1
|
Huang T, Zhou MY, Zou GL, Hu RH, Han L, Zhang QX, Zhao XK. Focal adhesion kinase promotes aerobic glycolysis in hepatic stellate cells via the cyclin D1/c-Myc/MCT-1 pathway to induce liver fibrosis. Sci Rep 2025; 15:4552. [PMID: 39915293 PMCID: PMC11802747 DOI: 10.1038/s41598-025-88538-8] [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: 09/11/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
Hepatic stellate cells (HSCs) transdifferentiate into myofibroblasts during liver fibrosis and exhibit increased glycolysis. Phosphorylated focal adhesion kinase (FAK) (pY397-FAK) promotes monocarboxylate transporter 1 (MCT-1) expression in HSCs to increase aerobic glycolysis and cause liver fibrosis. A combined multiomics analysis of C57BL/6 mice with tetrachloromethane (CCl4)-induced liver fibrosis was performed to identify the downstream FAK signaling pathway. The effect of the FAK inhibitor PF562271 on CCl4-induced liver fibrosis was explored by immunofluorescence of liver tissues. The migration, proliferation and aerobic glycolysis of LX-2 cells after stimulation and activation by transforming growth factor beta-1 (TGF-β1) or suppression by PF562271 was assessed in vitro. Multiomics analysis of a successfully generated CCl4-induced liver fibrosis mouse model was performed. FAK and cyclin D1 were significantly enriched in mice with CCl4-induced liver fibrosis. In vivo, the MCT-1 and alpha smooth muscle actin (α-SMA) levels were increased in mice with CCl4-induced liver fibrosis, and MCT-1 and α-SMA expression decreased after PF562271 treatment. In vitro, PF562271 alleviated TGF-β1-induced LX-2 activation. LX-2 cells showed diminished migration, proliferation, and aerobic glycolysis after PF562271 intervention. FAK promotes aerobic glycolysis in LX-2 cells through the cyclin D1/c-Myc/MCT-1 pathway, thereby increasing liver fibrosis.
Collapse
Affiliation(s)
- Tao Huang
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Ming-Yu Zhou
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Gao-Liang Zou
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Rui-Han Hu
- Department of Cardiology, Guiqian International General Hospital, Guiyang, Guizhou Province, China
| | - Lu Han
- Department of Comprehensive Ward, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Qing-Xiu Zhang
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Xue-Ke Zhao
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China.
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, No. 9 Beijing Road, Guiyang, 550004, Guizhou Province, China.
| |
Collapse
|
2
|
Liu Y, Yan Z, Liu C, Yang R, Zheng Q, Jian J, Wang M, Wang L, Weng X, Chen Z, Liu X. Integrated RNA sequencing analysis and machine learning identifies a metabolism-related prognostic signature in clear cell renal cell carcinoma. Sci Rep 2025; 15:1691. [PMID: 39799252 PMCID: PMC11724983 DOI: 10.1038/s41598-025-85618-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025] Open
Abstract
The connection between metabolic reprogramming and tumor progression has been demonstrated in an increasing number of researches. However, further research is required to identify how metabolic reprogramming affects interpatient heterogeneity and prognosis in clear cell renal cell carcinoma (ccRCC). In this work, single-cell RNA sequencing (scRNA-seq) based deconvolution was utilized to create a malignant cell hierarchy with metabolic differences and to investigate the relationship between metabolic biomarkers and prognosis. Simultaneously, we created a machine learning-based approach for creating metabolism-related prognostic signature (MRPS). Gamma-glutamyltransferase 6 (GGT6) was further explored for deep biological insights through in vitro experiments. Compared to 51 published signatures and conventional clinical features, MRPS showed substantially higher accuracy. Meanwhile, high MRPS-risk samples demonstrated an immunosuppressive phenotype with more infiltrations of regulatory T cell (Treg) and tumour-associated macrophage (TAM). Following the administration of immune checkpoint inhibitors (ICIs), MRPS showed consistent and strong performance and was an independent risk factor for overall survival. GGT6, an essential metabolic indicator and component of MRPS, has been proven to support proliferation and invasion in ccRCC. MRPS has the potential to be a highly effective tool in improving the clinical results of patients with ccRCC.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/mortality
- Carcinoma, Renal Cell/drug therapy
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Kidney Neoplasms/mortality
- Kidney Neoplasms/drug therapy
- Machine Learning
- Prognosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Sequence Analysis, RNA/methods
- Male
- Female
- Gene Expression Regulation, Neoplastic
- Middle Aged
- Single-Cell Analysis
- gamma-Glutamyltransferase/metabolism
