1
|
Lyu H, Bao S, Cai L, Wang M, Liu Y, Sun Y, Hu X. The role and research progress of serine metabolism in tumor cells. Front Oncol 2025; 15:1509662. [PMID: 40265021 PMCID: PMC12011608 DOI: 10.3389/fonc.2025.1509662] [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: 10/11/2024] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
Serine is crucial for tumor initiation, progression, and adaptive immunity. Metabolic pathways for serine synthesis, acquisition, and utilization in tumors and tumor-associated cells are influenced by various physiological factors and the tumor microenvironment, leading to metabolic reprogramming and amplification. Excessive serine metabolism promotes abnormal macromolecule biosynthesis, mitochondrial dysfunction, and epigenetic modifications, driving malignant transformation, proliferation, metastasis, immune suppression, and drug resistance in tumor cells. Restricting dietary serine intake or reducing the expression of serine synthetic enzymes can effectively slow tumor growth and extend patient survival. Consequently, targeting serine metabolism has emerged as a novel and promising research focus in cancer research. This paper reviews serine metabolic pathways and their roles in tumor development. It summarizes the influencing factors of serine metabolism. The article explores the significance of serine synthesis and metabolizing enzymes, along with related biomarkers, in tumor diagnosis and treatment, providing new insights for developing targeted therapies that modulate serine metabolism in cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Yang Sun
- School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xiaoyang Hu
- School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
2
|
Gu HC, Wang LF, Zhang YW, Zhuo YQ, Zhang ZH, Wei XY, Liu QW, Deng KY, Xin HB. Human urine stem cells protect against cyclophosphamide-induced premature ovarian failure by inhibiting SLC1A4-mediated outflux of intracellular serine in ovarian granulosa cells. Cell Mol Biol Lett 2025; 30:21. [PMID: 39972244 PMCID: PMC11840982 DOI: 10.1186/s11658-025-00701-1] [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: 07/23/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Cyclophosphamide (CTX) is the first-line medication for the treatment of breast cancer, although it potentially leads to severe ovarian dysfunction and even premature ovarian failure (POF). However, the mechanism of CTX-induced POF remains unclear. Mesenchymal stem cell-based therapy has been wildly used for treating numerous diseases. Therefore, our study aims to elucidate the underlying mechanism of CTX-induced POF and to explore the therapeutic effect of human urine stem cells (hUSCs) in POF. METHODS CTX-induced POF or ovarian granulosa cell (GCs) apoptosis were treated with hUSCs and their exosomes in vitro and in vivo. Morphological, histological, and functional alternations were examined using multiple approaches. The effector molecules of hUSC-derived exosomes (hUSC-Exo) were determined by differential expression analysis in the ovaries. The target genes of miRNA were accessed by transcriptome sequencing in GCs, and the underlying mechanisms were further elucidated. RESULTS hUSCs remarkably inhibited CTX-induced apoptosis and promoted the proliferation of GCs, respectively. In addition, we observed that miR-27b-3p was highly expressed in hUSC-Exo and markedly suppressed CTX-induced GC apoptosis by specifically inhibiting the expression of SLC1A4, a serine transporter, in ovarian GCs, which, in turn, elevated the concentration of the intracellular serine by inhibiting the outflux of cellular serine. More importantly, the knockdown of SLC1A4 or simple supplementation of serine suppressed CTX-induced apoptosis of GCs. Finally, we demonstrated that CTX-induced apoptosis of ovarian GCs was essential for POF by reducing the intracellular serine concentration via elevating the expression of SLC1A4, whereas hUSCs protected against CTX-induced POF via miR-27b-3p/SLC1A4/serine axis-mediated activation of the PI3K/AKT/mTOR signaling pathway. CONCLUSIONS Our study suggests that hUSC-based cell therapy or simple supplementation of serine may provide an efficient therapeutic approach for the prevention and treatment of CTX-induced POF clinically.
Collapse
Affiliation(s)
- Hao-Cheng Gu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang, 330031, People's Republic of China
- School of Life and Science, Nanchang University, Nanchang, 330031, People's Republic of China
| | - Ling-Fang Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang, 330031, People's Republic of China
| | - Yu-Wei Zhang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang, 330031, People's Republic of China
- School of Life and Science, Nanchang University, Nanchang, 330031, People's Republic of China
| | - You-Qiong Zhuo
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang, 330031, People's Republic of China
- School of Food Science and Technology, Nanchang University, Nanchang, 330031, People's Republic of China
| | - Zhou-Hang Zhang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang, 330031, People's Republic of China
| | - Xing-Yu Wei
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang, 330031, People's Republic of China
| | - Quan-Wen Liu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang, 330031, People's Republic of China
| | - Ke-Yu Deng
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang, 330031, People's Republic of China.
- School of Life and Science, Nanchang University, Nanchang, 330031, People's Republic of China.
| | - Hong-Bo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang, 330031, People's Republic of China.
- School of Life and Science, Nanchang University, Nanchang, 330031, People's Republic of China.
- School of Food Science and Technology, Nanchang University, Nanchang, 330031, People's Republic of China.
| |
Collapse
|
3
|
Fu Y, Chen J, Zhu X, Ding M, Wang H, Fu S. Roles and therapeutic potential of the SLC family in prostate cancer-literature review. BMC Urol 2025; 25:32. [PMID: 39966814 PMCID: PMC11837367 DOI: 10.1186/s12894-025-01714-w] [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: 10/01/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
Prostate cancer (PCa) is one of the most common malignancies in men worldwide. Despite advances in treatment, many patients develop resistance to conventional therapies. Solute carrier (SLC) proteins, as transmembrane transporters, have recently emerged as potential therapeutic targets due to their role in tumor metabolism and progression. This review summarizes the key roles of six SLC proteins in PCa, including their involvement in metabolic reprogramming, regulation of signaling pathways, and effects on the tumor microenvironment. Although targeting of SLC family members in prostate cancer remains an underexplored area, the growing body of evidence suggests that it holds potential for future development.
