Engineering
Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan 250100, China
- Center
of Osteoporosis and Bone Mineral Research, Shandong University, Jinan 250100, China
| |
Collapse
|
29
|
Shao W, Liu L, Zheng F, Ma Y, Zhang J. The potent role of Src kinase-regulating glucose metabolism in cancer. Biochem Pharmacol 2022; 206:115333. [DOI: 10.1016/j.bcp.2022.115333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/02/2022]
|
30
|
Yu Z, Liang C, Tu H, Qiu S, Dong X, Zhang Y, Ma C, Li P. Common Core Genes Play Vital Roles in Gastric Cancer With Different Stages. Front Genet 2022; 13:881948. [PMID: 35938042 PMCID: PMC9352954 DOI: 10.3389/fgene.2022.881948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/31/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Owing to complex molecular mechanisms in gastric cancer (GC) oncogenesis and progression, existing biomarkers and therapeutic targets could not significantly improve diagnosis and prognosis. This study aims to identify the key genes and signaling pathways related to GC oncogenesis and progression using bioinformatics and meta-analysis methods. Methods: Eligible microarray datasets were downloaded and integrated using the meta-analysis method. According to the tumor stage, GC gene chips were classified into three groups. Thereafter, the three groups’ differentially expressed genes (DEGs) were identified by comparing the gene data of the tumor groups with those of matched normal specimens. Enrichment analyses were conducted based on common DEGs among the three groups. Then protein–protein interaction (PPI) networks were constructed to identify relevant hub genes and subnetworks. The effects of significant DEGs and hub genes were verified and explored in other datasets. In addition, the analysis of mutated genes was also conducted using gene data from The Cancer Genome Atlas database. Results: After integration of six microarray datasets, 1,229 common DEGs consisting of 1,065 upregulated and 164 downregulated genes were identified. Alpha-2 collagen type I (COL1A2), tissue inhibitor matrix metalloproteinase 1 (TIMP1), thymus cell antigen 1 (THY1), and biglycan (BGN) were selected as significant DEGs throughout GC development. The low expression of ghrelin (GHRL) is associated with a high lymph node ratio (LNR) and poor survival outcomes. Thereafter, we constructed a PPI network of all identified DEGs and gained 39 subnetworks and the top 20 hub genes. Enrichment analyses were performed for common DEGs, the most related subnetwork, and the top 20 hub genes. We also selected 61 metabolic DEGs to construct PPI networks and acquired the relevant hub genes. Centrosomal protein 55 (CEP55) and POLR1A were identified as hub genes associated with survival outcomes. Conclusion: The DEGs, hub genes, and enrichment analysis for GC with different stages were comprehensively investigated, which contribute to exploring the new biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Zhiyuan Yu
- School of Medicine, Nankai University, Tianjin, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Chen Liang
- First Department of Liver Disease / Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing You’an Hospital, Capital Medical University, Beijing, China
| | - Huaiyu Tu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shuzhong Qiu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaoyu Dong
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yonghui Zhang
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Chao Ma
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Peiyu Li
- School of Medicine, Nankai University, Tianjin, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Peiyu Li,
| |
Collapse
|
31
|
Zhao Y, Liu H, Fan M, Miao Y, Zhao X, Wei Q, Ma B. G protein-coupled receptor 30 mediates cell proliferation of goat mammary epithelial cells via MEK/ERK&PI3K/AKT signaling pathway. Cell Cycle 2022; 21:2027-2037. [PMID: 35659445 DOI: 10.1080/15384101.2022.2083708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The mammary gland of mammals possesses the specific function of synthesizing, secreting, and delivering milk. Notably, mammary epithelial cells are considered to be central to control the expansion and remodeling of mammary gland into a milk-secretory organ. And the biological function of mammary gland is mainly regulated by the endocrine system, especially for estrogen. G protein-coupled receptor 30 (GPR30), an