- gamma-Glutamyltransferase/genetics
- Cell Line, Tumor
Collapse
Affiliation(s)
- Yunxun Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhiwei Yan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Cheng Liu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Rui Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qingyuan Zheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jun Jian
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Minghui Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaodong Weng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
3
|
Zhang F, Gu T, Li J, Zhu Y, Chu M, Zhou Q, Liu J. Emodin regulated lactate metabolism by inhibiting MCT1 to delay non-small cell lung cancer progression. Hum Cell 2024; 38:11. [PMID: 39465441 DOI: 10.1007/s13577-024-01140-4] [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: 06/25/2024] [Accepted: 09/09/2024] [Indexed: 10/29/2024]
Abstract
Lung cancer is one of the most common malignant tumors in the world, with high incidence rate and mortality. Monocarboxylate transporter (MCT) 1 has been found to be widely expressed in various tumors and plays a crucial role in regulating energy metabolism. Emodin, as an important traditional Chinese medicine in China, has been reported to inhibit the progression of lung cancer. However, its potential mechanism has not been fully elucidated. The effects of emodin and MCT1 inhibitor AZD3965 on the proliferation, migration, and invasion of lung cancer cells were detected using cell counting kit-8 (CCK-8) assay, wound-healing assay, and transwell small chamber assay. The content of glucose, lactate, and pyruvate in the cell culture medium was detected using a glucose, lactate, and pyruvate detection kit, and also detected protein expression using western blotting. In addition, to investigate the effects of emodin and AZD3965 on lung cancer in vivo, we constructed nude mice subcutaneous transplant tumor model by subcutaneous injection of lung cancer cells. The results showed that emodin and AZD3965 could inhibit the proliferation, migration, and invasion of lung cancer cells. At the same time, they could inhibit the expression of MCT1 in lung cancer cells and promote the release of lactate, but did not affect the content of glucose and pyruvate. In vivo experiments had shown that emodin and AZD3965 could effectively inhibit the growth of lung cancer and inhibit the expression of MCT1. All in all, our data suggested that emodin inhibited the proliferation, migration, and invasion of lung cancer cells, possibly by inhibiting MCT1, providing important theoretical basis for elucidating the mechanism of emodin in treating lung cancer.
Collapse
Affiliation(s)
- Fei Zhang
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Tian Gu
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Jin Li
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Yanqiu Zhu
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Mingliang Chu
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China.
| | - Qing Zhou
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China.
| | - Jiemin Liu
- Department of Endoscopy, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
| |
Collapse
|
4
|
Zhao W, Ouyang C, Zhang L, Wang J, Zhang J, Zhang Y, Huang C, Xiao Q, Jiang B, Lin F, Zhang C, Zhu M, Xie C, Huang X, Zhang B, Zhao W, He J, Chen S, Liu X, Lin D, Li Q, Wang Z. The proto-oncogene tyrosine kinase c-SRC facilitates glioblastoma progression by remodeling fatty acid synthesis. Nat Commun 2024; 15:7455. [PMID: 39198451 PMCID: PMC11358276 DOI: 10.1038/s41467-024-51444-0] [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: 01/05/2024] [Accepted: 08/08/2024] [Indexed: 09/01/2024] Open
Abstract
Increased fatty acid synthesis benefits glioblastoma malignancy. However, the coordinated regulation of cytosolic acetyl-CoA production, the exclusive substrate for fatty acid synthesis, remains unclear. Here, we show that proto-oncogene tyrosine kinase c-SRC is activated in glioblastoma and remodels cytosolic acetyl-CoA production for fatty acid synthesis. Firstly, acetate is an important substrate for fatty acid synthesis in glioblastoma. c-SRC phosphorylates acetyl-CoA synthetase ACSS2 at Tyr530 and Tyr562 to stimulate the conversion of acetate to acetyl-CoA in cytosol. Secondly, c-SRC inhibits citrate-derived acetyl-CoA synthesis by phosphorylating ATP-citrate lyase ACLY at Tyr682. ACLY phosphorylation shunts citrate to IDH1-catalyzed NADPH production to provide reducing equivalent for fatty acid synthesis. The c-SRC-unresponsive double-mutation of ACSS2 and ACLY significantly reduces fatty acid synthesis and hampers glioblastoma progression. In conclusion, this remodeling fulfills the dual needs of glioblastoma cells for both acetyl-CoA and NADPH in fatty acid synthesis and provides evidence for glioma treatment by c-SRC inhibition.