Collapse
Affiliation(s)
- Yuanzhi Fu
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650101, Yunnan, China
- Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Junhao Chen
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650101, Yunnan, China
| | - Xingcheng Zhu
- Department of Clinical Laboratory, The Second People's Hospital of Qujing City Qujing, Yunnan, China
| | - Mingxia Ding
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650101, Yunnan, China
| | - Haifeng Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650101, Yunnan, China.
| | - Shi Fu
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650101, Yunnan, China.
| |
Collapse
|
4
|
Zheng J, Gong J. SLC1A4 Promotes Malignant Transformation of Hepatocellular Carcinoma by Activating the AKT Signaling. Anal Cell Pathol (Amst) 2025; 2025:1115184. [PMID: 39949345 PMCID: PMC11824774 DOI: 10.1155/ancp/1115184] [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: 12/22/2023] [Revised: 10/04/2024] [Accepted: 11/30/2024] [Indexed: 02/16/2025] Open
Abstract
Due to the difficulty in early diagnosis and the lack of treatment for advanced disease, the mortality rate of hepatocellular carcinoma (HCC) is high, and the 5-year overall survival rate is low at present. SLC1A4 is a neutral amino acid transporter, but its regulatory role and mechanism in HCC are still unclear. Through analyzing the TCGA database and clinical tissue specimens, this study uncovered the high expression of SLC1A4 in tumor tissues of HCC. Worse more, a high level of SLC1A4 may lead to a poor prognosis of HCC. Mechanically, silencing SLC1A4 inhibited the phosphorylation activation of AKT by suppressing the ubiquitin modification of AKT at lysine 63 and amino acid influx represented by D-serine, decreasing the protein level of β-catenin in the cell nucleus and suppressing the transcriptional activity of c-Myc and EpCAM promoters. As a result, silencing SLC1A4 inhibited the proliferation, migration, and stemness of hepatic cancer cells, which was successfully reversed by the introduction of exogenous AKT. Moreover, epithelial-mesenchymal transition (EMT) in vitro and metastasis potential in vivo of hepatic cancer cells was suppressed by the downregulated SLC1A4 level. In conclusion, SLC1A4 promotes the malignant transformation of HCC through activating signal transduction mediated by AKT. The findings in this study suggested that SLC1A4 may be a diagnostic indicator for the early HCC and a therapeutic target for the advanced HCC.
Collapse
Affiliation(s)
- Jiaoyun Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Jian Gong
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
- Research Center of Digestive Disease, Central South University, Changsha 410011, Hunan, China
- Clinical Research Center of Digestive Diseases of Hunan Province, Changsha 410011, Hunan, China
| |
Collapse
|
5
|
Dyachenko EI, Bel’skaya LV. Transmembrane Amino Acid Transporters in Shaping the Metabolic Profile of Breast Cancer Cell Lines: The Focus on Molecular Biological Subtype. Curr Issues Mol Biol 2024; 47:4. [PMID: 39852119 PMCID: PMC11763447 DOI: 10.3390/cimb47010004] [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: 11/29/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/26/2025] Open
Abstract
Amino acid metabolism in breast cancer cells is unique for each molecular biological subtype of breast cancer. In this review, the features of breast cancer cell metabolism are considered in terms of changes in the amino acid composition due to the activity of transmembrane amino acid transporters. In addition to the main signaling pathway PI3K/Akt/mTOR, the activity of the oncogene c-Myc, HIF, p53, GATA2, NF-kB and MAT2A have a direct effect on the amino acid metabolism of cancer cells, their growth and proliferation, as well as the maintenance of homeostatic equilibrium. A distinctive feature of luminal subtypes of breast cancer from TNBC is the ability to perform gluconeogenesis. Breast cancers with a positive expression of the HER2 receptor, in contrast to TNBC and luminal A subtype, have a distinctive active synthesis and consumption of fatty acids. It is interesting to note that amino acid transporters exhibit their activity depending on the pH level inside the cell. In the most aggressive forms of breast cancer or with the gradual progression of the disease, pH will also change, which will directly affect the metabolism of amino acids. Using the cell lines presented in this review, we can trace the characteristic features inherent in each of the molecular biological subtypes of breast cancer and develop the most optimal therapeutic targets.