estrogen membrane receptor, mediates estrogen-induced functions of physiology and pathophysiology. However, the relationship between estrogen/GPR30 signaling and proliferation of goat mammary epithelial cells (gMECs) is still unclear. Herein, estrogen promoted cell proliferation than control, as evidence by upregulation of cell numbers, BrdU-positive cell counts, and cell viability. Of note, these activities were all obviously reduced by treatment with GPR30 antagonist G15, yet GPR30 agonist G1 increased cell proliferation than control. Further, GPR30 silencing inhibited cell proliferation than negative control. This inhibition was accompanied by a G2/M phase arrest and downregulation of cell cycle regulators. Meanwhile, estrogen increased the phosphorylation of ERK1/2 and AKT. Further, the protein level of p-ERK1/2 and p-AKT was enhanced by GPR30 agonist G1 but inhibited by GPR30 antagonist G15 and GPR30 silencing. Importantly, MEK inhibitor and PI3K inhibitor decreased the expression of cell cycle regulators, and repressed estrogen-induced and G1-driven promotion of cell proliferation, suggesting that estrogen regulated cell proliferation of gMECs through mechanisms involving cell cycle, dependent of GPR30 and MEK/ERK and PI3K/AKT signaling pathway. This may provide a strong theoretical basis for researching estrogen sustained-release drugs promoting breast development and improving lactation performance.Abbreviations: gMECs, goat mammary epithelial cells; E2, 17β-estradiol; GPR30, G protein-coupled receptor 30; shRNA, small hairpin RNA; CDK, cyclin-dependent kinase; PI3K, phosphatidylinositol 3-kinase; AKT, proteinkinase B; MAPK, mitogen-activated protein kinase; MEK, mitogen-activated protein kinase kinase; ERK1/2, extracellular signal-regulated kinase 1/2.
Collapse
Affiliation(s)
- Ying Zhao
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Haokun Liu
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Mingzhen Fan
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuyang Miao
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaoe Zhao
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Qing Wei
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Baohua Ma
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
32
|
Yang L, Liu D, Yan H, Chen K. Dapagliflozin attenuates cholesterol overloading-induced injury in mice hepatocytes with type 2 diabetes mellitus (T2DM) via eliminating oxidative damages. Cell Cycle 2022; 21:641-654. [PMID: 35100086 PMCID: PMC8942414 DOI: 10.1080/15384101.2022.2031429] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Cholesterol overloading-induced damages on hepatocytes cause liver dysfunctions, which further damages cholesterol metabolism and results in visceral fat accumulation in patients with type 2 diabetes mellitus (T2DM). The sodium-glucose cotransporter 2 (SGLT2) inhibitor Dapagliflozin has been reported to regulate cholesterol levels in T2DM patients, but the underlying mechanisms have not been studied. In the present study, we initially established in vivo T2DM mice models, and our results showed that both free cholesterol (FC) and cholesteryl ester (CE) were accumulated, while the pro-proliferation associated genes were downregulated in T2DM mice liver tissues, which were reversed by Dapagliflozin co-treatment. Similarly, the mice primary hepatocytes were loaded with cholesterol to establish in vitro models, and we expectedly found that Dapagliflozin attenuated cholesterol-overloading induced cytotoxicity and cellular senescence in the hepatocytes. Then, we noticed that oxidative damages occurred in T2DM mice liver tissues and cholesterol treated hepatocytes, which could be suppressed by Dapagliflozin. Also, elimination of Reactive Oxygen Species (ROS) by N-acetyl-L-cysteine (NAC) recovered cellular functions of hepatocytes in vitro and in vivo. Furthermore, the potential underlying mechanisms were uncovered, and our data suggested that Dapagliflozin activated the anti-oxidant Nrf2/HO-1 pathway in mice hepatocytes, and silencing of Nrf2 abrogated the protective effects of Dapagliflozin on cholesterol-overloaded hepatocytes. Collectively, we concluded that Dapagliflozin recovered cholesterol metabolism functions in T2DM mice liver via activating the anti-oxidant Nrf2/HO-1 pathway, and our data supported that Dapagliflozin was a potential therapeutic drug to eliminate cholesterol-induced cytotoxicity during T2DM pathogenesis.