Collapse
Affiliation(s)
- Wentao Zhao
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
| | - Cong Ouyang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Liang Zhang
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jinyang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Jiaojiao Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Yan Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Chen Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Qiao Xiao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Bin Jiang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Furong Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Cixiong Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Mingxia Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Changchuan Xie
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Xi Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Bingchang Zhang
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wenpeng Zhao
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jiawei He
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Sifang Chen
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiyao Liu
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Donghai Lin
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Qinxi Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
| | - Zhanxiang Wang
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
5
|
Zhao J, Jin D, Huang M, Ji J, Xu X, Wang F, Zhou L, Bao B, Jiang F, Xu W, Lu X, Xiao M. Glycolysis in the tumor microenvironment: a driver of cancer progression and a promising therapeutic target. Front Cell Dev Biol 2024; 12:1416472. [PMID: 38933335 PMCID: PMC11199735 DOI: 10.3389/fcell.2024.1416472] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Even with sufficient oxygen, tumor cells use glycolysis to obtain the energy and macromolecules they require to multiply, once thought to be a characteristic of tumor cells known as the "Warburg effect". In fact, throughout the process of carcinogenesis, immune cells and stromal cells, two major cellular constituents of the tumor microenvironment (TME), also undergo thorough metabolic reprogramming, which is typified by increased glycolysis. In this review, we provide a full-scale review of the glycolytic remodeling of several types of TME cells and show how these TME cells behave in the acidic milieu created by glucose shortage and lactate accumulation as a result of increased tumor glycolysis. Notably, we provide an overview of putative targets and inhibitors of glycolysis along with the viability of using glycolysis inhibitors in combination with immunotherapy and chemotherapy. Understanding the glycolytic situations in diverse cells within the tumor immunological milieu will aid in the creation of subsequent treatment plans.
Collapse
Affiliation(s)
- Junpeng Zhao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Dandan Jin
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Mengxiang Huang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Jie Ji
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Xuebing Xu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Fei Wang
- Department of Laboratory Medicine, Affiliated Hospital and Medical School of Nantong University, Nantong, Jiangsu, China
| | - Lirong Zhou
- Department of Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Baijun Bao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Feng Jiang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Weisong Xu
- Department of Gastroenterology, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xiaomin Lu
- Department of Oncology Affiliated Haian Hospital of Nantong University, Nantong, Jiangsu, China
| | - Mingbing Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
- Department of Laboratory Medicine, Affiliated Hospital and Medical School of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
6
|
Koltai T, Fliegel L. Exploring monocarboxylate transporter inhibition for cancer treatment. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:135-169. [PMID: 38464385 PMCID: PMC10918235 DOI: 10.37349/etat.2024.00210] [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: 10/02/2023] [Accepted: 12/01/2023] [Indexed: 03/12/2024] Open
Abstract
Cells are separated from the environment by a lipid bilayer membrane that is relatively impermeable to solutes. The transport of ions and small molecules across this membrane is an essential process in cell biology and metabolism. Monocarboxylate transporters (MCTs) belong to a vast family of solute carriers (SLCs) that facilitate the transport of certain hydrophylic small compounds through the bilipid cell membrane. The existence of 446 genes that code for SLCs is the best evidence of their importance. In-depth research on MCTs is quite recent and probably promoted by their role in cancer development and progression. Importantly, it has recently been realized that these transporters represent an interesting target for cancer treatment. The search for clinically useful monocarboxylate inhibitors is an even more recent field. There is limited pre-clinical and clinical experience with new inhibitors and their precise mechanism of action is still under investigation. What is common to all of them is the inhibition of lactate transport. This review discusses the structure and function of MCTs, their participation in cancer, and old and newly developed inhibitors. Some suggestions on how to improve their anticancer effects are also discussed.