Collapse
Affiliation(s)
| | - Lyudmila V. Bel’skaya
- Biochemistry Research Laboratory, Omsk State Pedagogical University, 644099 Omsk, Russia;
| |
Collapse
|
6
|
Zhang S, Huang F, Wang Y, Long Y, Li Y, Kang Y, Gao W, Zhang X, Wen Y, Wang Y, Pan L, Xia Y, Yang Z, Yang Y, Mo H, Li B, Hu J, Song Y, Zhang S, Dong S, Du X, Li Y, Liu Y, Liao W, Gao Y, Zhang Y, Chen H, Liang Y, Chen J, Weng H, Huang H. NAT10-mediated mRNA N 4-acetylcytidine reprograms serine metabolism to drive leukaemogenesis and stemness in acute myeloid leukaemia. Nat Cell Biol 2024; 26:2168-2182. [PMID: 39506072 PMCID: PMC11628400 DOI: 10.1038/s41556-024-01548-y] [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: 02/28/2024] [Accepted: 09/27/2024] [Indexed: 11/08/2024]
Abstract
RNA modification has emerged as an important epigenetic mechanism that controls abnormal metabolism and growth in acute myeloid leukaemia (AML). However, the roles of RNA N4-acetylcytidine (ac4C) modification in AML remain elusive. Here, we report that ac4C and its catalytic enzyme NAT10 drive leukaemogenesis and sustain self-renewal of leukaemic stem cells/leukaemia-initiating cells through reprogramming serine metabolism. Mechanistically, NAT10 facilitates exogenous serine uptake and de novo biosynthesis through ac4C-mediated translation enhancement of the serine transporter SLC1A4 and the transcription regulators HOXA9 and MENIN that activate transcription of serine synthesis pathway genes. We further characterize fludarabine as an inhibitor of NAT10 and demonstrate that pharmacological inhibition of NAT10 targets serine metabolic vulnerability, triggering substantial anti-leukaemia effects both in vitro and in vivo. Collectively, our study demonstrates the functional importance of ac4C and NAT10 in metabolism control and leukaemogenesis, providing insights into the potential of targeting NAT10 for AML therapy.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Humans
- Animals
- Mice
- Serine/metabolism
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/drug effects
- RNA, Messenger/metabolism
- RNA, Messenger/genetics
- N-Terminal Acetyltransferases/metabolism
- N-Terminal Acetyltransferases/genetics
- Cell Line, Tumor
- Homeodomain Proteins/metabolism
- Homeodomain Proteins/genetics
- Cytidine/analogs & derivatives
- Cytidine/pharmacology
- Cytidine/metabolism
- Gene Expression Regulation, Leukemic/drug effects
- Mice, Inbred NOD
- Carcinogenesis/genetics
- Carcinogenesis/metabolism
Collapse
Affiliation(s)
- Subo Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Feng Huang
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Yushuai Wang
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Yifei Long
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuanpei Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yalin Kang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Weiwei Gao
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiuxin Zhang
- Bioland Laboratory, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Yueting Wen
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Yun Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lili Pan
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
- Union Clinical Medical Colleges, Fujian Medical University, Fuzhou, China
| | - Youmei Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhoutian Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ying Yang
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Hongjie Mo
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Baiqing Li
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Jiacheng Hu
- Bioland Laboratory, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Yunda Song
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shilin Zhang
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Shenghua Dong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao Du
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yingmin Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yadi Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenting Liao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yijun Gao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yaojun Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hongming Chen
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Yang Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianjun Chen
- Department of Systems Biology & Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA
| | - Hengyou Weng
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China.
- Bioland Laboratory, Guangzhou, China.
| | - Huilin Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
7
|
Xing XW, Huang X, Li WP, Wang MK, Yang JS. Clinical application value of long non-coding RNAs signatures of genomic instability in predicting prognosis of hepatocellular carcinoma. World J Gastrointest Surg 2024; 16:2386-2392. [PMID: 39220063 PMCID: PMC11362949 DOI: 10.4240/wjgs.v16.i8.2386] [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: 03/19/2024] [Revised: 05/16/2024] [Accepted: 06/05/2024] [Indexed: 08/16/2024] Open
Abstract
Hepatocellular carcinoma (HCC) presents challenges due to its high recurrence and metastasis rates and poor prognosis. While current clinical diagnostic and prognostic indicators exist, their accuracy remains imperfect due to their biological complexity. Therefore, there is a quest to identify improved biomarkers for HCC diagnosis and prognosis. By combining long non-coding RNA (lncRNA) expression and somatic mutations, Duan et al identified five representative lncRNAs from 88 lncRNAs related to genomic instability (GI), forming a GI-derived lncRNA signature (LncSig). This signature outperforms previously reported LncSig and TP53 mutations in predicting HCC prognosis. In this editorial, we comprehensively evaluate the clinical application value of such prognostic evaluation model based on sequencing technology in terms of cost, time, and practicability. Additionally, we provide an overview of various prognostic models for HCC, aiding in a comprehensive understanding of research progress in prognostic evaluation methods.
Collapse
Affiliation(s)
- Xiao-Wen Xing
- Department of Disease Control and Prevention, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Xiao Huang
- Department of Disease Control and Prevention, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Wei-Peng Li
- Department of Disease Control and Prevention, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Ming-Ke Wang
- Department of Disease Control and Prevention, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Ji-Shun Yang
- Medical Care Center, Naval Medical University, Shanghai 200052, China
| |
Collapse
|
8
|
Luo MY, Han Z, Wang J, Zhong C, Chen J. TARDBP is a candidate diagnostic biomarker promoting tumor progression via impacting tumor immunity and tumor microenvironment. J Cancer 2024; 15:4113-4127. [PMID: 38947395 PMCID: PMC11212099 DOI: 10.7150/jca.96800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/23/2024] [Indexed: 07/02/2024] Open
Abstract
In the realm of cancer research, particularly hepatocellular carcinoma (HCC), TAR DNA-binding protein (TARDBP) has transitioned from being associated with neurodegenerative diseases to emerging as a significant molecule in oncology due to its aberrant expression in HCC and other malignancies. This shift underlines the versatility of TARDBP and its critical role in tumorigenesis. Our study illuminates TARDBP's universal upregulation across various cancers, indicating its involvement in fundamental oncogenic processes and potential impact on genomic instability. The relationship between TARDBP expression and tumor mutational burden (TMB) across several cancers highlights its influence on a key hallmark of cancer progression. Additionally, TARDBP's interaction with immune and inflammatory factors within the tumor microenvironment, including its association with immune-stimulatory factors and inverse relationship with immune inhibitors, suggests its role in modulating immune evasion. Clinically, TARDBP's aberrant expression correlates with adverse patient outcomes in HCC, making it a promising candidate for therapeutic targeting. The study concludes that TARDBP holds significant potential as a novel therapeutic target in HCC and possibly other malignancies, meriting further exploration to integrate TARDBP-targeted therapies into cancer treatment protocols, thereby advancing the field of precision medicine.