Collapse
Affiliation(s)
- Liu Yang
- Department of Endocrinology, Jingjiang People’s Hospital, Jingjiang, China
| | - Dan Liu
- Department of Endocrinology, Jingjiang People’s Hospital, Jingjiang, China
| | - Hongqin Yan
- Department of Endocrinology, Jingjiang People’s Hospital, Jingjiang, China
| | - Kaixia Chen
- Department of Pharmacy, Jingjiang People’s Hospital, Jingjiang, China,CONTACT Kaixia Chen Department of Pharmacy, Jingjiang People’s Hospital, Jingjiang, China
| |
Collapse
|
33
|
Liu D, Liu S, Fang Y, Liu L, Hu K. Comprehensive Analysis of the Expression and Prognosis for ITGBs: Identification of ITGB5 as a Biomarker of Poor Prognosis and Correlated with Immune Infiltrates in Gastric Cancer. Front Cell Dev Biol 2022; 9:816230. [PMID: 35223869 PMCID: PMC8863963 DOI: 10.3389/fcell.2021.816230] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/27/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Integrin β superfamily members (ITGBs) are documented to play important roles in various biological processes, and accumulating evidence suggests that ITGBs are associated with carcinogenic effects in several malignancies. Gastric cancer (GC) is a complicated and highly heterogeneous disease; however, the expression and prognostic values of eight ITGBs and potential mechanism in GC remain largely unclear. Methods: The expression and prognostic significance of ITGBs in GC were systematically analyzed through Gene Expression Profiling Interactive Analysis, Human Protein Atlas, Kaplan–Meier Plotter, and cBioPortal databases. Then, the mRNA transcription data and corresponding clinical data of GC were downloaded from the Gene Expression Omnibus database as a testing cohort, and differentially expressed and prognostic genes were identified. The correlation between ITGB5 expression and overall survival and various clinical parameters were found by using univariate/multivariable Cox regression and Kaplan–Meier survival analysis. Additionally, differential analysis of gene expression profiles in low- and high-ITGB5 expression groups and pathway enrichment analysis was performed. Finally, the correlation of ITGB5 expression with immune infiltrates in GC was clarified. Results: Compared with adjacent normal tissue, the results reveal that the mRNA levels of ITGB1-2 and ITGB4-8 are significantly higher in GC, and immunohistochemistry results show the consistency between RNA and protein expression levels. Cox regression and Kaplan–Meier survival analysis indicate that high ITGB5 expression contributes to a poor prognosis and could be an independent prognostic factor in GC patients. Besides this, gene functional enrichment analysis indicates that ITGB5 expression is significantly associated with extracellular matrix organization, cell-substrate adhesion, and ossification. The KEGG pathway analysis of ITGB5 shows a close association between ITGB5 and focal adhesion, ECM-receptor interaction, phagosome, and PI3K-Akt signaling pathway. Last, the infiltrating level of CD4+ T cells, macrophages, and dendritic cells are positively related to the expression of ITGB5, especially macrophages, and lower levels of macrophages predict a better prognosis in GC in our study. Conclusion: Our findings investigate that ITGB5 may function as a valid biomarker of prognosis, and high expression of ITGB5 predicts poor prognosis for patients with GC. Besides this, it might be a potential target of precision therapy against GC.
Collapse
Affiliation(s)
- Dongliang Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shaojun Liu
- Department of General Surgery, The First Hospital Affiliated to the University of Science and Technology of China, Hefei, China
| | - Yu Fang
- Department of General Surgery, The First Hospital Affiliated to the University of Science and Technology of China, Hefei, China
| | - Liu Liu
- Department of General Surgery, The First Hospital Affiliated to the University of Science and Technology of China, Hefei, China
- *Correspondence: Liu Liu, ; Kongwang Hu,
| | - Kongwang Hu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Liu Liu, ; Kongwang Hu,
| |
Collapse
|
34
|
Bin YL, Hu HS, Tian F, Wen ZH, Yang MF, Wu BH, Wang LS, Yao J, Li DF. Metabolic Reprogramming in Gastric Cancer: Trojan Horse Effect. Front Oncol 2022; 11:745209. [PMID: 35096565 PMCID: PMC8790521 DOI: 10.3389/fonc.2021.745209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/12/2021] [Indexed: 12/24/2022] Open
Abstract
Worldwide, gastric cancer (GC) represents the fifth most common cancer for incidence and the third leading cause of death in developed countries. Despite the development of combination chemotherapies, the survival rates of GC patients remain unsatisfactory. The reprogramming of energy metabolism is a hallmark of cancer, especially increased dependence on aerobic glycolysis. In the present review, we summarized current evidence on how metabolic reprogramming in GC targets the tumor microenvironment, modulates metabolic networks and overcomes drug resistance. Preclinical and clinical studies on the combination of metabolic reprogramming targeted agents and conventional chemotherapeutics or molecularly targeted treatments [including vascular endothelial growth factor receptor (VEGFR) and HER2] and the value of biomarkers are examined. This deeper understanding of the molecular mechanisms underlying successful pharmacological combinations is crucial in finding the best-personalized treatment regimens for cancer patients.