Collapse
Affiliation(s)
- Tomas Koltai
- Hospital del Centro Gallego de Buenos Aires, Buenos Aires 2199, Argentina
| | - Larry Fliegel
- Department of Biochemistry, Faculty of Medicine, University of Alberta, Edmonton T6G 2R3, Alberta, Canada
| |
Collapse
|
7
|
Chen L, Li Y, Deng X. Comprehensive analysis of pan-cancer reveals the potential of SLC16A1 as a prognostic and immunological biomarker. Medicine (Baltimore) 2023; 102:e33242. [PMID: 36930112 PMCID: PMC10019278 DOI: 10.1097/md.0000000000033242] [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: 11/11/2022] [Accepted: 02/20/2023] [Indexed: 03/18/2023] Open
Abstract
SLC16A1 plays an important role in the development of multiple cancer types. Pan-cancer analysis may have significant impacts on the exploration of the relationship between SLC16A1 gene expression, prognosis and the molecular mechanisms of tumorigenesis. In this study, through the analysis of TCGA and GEO datasets, we explored the expression level and survival prognosis of SLC16A1 in pan-cancer, and further explored the differences in SLC16A1 gene mutation, methylation, and phosphorylation between tumor and normal tissues. In addition, we focused on the biological function of this gene and the relationship between the prognosis and immune infiltration by immune infiltration analysis and enrichment analysis, in order to evaluate the diagnostic and prognostic significance of SLC16A1 in carcinomas. The study found that SLC16A1 was highly expressed in 14 kinds of tumors, and there were statistically significant differences in the prognosis of 9 tumors. The phosphorylation level of S467 increased in OV, RCC, and UCEC. There was a statistically negative correlation between the CD8+ T-cell infiltration level and the SLC16A1 expression in HNSC, LUSC, SARC, TGCT, and KIRC. The cancer-related fibroblasts were positively correlated with SLC16A1 expression in BLCA, BRCA, KIRC, KIRP, PAAD, PCPG, and THCA. The enrichment analysis indicated that the tumorigenesis mechanism of this gene was mainly related to "glycolysis and glucose metabolism synthesis." SLC16A1 was a promising prognostic and immunological biomarker in pan-cancer.
Collapse
Affiliation(s)
- Lingyun Chen
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
- Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Yang Li
- Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Xinna Deng
- Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
8
|
Zhou D, Duan Z, Li Z, Ge F, Wei R, Kong L. The significance of glycolysis in tumor progression and its relationship with the tumor microenvironment. Front Pharmacol 2022; 13:1091779. [PMID: 36588722 PMCID: PMC9795015 DOI: 10.3389/fphar.2022.1091779] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022] Open
Abstract
It is well known that tumor cells rely mainly on aerobic glycolysis for energy production even in the presence of oxygen, and glycolysis is a known modulator of tumorigenesis and tumor development. The tumor microenvironment (TME) is composed of tumor cells, various immune cells, cytokines, and extracellular matrix, among other factors, and is a complex niche supporting the survival and development of tumor cells and through which they interact and co-evolve with other tumor cells. In recent years, there has been a renewed interest in glycolysis and the TME. Many studies have found that glycolysis promotes tumor growth, metastasis, and chemoresistance, as well as inhibiting the apoptosis of tumor cells. In addition, lactic acid, a metabolite of glycolysis, can also accumulate in the TME, leading to reduced extracellular pH and immunosuppression, and affecting the TME. This review discusses the significance of glycolysis in tumor development, its association with the TME, and potential glycolysis-targeted therapies, to provide new ideas for the clinical treatment of tumors.