Collapse
Affiliation(s)
- Min-Yi Luo
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, 510080, Guangzhou, Guangdong, China
- Department of Coloproctology, The Sixth Affiliated Hospital, Sun Yat-sen University, 510080, Guangzhou, Guangdong, China
| | - Zhe Han
- Department of Neurology, The First Bethune Hospital of Jilin University, 130000, Changchun, Jilin, China
| | - Jiaqi Wang
- Department of Neonatology, Jiangmen People's Hospital, 52900, Jiangmen, Guangdong, China
| | - Cheng Zhong
- Department of Orthopedics, Jiangmen Hospital of Traditional Chinese Medicine Affiliated to Jinan University, 52900, Jiangmen, Guangdong, China
| | - Jiancong Chen
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, 510080, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Agrawal P, Chen S, de Pablos A, Jame-Chenarboo F, Miera Saenz de Vega E, Darvishian F, Osman I, Lujambio A, Mahal LK, Hernando E. Integrated in vivo functional screens and multi-omics analyses identify α-2,3-sialylation as essential for melanoma maintenance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584072. [PMID: 38559078 PMCID: PMC10979837 DOI: 10.1101/2024.03.08.584072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Glycosylation is a hallmark of cancer biology, and altered glycosylation influences multiple facets of melanoma growth and progression. To identify glycosyltransferases, glycans, and glycoproteins essential for melanoma maintenance, we conducted an in vivo growth screen with a pooled shRNA library of glycosyltransferases, lectin microarray profiling of benign nevi and melanoma patient samples, and mass spectrometry-based glycoproteomics. We found that α-2,3 sialyltransferases ST3GAL1 and ST3GAL2 and corresponding α-2,3-linked sialosides are upregulated in melanoma compared to nevi and are essential for melanoma growth in vivo and in vitro. Glycoproteomics revealed that glycoprotein targets of ST3GAL1 and ST3GAL2 are enriched in transmembrane proteins involved in growth signaling, including the amino acid transporter Solute Carrier Family 3 Member 2 (SLC3A2/CD98hc). CD98hc suppression mimicked the effect of ST3GAL1 and ST3GAL2 silencing, inhibiting melanoma cell proliferation. We found that both CD98hc protein stability and its pro-survival effect in melanoma are dependent upon α-2,3 sialylation mediated by ST3GAL1 and ST3GAL2. In summary, our studies reveal that α-2,3-sialosides functionally contribute to melanoma maintenance, supporting ST3GAL1 and ST3GAL2 as novel therapeutic targets in these tumors.
Collapse
Affiliation(s)
- Praveen Agrawal
- Department of Pathology, NYU Grossman School of Medicine, New York
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Shuhui Chen
- Department of Chemistry, New York University
| | - Ana de Pablos
- Department of Pathology, NYU Grossman School of Medicine, New York
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health
- Centro Nacional de Investigaciones Oncologicas (CNIO), Madrid, Spain
| | | | | | | | - Iman Osman
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health
- Department of Dermatology, NYU Grossman School of Medicine, New York
| | | | - Lara K. Mahal
- Department of Chemistry, New York University
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Eva Hernando
- Department of Pathology, NYU Grossman School of Medicine, New York
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health
| |
Collapse
|
10
|
Pang S, Zhao S, Dongye Y, Fan Y, Liu J. Identification and validation of m6A-associated ferroptosis genes in renal clear cell carcinoma. Cell Biol Int 2024. [PMID: 38440906 DOI: 10.1002/cbin.12146] [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: 10/11/2023] [Revised: 01/09/2024] [Accepted: 02/17/2024] [Indexed: 03/06/2024]
Abstract
Urinary cancer is synonymous with clear cell renal cell carcinoma (ccRCC). Unfortunately, existing treatments for this illness are ineffective and unpromising. Finding novel ccRCC biomarkers is crucial to creating successful treatments. The Cancer Genome Atlas provided clear cell renal cell carcinoma transcriptome data. Functional enrichment analysis was performed on ccRCC and control samples' differentially expressed N6-methyladenosine RNA methylation and ferroptosis-related genes (DEMFRGs). Machine learning was used to find and model ccRCC patients' predicted genes. A nomogram was created for clear cell renal cell carcinoma patients. Prognostic genes were enriched. We examined patients' immune profiles by risk score. Our prognostic genes predicted ccRCC treatment drugs. We found 37 DEMFRGs by comparing 1913 differentially expressed ccRCC genes to 202 m6A RNA methylation FRGs. Functional enrichment analysis showed that hypoxia-induced cell death and metabolism pathways were the most differentially expressed methylation functional regulating genes. Five prognostic genes were found by machine learning: TRIB3, CHAC1, NNMT, EGFR, and SLC1A4. An advanced renal cell carcinoma nomogram with age and risk score accurately predicted the outcome. These five prognostic genes were linked to various cancers. Immunological cell number and checkpoint expression differed between high- and low-risk groups. The risk model successfully predicted immunotherapy outcome, showing high-risk individuals had poor results. NIACIN, TAE-684, ROCILETINIB, and others treat ccRCC. We found ccRCC prognostic genes that work. This discovery may lead to new ccRCC treatments.