Collapse
Affiliation(s)
- Yu-Ling Bin
- Department of Rheumatology and Immunology, ZhuZhou Central Hospital, Zhuzhou, China
| | - Hong-Sai Hu
- Department of Gastroenterology, ZhuZhou Central Hospital, Zhuzhou, China
| | - Feng Tian
- Department of Rheumatology and Immunology, ZhuZhou Central Hospital, Zhuzhou, China
| | - Zhen-Hua Wen
- Department of Rheumatology and Immunology, ZhuZhou Central Hospital, Zhuzhou, China
| | - Mei-Feng Yang
- Department of Hematology, Yantian District People's Hospital, Shenzhen, China
| | - Ben-Hua Wu
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - De-Feng Li
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| |
Collapse
|
35
|
Zhong X, Yao L, Xu L, Ma Q, Huang G, Yang M, Gao C, Cheng J, Zhou X, Li Q, Guo X. Comprehensive Analysis of Potential Correlation Between Solute Carrier 1A (SLC1A) Family and Lung Adenocarcinoma. Int J Gen Med 2022; 15:2101-2117. [PMID: 35241927 PMCID: PMC8886152 DOI: 10.2147/ijgm.s350986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/15/2022] [Indexed: 12/12/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD) is the most common dangerous malignant tumor and the leading cause of global cancer incidence and mortality. The Solute Carrier 1A (SLC1A) family play a significant part in cellular biological process, inflammation, and immunity. Specific functions of the SLC1A family in lung cancer are still not systematically described. Objective This study aimed to explore the best biological understanding of SLC1A family in lung cancer. Methods To study the expression and role of the SLC1A family in lung cancer, researchers used a variety of bioinformatics databases and tools. Results Aberrant expression of SLC1A family genes were demonstrated and analyzed the association with gender, tumor grade, cancer stages, and nodal metastasis status. The ectopic expression of SLC1A family genes has prognostic value for LUAD patients. Immune infiltration revealed a significant correlation between SLC1A family genes expression in LUAD. SLC1A family genes were involved in manifold biological processes and have different levels of DNA methylation and genetic alteration. Conclusions These findings suggested that members of the SLC1A family could be a potential target for the development of LUAD therapeutics as well as a reliable indicator of LUAD prognostic value.
Collapse
Affiliation(s)
- Xiaowu Zhong
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Lihua Yao
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Lei Xu
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Qiang Ma
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Guangcheng Huang
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Miyuan Yang
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Chuanli Gao
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Jibing Cheng
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Xi Zhou
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Qinrong Li
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Xiaolan Guo
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Correspondence: Xiaolan Guo, Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China, Tel +86-817-2282059, Fax +86-817-2282059, Email
| |
Collapse
|
36
|
García-Gaytán AC, Hernández-Abrego A, Díaz-Muñoz M, Méndez I. Glutamatergic system components as potential biomarkers and therapeutic targets in cancer in non-neural organs. Front Endocrinol (Lausanne) 2022; 13:1029210. [PMID: 36457557 PMCID: PMC9705578 DOI: 10.3389/fendo.2022.1029210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
Glutamate is one of the most abundant amino acids in the blood. Besides its role as a neurotransmitter in the brain, it is a key substrate in several metabolic pathways and a primary messenger that acts through its receptors outside the central nervous system (CNS). The two main types of glutamate receptors, ionotropic and metabotropic, are well characterized in CNS and have been recently analyzed for their roles in non-neural organs. Glutamate receptor expression may be particularly important for tumor growth in organs with high concentrations of glutamate and might also influence the propensity of such tumors to set metastases in glutamate-rich organs, such as the liver. The study of glutamate transporters has also acquired relevance in the physiology and pathologies outside the CNS, especially in the field of cancer research. In this review, we address the recent findings about the expression of glutamatergic system components, such as receptors and transporters, their role in the physiology and pathology of cancer in non-neural organs, and their possible use as biomarkers and therapeutic targets.