Collapse
Affiliation(s)
- Daoying Zhou
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China,Department of Provincial Clinical College, Wannan Medical College, Wuhu, China
| | - Zhen Duan
- Function Examination Center, Anhui Chest Hospital, Hefei, China
| | - Zhenyu Li
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China,Department of Provincial Clinical College, Wannan Medical College, Wuhu, China
| | - Fangfang Ge
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China,Department of Provincial Clinical College, Wannan Medical College, Wuhu, China
| | - Ran Wei
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lingsuo Kong
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China,*Correspondence: Lingsuo Kong,
| |
Collapse
|
9
|
Epigenetic connection between gut microbiota-derived short-chain fatty acids and chromatin histone modification in kidney diseases. Chin Med J (Engl) 2022; 135:1692-1694. [PMID: 36193977 PMCID: PMC9509131 DOI: 10.1097/cm9.0000000000002295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
10
|
Abstract
Renal cell carcinoma (RCC) is the sixth most common cancer among men and the ninth among women, and its prognosis is closely correlated with metastasis. Targeted therapy and immunotherapy are the main adjuvant treatments for advanced RCC and require early diagnosis, precise assessment, and prediction of the therapeutic responses. Current conventional imaging methods of RCC only provide structural information rather than biological processes. Noninvasive diagnostic tools are therefore needed to image RCC early and accurately at the molecular level. Nuclear medicine imaging combines the high sensitivity of radionuclides with the high resolution of structural imaging to visualize the metabolic processes and specific targets of RCC for more accurate and reliable diagnosis, staging, prognosis prediction, and response assessment. This review summarizes the most recent applications of nuclear medicine receptor imaging and metabolic imaging in RCC and highlights future development perspectives in the field.
Collapse
Affiliation(s)
- Qianyun Wu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| |
Collapse
|
11
|
Li F, Wang S, Yao Y, Sun X, Wang X, Wang N, You Y, Zhang Y. Visual analysis on the research of monocarboxylate transporters based on CiteSpace. Medicine (Baltimore) 2021; 100:e27466. [PMID: 34871210 PMCID: PMC8568392 DOI: 10.1097/md.0000000000027466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 09/20/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Monocarboxylate transports (MCTs), a family of solute carrier protein, play an important role in maintenance of cellular stability in tumor cells by mediating lactate exchange across membranes. The objective of this paper is to evaluate the knowledge structure, development trend, and research hotspot of MCTs research field systematically and comprehensively. METHODS Based on the 1526 publications from 2010 to 2020 retrieved from "Web of Science Core Collection" (WoSCC), we visually analyzed the MCTs research in terms of subject category, scientific collaboration network, keywords, and high-frequency literature using CiteSpace. RESULTS The number of publications exhibits an upward trend from 2010 to 2020 and the top 5 countries in the MCTs research were the United States, China, Japan, Germany, and England. Visser TJ was the most prolific author, while Halestrap AP was the most influential author with the highest citations. Analysis of the 7 cluster units from the co-cited references and keywords revealed that high expression of MCTs induced by oxidative stress and glycolysis was the pivotal point in the MCTs research field, while regulation of metabolism in tumor microenvironment, prognostic markers of cancer, and targeted inhibitors are the top 3 research frontiers topics. CONCLUSION This study will help the new researcher to understand the MCTs related field, master the research frontier, and obtain valuable scientific information, thus providing directions for follow-up research.
Collapse
Affiliation(s)
- Feifei Li
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Shuqi Wang
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Youlong Yao
- Department of computer science, Jinan Vocational College, Shandong, China
| | - Xueming Sun
- Weifang Yidu Central Hospital, Weifang, Shandong, China
| | - Xiaoyan Wang
- Weifang Yidu Central Hospital, Weifang, Shandong, China
| | - Ning Wang
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yulin You
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yanli Zhang
- Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|