Collapse
Affiliation(s)
- Shuo Pang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
- Department of Urinary Surgery, Jinan Third People's Hospital, Jinan, Shandong, P.R. China
| | - Shuo Zhao
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
| | - Yuxi Dongye
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
- Department of Urinary Surgery, Jinan Third People's Hospital, Jinan, Shandong, P.R. China
| | - Yidong Fan
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
| | - Jikai Liu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
11
|
Kim YH, Yoon SJ, Kim M, Kim HH, Song YS, Jung JW, Han D, Cho SW, Kwon SW, Park YJ. Integrative Multi-omics Analysis Reveals Different Metabolic Phenotypes Based on Molecular Characteristics in Thyroid Cancer. Clin Cancer Res 2024; 30:883-894. [PMID: 38088902 DOI: 10.1158/1078-0432.ccr-23-2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/06/2023] [Accepted: 12/11/2023] [Indexed: 02/17/2024]
Abstract
PURPOSE Thyroid cancer metabolic characteristics vary depending on the molecular subtype determined by mutational status. We aimed to investigate the molecular subtype-specific metabolic characteristics of thyroid cancers. EXPERIMENTAL DESIGN An integrative multi-omics analysis was conducted, incorporating transcriptomics, metabolomics, and proteomics data obtained from human tissues representing distinct molecular characteristics of thyroid cancers: BRAF-like (papillary thyroid cancer with BRAFV600E mutation; PTC-B), RAS-like (follicular thyroid cancer with RAS mutation; FTC-R), and ATC-like (anaplastic thyroid cancer with BRAFV600E or RAS mutation; ATC-B or ATC-R). To validate our findings, we employed tissue microarray of human thyroid cancer tissues and performed in vitro analyses of cancer cell phenotypes and metabolomic assays after inducing genetic knockdown. RESULTS Metabolic properties differed between differentiated thyroid cancers of PTC-B and FTC-R, but were similar in dedifferentiated thyroid cancers of ATC-B/R, regardless of their mutational status. Tricarboxylic acid (TCA) intermediates and branched-chain amino acids (BCAA) were enriched with the activation of TCA cycle only in FTC-R, whereas one-carbon metabolism and pyrimidine metabolism increased in both PTC-B and FTC-R and to a great extent in ATC-B/R. However, the protein expression levels of the BCAA transporter (SLC7A5) and a key enzyme in one-carbon metabolism (SHMT2) increased in all thyroid cancers and were particularly high in ATC-B/R. Knockdown of SLC7A5 or SHMT2 inhibited the migration and proliferation of thyroid cancer cell lines differently, depending on the mutational status. CONCLUSIONS These findings define the metabolic properties of each molecular subtype of thyroid cancers and identify metabolic vulnerabilities, providing a rationale for therapies targeting its altered metabolic pathways in advanced thyroid cancer.
Collapse
Affiliation(s)
- Yoo Hyung Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, the Republic of South Korea
| | - Sang Jun Yoon
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, the Republic of South Korea
| | - Mina Kim
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, the Republic of South Korea
| | - Hwan Hee Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, the Republic of South Korea
| | - Young Shin Song
- Department of Internal Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, the Republic of South Korea
| | - Jin Woo Jung
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, the Republic of South Korea
| | - Dohyun Han
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, the Republic of South Korea
- Transdisciplinary Department of Medicine & Advanced Technology, Seoul National University Hospital, Seoul, the Republic of South Korea
| | - Sun Wook Cho
- Department of Internal Medicine, Seoul National University Hospital, Seoul, the Republic of South Korea
| | - Sung Won Kwon
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, the Republic of South Korea
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, the Republic of South Korea
- Department of Internal Medicine and Genomic Medicine Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, the Republic of South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, the Republic of South Korea
| |
Collapse
|
12
|
Tombari C, Zannini A, Bertolio R, Pedretti S, Audano M, Triboli L, Cancila V, Vacca D, Caputo M, Donzelli S, Segatto I, Vodret S, Piazza S, Rustighi A, Mantovani F, Belletti B, Baldassarre G, Blandino G, Tripodo C, Bicciato S, Mitro N, Del Sal G. Mutant p53 sustains serine-glycine synthesis and essential amino acids intake promoting breast cancer growth. Nat Commun 2023; 14:6777. [PMID: 37880212 PMCID: PMC10600207 DOI: 10.1038/s41467-023-42458-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/11/2023] [Indexed: 10/27/2023] Open
Abstract
Reprogramming of amino acid metabolism, sustained by oncogenic signaling, is crucial for cancer cell survival under nutrient limitation. Here we discovered that missense mutant p53 oncoproteins stimulate de novo serine/glycine synthesis and essential amino acids intake, promoting breast cancer growth. Mechanistically, mutant p53, unlike the wild-type counterpart, induces the expression of serine-synthesis-pathway enzymes and L-type amino acid transporter 1 (LAT1)/CD98 heavy chain heterodimer. This effect is exacerbated by amino acid shortage, representing a mutant p53-dependent metabolic adaptive response. When cells suffer amino acids scarcity, mutant p53 protein is stabilized and induces metabolic alterations and an amino acid transcriptional program that sustain cancer cell proliferation. In patient-derived tumor organoids, pharmacological targeting of either serine-synthesis-pathway and LAT1-mediated transport synergizes with amino acid shortage in blunting mutant p53-dependent growth. These findings reveal vulnerabilities potentially exploitable for tackling breast tumors bearing missense TP53 mutations.