Collapse
|
37
|
Fang SQ, Liu YH, Zhao KP, Zhang HX, Wang HW, Deng YH, Zhou YX, Ge GB, Ni HM, Chen QL. Transcriptional profiling and network pharmacology analysis identify the potential biomarkers from Chinese herbal formula Huosu Yangwei Formula treated gastric cancer in vivo. Chin J Nat Med 2021; 19:944-953. [PMID: 34961592 DOI: 10.1016/s1875-5364(22)60154-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Indexed: 02/07/2023]
Abstract
Huosu Yangwei (HSYW) Formula is a traditioanl Chinese herbal medicine that has been extensively used to treat chronic atrophic gastritis, precancerous lesions of gastric cancer and advanced gastric cancer. However, the effective compounds of HSYW and its related anti-tumor mechanisms are not completely understood. In the current study, 160 ingredients of HSYW were identified and 64 effective compounds were screened by the ADMET evaluation. Furthermore, 64 effective compounds and 2579 potential targets were mapped based on public databases. Animal experiments demonstrated that HSYW significantly inhibited tumor growth in vivo. Transcriptional profiles revealed that 81 mRNAs were differentially expressed in HSYW-treated N87-bearing Balb/c mice. Network pharmacology and PPI network showed that 12 core genes acted as potential markers to evaluate the curative effects of HSYW. Bioinformatics and qRT-PCR results suggested that HSYW might regulate the mRNA expression of DNAJB4, CALD, AKR1C1, CST1, CASP1, PREX1, SOCS3 and PRDM1 against tumor growth in N87-bearing Balb/c mice.
Collapse
Affiliation(s)
- Sheng-Quan Fang
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yue-Han Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Kun-Peng Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hui-Xing Zhang
- School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hong-Wei Wang
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yu-Hai Deng
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yu-Xuan Zhou
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hong-Mei Ni
- School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Qi-Long Chen
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
38
|
Yan X, Chen M, Xiao C, Fu J, Sun X, Hu Z, Zhou H. Effect of unfolded protein response on the immune infiltration and prognosis of transitional cell bladder cancer. Ann Med 2021; 53:1048-1058. [PMID: 34187252 PMCID: PMC8253203 DOI: 10.1080/07853890.2021.1918346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/12/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Bladder cancer (BC) is one of the most common human malignancies worldwide. Previous researches have shown that the unfolded protein response (UPR) pathway could contribute to the tumorigenesis of BC. However, the role of UPR in the immune infiltration, progression, and prognosis of BC is unclear.Methods: The GSVA and ssGSEA methods were used for assessing the UPR score and immune cells infiltration score in three BC public datasets, respectively. The relationship between the UPR pathway and clinicopathological characteristics was analyzed by the Kruskal-Wallis, Wilcox test, and log-rank test. The association of the UPR pathway with various tumor-infiltrating immune cells was evaluated with the correlation analysis. Univariate Cox regression analysis was performed to identify risk factors significantly associated with prognosis. The predictive models were built based on risk factors and visualized with nomograms. The performance of our models was evaluated with the calibration curve, Harrell's concordance index (c-index), and receiver operating characteristic (ROC) analysis.Results: We found that the UPR pathway and many UPR-related genes were significantly associated with the pathologic grade, tumor type, and invasive progression of transitional cell bladder cancer (TCBC), and a high UPR score predicted a poor prognosis in patients. The UPR score was positively correlated with the infiltration abundance of many tumor immune cells in TCBC. Besides, we constructed predictive models based on the UPR score, and good performance was observed, with c-indexes ranging from 0.74 to 0.87.Conclusions: Our study proved that the UPR pathway may have an important impact on the progression, prognosis, and tumor immune infiltration in TCBC, and the models we built may provide effective and reliable guides for prognosis assessment and treatment decision-making for TCBC patients.