Collapse
Affiliation(s)
- Camilla Tombari
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, 34149, Trieste, Italy
| | - Alessandro Zannini
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, 34149, Trieste, Italy
| | - Rebecca Bertolio
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, 34149, Trieste, Italy
| | - Silvia Pedretti
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari, University of Milan, Milan, Italy
| | - Matteo Audano
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari, University of Milan, Milan, Italy
| | - Luca Triboli
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, 34149, Trieste, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Science, Human Pathology Section, School of Medicine, University of Palermo, 90133, Palermo, Italy
| | - Davide Vacca
- Tumor Immunology Unit, Department of Health Science, Human Pathology Section, School of Medicine, University of Palermo, 90133, Palermo, Italy
| | - Manuel Caputo
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, 34149, Trieste, Italy
| | - Sara Donzelli
- Translational Oncology Research Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Ilenia Segatto
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, 33081, Aviano, Italy
| | - Simone Vodret
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, 34149, Trieste, Italy
| | - Silvano Piazza
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, 34149, Trieste, Italy
| | - Alessandra Rustighi
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, 34149, Trieste, Italy
| | - Fiamma Mantovani
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Barbara Belletti
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, 33081, Aviano, Italy
| | - Gustavo Baldassarre
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, 33081, Aviano, Italy
| | - Giovanni Blandino
- Translational Oncology Research Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Science, Human Pathology Section, School of Medicine, University of Palermo, 90133, Palermo, Italy
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Silvio Bicciato
- Center for Genome Research, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Nico Mitro
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari, University of Milan, Milan, Italy
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Giannino Del Sal
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, 34149, Trieste, Italy.
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy.
| |
Collapse
|
13
|
Whitley JA, Cai H. Engineering extracellular vesicles to deliver CRISPR ribonucleoprotein for gene editing. J Extracell Vesicles 2023; 12:e12343. [PMID: 37723839 PMCID: PMC10507228 DOI: 10.1002/jev2.12343] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 09/20/2023] Open
Abstract
Clustered regularly interspaced palindromic repeats (CRISPR) is a gene editing tool with tremendous therapeutic potential. Recently, ribonucleoprotein (RNP) complex-based CRISPR systems have gained momentum due to their reduction of off-target editing. This has coincided with the emergence of extracellular vesicles (EVs) as a therapeutic delivery vehicle due to its low immunogenicity and high capacity for manipulation. EVs are cell-derived membranous nanoparticles which mediate the intercellular transfer of molecular components. Current technologies achieve CRISPR RNP encapsulation into EVs through EVs biogenesis, thereby avoiding unnecessary physical, chemical or biological manipulations to the vesicles directly. Herein, we identify sixteen EVs-based CRISPR RNP encapsulation strategies, each with distinct genetic features to encapsulate CRISPR RNP. According to the molecular mechanism facilitating the encapsulation process, there are six strategies of encapsulating Cas9 RNP into virus-like particles based on genetic fusion, seven into EVs based on protein tethering, and three based on sgRNA-coupled encapsulation. Additionally, the incorporation of a targeting moiety to the EVs membrane surface through EVs biogenesis confers tropism and increases delivery efficiency to specific cell types. The targeting moieties include viral envelope proteins, recombinant proteins containing a ligand peptide, single-chain fragment variable (scFv) antibodies, and integrins. However, current strategies still have a number of limitations which prevent their use in clinical trials. Among those, the incorporation of viral proteins for encapsulation of Cas9 RNP have raised issues of biocompatibility due to host immune response. Future studies should focus on genetically engineering the EVs without viral proteins, enhancing EVs delivery specificity, and promoting EVs-based homology directed repair. Nevertheless, the integration of CRISPR RNP encapsulation and tropism technologies will provide strategies for the EVs-based delivery of CRISPR RNP in gene therapy and disease treatment.
Collapse
Affiliation(s)
- Joseph Andrew Whitley
- Department of Pharmaceutical and Biomedical SciencesCollege of PharmacyUniversity of GeorgiaAthensGeorgiaUSA
| | - Houjian Cai
- Department of Pharmaceutical and Biomedical SciencesCollege of PharmacyUniversity of GeorgiaAthensGeorgiaUSA
| |
Collapse
|
14
|
Ding Y, Gong Y, Zeng H, Zhou X, Yu Z, Pan J, Zhou M, Liu S, Lai W. Biological function analysis of ARHGAP39 as an independent prognostic biomarker in hepatocellular carcinoma. Aging (Albany NY) 2023; 15:2631-2666. [PMID: 37059586 PMCID: PMC10120899 DOI: 10.18632/aging.204635] [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: 01/01/2023] [Accepted: 03/11/2023] [Indexed: 04/16/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common subtype of liver cancer, with a high morbidity and low survival rate. Rho GTPase activating protein 39 (ARHGAP39) is a crucial activating protein of Rho GTPases, a novel target in cancer therapy, and it was identified as a hub gene for gastric cancer. However, the expression and role of ARHGAP39 in hepatocellular carcinoma remain unclear. Accordingly, the cancer genome atlas (TCGA) data were used to analyze the expression and clinical value of ARHGAP39 in hepatocellular carcinoma. Further, the LinkedOmics tool suggested functional enrichment pathways for ARHGAP39. To investigate in depth the possible role of ARHGAP39 on immune infiltration, we analyzed the relationship between ARHGAP39 and chemokines in HCCLM3 cells. Finally, the GSCA website was used to explore drug resistance in patients with high ARHGAP39 expression. Studies have shown that ARHGAP39 is highly expressed in hepatocellular carcinoma and relevant to clinicopathological features. In addition, the overexpression of ARHGAP39 leads to a poor prognosis. Besides, co-expressed genes and enrichment analysis showed a correlation with the cell cycle. Notably, ARHGAP39 may worsen the survival of hepatocellular carcinoma patients by increasing the level of immune infiltration through chemokines. Moreover, N6-methyladenosine (m6A) modification-related factors and drug sensitivity were also found to be associated with ARHGAP39. In brief, ARHGAP39 is a promising prognostic factor for hepatocellular carcinoma patients that is closely related to cell cycle, immune infiltration, m6A modification, and drug resistance.