Collapse
Affiliation(s)
- Xiaokai Yan
- Department of Oncology, the Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Min Chen
- Department of Oncology, the Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chiying Xiao
- Department of Oncology, the Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiandong Fu
- Department of Oncology, the Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xia Sun
- Department of Oncology, the Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zuohuai Hu
- Department of Oncology, the Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hang Zhou
- Department of Oncology, the Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
39
|
Xue C, Li G, Lu J, Li L. Crosstalk between circRNAs and the PI3K/AKT signaling pathway in cancer progression. Signal Transduct Target Ther 2021; 6:400. [PMID: 34815385 PMCID: PMC8611092 DOI: 10.1038/s41392-021-00788-w] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 02/06/2023] Open
Abstract
Circular RNAs (circRNAs), covalently closed noncoding RNAs, are widely expressed in eukaryotes and viruses. They can function by regulating target gene expression, linear RNA transcription and protein generation. The phosphoinositide 3-kinase (PI3K)/AKT signaling pathway plays key roles in many biological and cellular processes, such as cell proliferation, growth, invasion, migration, and angiogenesis. It also plays a pivotal role in cancer progression. Emerging data suggest that the circRNA/PI3K/AKT axis modulates the expression of cancer-associated genes and thus regulates tumor progression. Aberrant regulation of the expression of circRNAs in the circRNA/PI3K/AKT axis is significantly associated with clinicopathological characteristics and plays an important role in the regulation of biological functions. In this review, we summarized the expression and biological functions of PI3K-AKT-related circRNAs in vitro and in vivo and assessed their associations with clinicopathological characteristics. We also further discussed the important role of circRNAs in the diagnosis, prognostication, and treatment of cancers.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Ganglei Li
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
40
|
Hao W, Wu L, Cao L, Yu J, Ning L, Wang J, Lin X, Chen Y. Radioresistant Nasopharyngeal Carcinoma Cells Exhibited Decreased Cisplatin Sensitivity by Inducing SLC1A6 Expression. Front Pharmacol 2021; 12:629264. [PMID: 33927617 PMCID: PMC8077170 DOI: 10.3389/fphar.2021.629264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 03/02/2021] [Indexed: 12/29/2022] Open
Abstract
Cisplatin-based regimens are commonly used for the treatment of nasopharyngeal carcinoma (NPC) in patients who receive concurrent chemoradiotherapy. The sensitivity of NPC cells to cisplatin is closely associated with the efficacy of radiation therapy. In this study, we established two radioresistant NPC cell lines, HONE1-IR and CNE2-IR, and found that both cell lines showed reduced sensitivity to cisplatin. RNA-sequence analysis showed that SLC1A6 was upregulated in both HONE1-IR and CNE2-IR cell lines. Downregulation of SLC1A6 enhanced cisplatin sensitivity in these two radioresistant NPC cell lines. It was also found that the expression of SLC1A6 was induced during radiation treatment and correlated with poor prognosis of NPC patients. Notably, we observed that upregulation of SLC1A6 led to elevating level of glutamate and the expression of drug-resistant genes, resulted in reduced cisplatin sensitivity. Our findings provide a rationale for developing a novel therapeutic target for NPC patients with cisplatin resistance.
Collapse
Affiliation(s)
- Wenwen Hao
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Head and Neck Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lisha Wu
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Linhui Cao
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinxiu Yu
- Department of Radiotherapy, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Li Ning
- Department of Head and Neck Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jingshu Wang
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoping Lin
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yanfeng Chen
- Department of Head and Neck Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
41
|
Cheng Z, Liu G, Huang C, Zhao X. Upregulation of circRNA_100395 sponges miR-142-3p to inhibit gastric cancer progression by targeting the PI3K/AKT axis. Oncol Lett 2021; 21:419. [PMID: 33841580 PMCID: PMC8020390 DOI: 10.3892/ol.2021.12680] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 01/20/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) has a high morbidity and mortality, hence it is very important to elucidate the molecular pathogenesis mechanism of GC progression in order to find new treatment strategies. The present study aimed to explore the biological function of circular RNA_100395 (circRNA_100395) in GC. The expression level of circRNA_100395 in GC tissues, as well as normal epithelial cells and various gastric cancer cell lines, was detected using reverse transcription-quantitative PCR. Cell Counting Kit-8, EdU assay, flow cytometry and Transwell assays were performed to investigate cell proliferation, apoptosis, migration and invasion, respectively. A dual-luciferase reporter assay was performed to detect the correlation between circRNA_100395 and micro (mi)RNA-142-3p. Western blotting was performed to elucidate the potential regulatory mechanism. circRNA_100395 expression was found to be increased in GC tissues and cell lines. However, miR-142-3p expression was significantly reduced. Besides, low expression levels of circRNA_100395 were associated with poor tumor differentiation, advanced Tumor-Node-Metastasis stage, lymph node metastasis and shorter overall survival time. Moreover, overexpression of circRNA_100395 suppressed cell proliferation, increased the apoptosis rate and suppressed cell invasion and migration by inhibiting the PI3K/AKT signaling pathway. These findings also showed that miRNA-142-3p rescued the antitumor effects induced by circRNA_100395-overexpression. cirRNA_100395-overexpression had antitumor effects via regulating the miR-142-3p signaling pathway, which might be a promising treatment target for GC.