Collapse
Affiliation(s)
- Yongqi Ding
- Department of Health Management Medical, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Yiyang Gong
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Hong Zeng
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Xuanrui Zhou
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Zichuan Yu
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Jingying Pan
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Minqin Zhou
- Second College of Clinical Medicine, Nanchang University, Nanchang, China
| | - Shiwen Liu
- Emergency Intensive Care Unit, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wei Lai
- Department of Health Management Medical, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
15
|
Zhang P, Zhao JH, Chen L, Bian ZL, Ju LL, Wang HX, Cai WH. Expression and function of myelin expression factor 2 in hepatocellular carcinoma. BMC Gastroenterol 2023; 23:20. [PMID: 36658471 PMCID: PMC9854206 DOI: 10.1186/s12876-023-02644-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023] Open
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is one of the most common malignant tumours in the world and has a high mortality rate. However, the pathogenesis of HCC remains unclear. This study aimed to investigate the potential biomarkers of HCC. METHODS ONCOMINE, HCCDB and THE HUMAN PROTEIN ATLAS were used to identify myelin expression factor 2 (MYEF2) as a potential biomarker for HCC. The Cancer Genome Atlas database was used to further validate and analyse the value of MYEF2. Kaplan-Meier Plotter was used for the prognostic analysis. The COX regression model and Kaplan-Meier method were used to investigate the clinical value of MYEF2 in the prognosis of HCC by reviewing the survival status of patients. Fluorescent quantitative polymerase chain reaction (qPCR) and immunohistochemistry were used to detect the expressions of the MYEF2 mRNA and protein in HCC tissues and cell lines. qPCR and Western blotting were used to validate the efficiency of MYEF2 knockout and overexpression in HCC cells. The invasion and migration abilities regulated by MYEF2 were detected by performing transwell and wound healing assays. RESULTS MYEF2 is significantly upregulated in HCC and is mainly located in the nucleus of HCC cells. MYEF2 expression is significantly associated with the tumour stage, histological grade and TNM stage. High MYEF2 expression is an independent prognostic factor for patients with HCC. Functionally, elevated MYEF2 facilitated cell migration and invasion in vitro. In contrast, decreased MYEF2 inhibited cell migration and invasion. CONCLUSIONS MYEF2 may be a novel biomarker with potential diagnosis and prognosis values and as a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Peng Zhang
- grid.260483.b0000 0000 9530 8833Medical School of Nantong University, Nantong Third People’s Hospital, Nantong, 226000 Jiangsu People’s Republic of China
| | - Jiang-Hua Zhao
- grid.260483.b0000 0000 9530 8833Medical School of Nantong University, Nantong Third People’s Hospital, Nantong, 226000 Jiangsu People’s Republic of China
| | - Lin Chen
- grid.260483.b0000 0000 9530 8833Nantong Institute of Liver Diseases, Nantong Third People’s Hospital, Nantong University, Nantong, 226000 Jiangsu People’s Republic of China
| | - Zhao-Lian Bian
- grid.260483.b0000 0000 9530 8833Nantong Institute of Liver Diseases, Nantong Third People’s Hospital, Nantong University, Nantong, 226000 Jiangsu People’s Republic of China
| | - Lin-Ling Ju
- grid.260483.b0000 0000 9530 8833Nantong Institute of Liver Diseases, Nantong Third People’s Hospital, Nantong University, Nantong, 226000 Jiangsu People’s Republic of China
| | - Hui-Xuan Wang
- grid.260483.b0000 0000 9530 8833Nantong Institute of Liver Diseases, Nantong Third People’s Hospital, Nantong University, Nantong, 226000 Jiangsu People’s Republic of China
| | - Wei-Hua Cai
- grid.260483.b0000 0000 9530 8833Nantong Institute of Liver Disease, Department of Hepatobiliary Surgery, Nantong Third People’s Hospital, Nantong University, Nantong, 226000 Jiangsu People’s Republic of China
| |
Collapse
|
16
|
Xie H, Wang L, Tang Y, Zhao M, Wang Z, Liu M, Zhao Q, Zhou J, Wu Y. Functional analysis of differently expressed ferroptosis-related genes in patients with mitral valve prolapse. Front Genet 2022; 13:1062212. [PMID: 36523770 PMCID: PMC9745071 DOI: 10.3389/fgene.2022.1062212] [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: 10/05/2022] [Accepted: 11/18/2022] [Indexed: 01/06/2025] Open
Abstract
Background: The prevalence of mitral valve prolapse (MVP) in heart valvular diseases is globally increasing. However, the understanding of its etiology and pathogenesis is limited. So far, the relationship between ferroptosis-related genes and long non-coding RNAs (lncRNAs) in MVP remains unexplored. This study investigates the potential pathogenesis of ferroptosis-related genes in MVP and provides a therapeutic target for the disease. Methods: Blood samples from patients with MVP and healthy volunteers were collected for transcriptomic sequencing to analyze the expression of ferroptosis-related differentially expressed genes (DEGs) and differentially expressed long non-coding RNAs (DElncRNAs Co-expression network of ferroptosis-related DEGs and DElncRNAs. Furthermore, this work conducted GO and KEGG enrichment analyses. Results: CDKN2A, SLC1A4, ATF3, and other core genes related to the mitral valve prolapse were screened out. CDKN2A, SLC1A4, and ATF3 genes were at the core position of the network, regulated by numerous lncRNAs. Notably, these genes are primarily involved in the extracellular region and p53 signaling pathway. Conclusion: In summary, CDKN2A, SLC1A4, and ATF3 regulate the pathophysiological process of MVP and are potential therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jingxin Zhou
- Department of Cardiovascular Surgery, Nanjing Medical University First Affiliated Hospital, Nanjing, China
| | - Yanhu Wu
- Department of Cardiovascular Surgery, Nanjing Medical University First Affiliated Hospital, Nanjing, China
| |
Collapse
|
17
|