Collapse
Affiliation(s)
- Zhiyi Cheng
- Department of Gastrointestinal Surgery, Hospital Affiliated 5 to Nantong University, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Guiyuan Liu
- Department of Gastrointestinal Surgery, Hospital Affiliated 5 to Nantong University, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Chuanjiang Huang
- Department of Gastrointestinal Surgery, Hospital Affiliated 5 to Nantong University, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Xiaojun Zhao
- Department of Gastrointestinal Surgery, Hospital Affiliated 5 to Nantong University, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| |
Collapse
|
42
|
Carusela MF, Miguel Rubi J. Computational Model for Membrane Transporters. Potential Implications for Cancer. Front Cell Dev Biol 2021; 9:642665. [PMID: 33693005 PMCID: PMC7937797 DOI: 10.3389/fcell.2021.642665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/04/2021] [Indexed: 01/13/2023] Open
Abstract
To explain the increased transport of nutrients and metabolites and to control the movement of drug molecules through the transporters to the cancer cells, it is important to understand the exact mechanism of their structure and activity, as well as their biological and physical characteristics. We propose a computational model that reproduces the functionality of membrane transporters by quantifying the flow of substrates through the cell membrane. The model identifies the force induced by conformational changes of the transporter due to hydrolysis of ATP, in ABC transporters, or by an electrochemical gradient of ions, in secondary transporters. The transport rate is computed by averaging the velocity generated by the force along the paths followed by the substrates. The results obtained are in accordance with the experiments. The model provides an overall framework for analyzing the membrane transport proteins that regulate the flows of ions, nutrients and other molecules across the cell membranes, and their activities.
Collapse
Affiliation(s)
- María Florencia Carusela
- Instituto de Ciencias, Universidad Nacional de General Sarmiento, Buenos Aires, Argentina
- National Scientific and Technical Research Council, Buenos Aires, Argentina
| | - J. Miguel Rubi
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
43
|
Kahya U, Köseer AS, Dubrovska A. Amino Acid Transporters on the Guard of Cell Genome and Epigenome. Cancers (Basel) 2021; 13:E125. [PMID: 33401748 PMCID: PMC7796306 DOI: 10.3390/cancers13010125] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/26/2020] [Accepted: 12/27/2020] [Indexed: 02/06/2023] Open
Abstract
Tumorigenesis is driven by metabolic reprogramming. Oncogenic mutations and epigenetic alterations that cause metabolic rewiring may also upregulate the reactive oxygen species (ROS). Precise regulation of the intracellular ROS levels is critical for tumor cell growth and survival. High ROS production leads to the damage of vital macromolecules, such as DNA, proteins, and lipids, causing genomic instability and further tumor evolution. One of the hallmarks of cancer metabolism is deregulated amino acid uptake. In fast-growing tumors, amino acids are not only the source of energy and building intermediates but also critical regulators of redox homeostasis. Amino acid uptake regulates the intracellular glutathione (GSH) levels, endoplasmic reticulum stress, unfolded protein response signaling, mTOR-mediated antioxidant defense, and epigenetic adaptations of tumor cells to oxidative stress. This review summarizes the role of amino acid transporters as the defender of tumor antioxidant system and genome integrity and discusses them as promising therapeutic targets and tumor imaging tools.
Collapse
Affiliation(s)
- Uğur Kahya
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (A.S.K.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
| | - Ayşe Sedef Köseer
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (A.S.K.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Anna Dubrovska
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (A.S.K.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|