Liu Y, Du S, Yuan M, He X, Zhu C, Han K, Zhu Y, Yang Q, Tong R. Identification of a novel ferroptosis-related gene signature associated with prognosis, the immune landscape, and biomarkers for immunotherapy in ovarian cancer. Front Pharmacol 2022; 13:949126. [PMID: 36386203 PMCID: PMC9641277 DOI: 10.3389/fphar.2022.949126] [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: 05/20/2022] [Accepted: 10/11/2022] [Indexed: 12/09/2022] Open
Abstract
Ferroptosis has been implicated in tumor progression and immunoregulation. Identification of ferroptosis-related prognostic gene is important for immunotherapy and prognosis in ovarian cancer (OV). We assessed the potential predictive power of a novel ferroptosis-related gene (FRG) signature for prognosis and immunotherapy in Asian and Caucasian OV populations. We collected gene expression profiles and clinicopathological data from public databases. The least absolute shrinkage and selection operator Cox regression algorithm was used to construct the FRG signature. Receiver operating characteristic (ROC) curve, Kaplan-Meier method, Cox regression model were used to evaluate the clinical benefits of FRG signature. Gene functional and gene set enrichment analyses were used for functional annotation and immune landscape analysis. A 15-FRG signature was constructed and used to stratify patients into two risk groups. Patients in the high-risk group had significantly worse survival. The risk score was a significant independent risk factor for OS. The area under the ROC curve indicated the good prediction performance of the FRG signature. Notably, the low-risk group showed a significant enrichment in immune-related pathways and a “hot” immune status. The risk score was found to be an efficient and robust predictor of response to immunotherapy. In conclusion, our study identified a novel 15-FRG prognostic signature that can be used for prognostic prediction and precision immunotherapy in Asian and Caucasian OV populations.
Collapse
Affiliation(s)
- Yilong Liu
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Suya Du
- Department of Clinical Pharmacy, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Mengying Yuan
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xia He
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Changyu Zhu
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ke Han
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuyan Zhu
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qianwen Yang
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Rongsheng Tong,
| |
Collapse
|
18
|
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: 0.3] [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
|
19
|
Guo C, Dong C, Zhang J, Wang R, Wang Z, Zhou J, Wang W, Ji B, Ma B, Ge Y, Wang Z. An Immune Signature Robustly Predicts Clinical Deterioration for Hepatitis C Virus-Related Early-Stage Cirrhosis Patients. Front Med (Lausanne) 2021; 8:716869. [PMID: 34350203 PMCID: PMC8326446 DOI: 10.3389/fmed.2021.716869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 06/24/2021] [Indexed: 12/22/2022] Open
Abstract
Hepatitis C virus (HCV)-related cirrhosis leads to a heavy global burden of disease. Clinical risk stratification in HCV-related compensated cirrhosis remains a major challenge. Here, we aim to develop a signature comprised of immune-related genes to identify patients at high risk of progression and systematically analyze immune infiltration in HCV-related early-stage cirrhosis patients. Bioinformatics analysis was applied to identify immune-related genes and construct a prognostic signature in microarray data set. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses were conducted with the “clusterProfiler” R package. Besides, the single sample gene set enrichment analysis (ssGSEA) was used to quantify immune-related risk term abundance. The nomogram and calibrate were set up via the integration of the risk score and clinicopathological characteristics to assess the effectiveness of the prognostic signature. Finally, three genes were identified and were adopted to build an immune-related prognostic signature for HCV-related cirrhosis patients. The signature was proved to be an independent risk element for HCV-related cirrhosis patients. In addition, according to the time-dependent receiver operating characteristic (ROC) curves, nomogram, and calibration plot, the prognostic model could precisely forecast the survival rate at the first, fifth, and tenth year. Notably, functional enrichment analyses indicated that cytokine activity, chemokine activity, leukocyte migration and chemotaxis, chemokine signaling pathway and viral protein interaction with cytokine and cytokine receptor were involved in HCV-related cirrhosis progression. Moreover, ssGSEA analyses revealed fierce immune-inflammatory response mechanisms in HCV progress. Generally, our work developed a robust prognostic signature that can accurately predict the overall survival, Child-Pugh class progression, hepatic decompensation, and hepatocellular carcinoma (HCC) for HCV-related early-stage cirrhosis patients. Functional enrichment and further immune infiltration analyses systematically elucidated potential immune response mechanisms.
Collapse
Affiliation(s)
- Cheng Guo
- Department of Gastroenterology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chenglai Dong
- Department of Thoracic and Cardiovascular Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junjie Zhang
- Department of Gastroenterology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rui Wang
- Department of Gastroenterology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhe Wang
- Department of Gastroenterology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jie Zhou
- Department of Gastroenterology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wei Wang
- Department of Gastroenterology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bing Ji
- Department of Gastroenterology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Boyu Ma
- Department of Gastroenterology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanli Ge
- Department of Gastroenterology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhirong Wang
- Department of Gastroenterology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|