1
|
Ni Z, Tian X, Zhao W, Hu W, Lv J, Sun X, Zhang Y, Zhang Y, Zhang Y, Li B, Liu F. The detrimental effects and mechanisms of Orlistat in disrupting energy homeostasis and reproduction in Daphnia magna. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 279:107201. [PMID: 39657302 DOI: 10.1016/j.aquatox.2024.107201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Orlistat (ORL) has been employed as an anti-obesity pharmaceutical for several decades. Given its low absorption rate, the majority of administered ORL is excreted into the environment with feces. It is crucial to collect scientific information regarding the possible ecological risks associated with ORL. Here, the effects of ORL on Daphnia magna were evaluated using a 21-day chronic test at concentrations of 1, 10, 100, and 1000 μg/L. We found the inhibition of feeding and swimming activities in the 100 and 1000 μg/L ORL exposed D. magna, respectively. Their digestive enzyme activities and metabolites were reduced even at 1 μg/L ORL exposure. It is noteworthy that exposure to 100 μg/L ORL induced a decrease in the reproductive capacity of D. magna, although no discernible genotoxicity was observed. To identify the toxicological mechanisms of ORL, a metabolic analysis was conducted on D. magna exposed to 1000 μg/L ORL. A comprehensive reduction in carbohydrates, lipids, and amino acids was observed, which resulted in a blockage of metabolic flux towards the TCA cycle, as evidenced by mitochondrial dysfunction. These findings substantiate the detrimental impact of ORL on D. magna and provide insights into the underlying molecular mechanisms from a metabolic perspective.
Collapse
Affiliation(s)
- Zhihua Ni
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China; Hebei Basic Science Center for Biotic Interaction, Hebei University, Baoding, 071002, China
| | - Xinling Tian
- College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China
| | - Wenbo Zhao
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China
| | - Wenkai Hu
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China
| | - Jinghua Lv
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China
| | - Xiaoli Sun
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China
| | - Yajie Zhang
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China
| | - Yiwen Zhang
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China
| | - Yuming Zhang
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China; Hebei Basic Science Center for Biotic Interaction, Hebei University, Baoding, 071002, China
| | - Baoku Li
- College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China.
| | - Fengsong Liu
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China; Hebei Basic Science Center for Biotic Interaction, Hebei University, Baoding, 071002, China.
| |
Collapse
|
2
|
Xia Y, An Y, Jin R, Huang W, Jin GZ, Xu J. Expression and Clinical Significance of Nuclear Phosphoglucomutase-1 in Hepatocellular Carcinoma. Appl Immunohistochem Mol Morphol 2024; 32:476-483. [PMID: 39351772 DOI: 10.1097/pai.0000000000001225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 08/15/2024] [Indexed: 11/08/2024]
Abstract
This study aimed to evaluate the predictive values of phosphoglucomutase-1 (PGM1) expression for prognosis in patients with hepatocellular carcinoma (HCC). PGM1 expression was assessed by immunohistochemistry in tissue microarrays. The relationship of PGM1 expression level with pathologic parameters and prognosis values was respectively analyzed by χ 2 test and Cox regression. The accuracy of independent risk factors in predicting prognosis was calculated by receiver operating characteristic curve. HCC patient-derived xenograft models were performed to evaluate the nuclear PGM1 antitumor effect. The results showed that PGM1 expression was low in HCC tissues. Nuclear PGM1 was an independent prognostic factor for overall survival and time to recurrence. Cox regression showed that nuclear PGM1, serum α-fetoprotein, liver cirrhosis, and TNM staging stage were independent risk predictors for HCC. Receiver operating characteristic curve demonstrated that combination of independent predictors had better prognostic value than TNM staging alone. Moreover, patient-derived xenograft models showed antitumor effect of nuclear PGM1. We found that low expression of nuclear PGM1 was detected in HCC tissues and associated with poor prognostic. Nuclear PGM1 was an independent prognostic factor in patients with HCC. Furthermore, nuclear PGM1 combining other independent risk factors showed a better prognostic value. Nuclear PGM1 was a useful prognostic biomarker for patients with HCC.
Collapse
Affiliation(s)
- Yechen Xia
- Hongqiao International Institute of Medicine, Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai
| | - Yan An
- Hongqiao International Institute of Medicine, Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai
| | - Riming Jin
- Department of First Surgery, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University
| | - Wentao Huang
- Department of Pathology, Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai
| | - Guang-Zhi Jin
- Department of Pathology, Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai
| | - Jing Xu
- Department of Laboratory Medicine, Wuxi Traditional Chinese Medicine Hospital, Wuxi, China
| |
Collapse
|
3
|
Liu X, Zhang Q, Wang P, Peng X, An Y, Chen J, Huang J, Qin S, He H, Hao M, Tian J, Yi L, Lei M, Luo P, Wang J, Zhang X. Dissection of Targeting Molecular Mechanisms of Celastrol-induced Nephrotoxicity via A Combined Deconvolution Strategy of Chemoproteomics and Metabolomics. Int J Biol Sci 2024; 20:4601-4617. [PMID: 39309437 PMCID: PMC11414378 DOI: 10.7150/ijbs.91751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 07/05/2024] [Indexed: 09/25/2024] Open
Abstract
Celastrol (Cel), derived from the traditional herb Tripterygium wilfordii Hook. f., has anti-inflammatory, anti-tumor, and immunoregulatory activities. Renal dysfunction, including acute renal failure, has been reported in patients following the administration of Cel-relative medications. However, the functional mechanism of nephrotoxicity caused by Cel is unknown. This study featured combined use of activity-based protein profiling and metabolomics analysis to distinguish the targets of the nephrotoxic effects of Cel. Results suggest that Cel may bind directly to several critical enzymes participating in metabolism and mitochondrial functions. These enzymes include voltage-dependent anion-selective channel protein 1 (essential for maintaining mitochondrial configurational and functional stability), pyruvate carboxylase (involved in sugar isomerization and the tricarboxylic acid cycle), fatty acid synthase (related to β-oxidation of fatty acids), and pyruvate kinase M2 (associated with aerobic respiration). Proteomics and metabolomics analysis confirmed that Cel-targeted proteins disrupt some metabolic biosynthetic processes and promote mitochondrial dysfunction. Ultimately, Cel aggravated kidney cell apoptosis. These cumulative results deliver an insight into the potential mechanisms of Cel-caused nephrotoxicity. They may also facilitate development of antagonistic drugs to mitigate the harmful effects of Cel on the kidneys and improve its clinical applications.
Collapse
Affiliation(s)
- Xueying Liu
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
- Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Qian Zhang
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Peili Wang
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Xin Peng
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
- Ningbo Municipal Hospital of TCM, Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo 310060, China
| | - Yehai An
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Junhui Chen
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
| | - Jingnan Huang
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
| | - Shuanglin Qin
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, TCM Precision Medicine Research Department, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, Hunan 410007, China
| | - Hengkai He
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
| | - Mingjing Hao
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
| | - Jiahang Tian
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Letai Yi
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
- Inner Mongolia Medical University, Hohhot, 010107, China
| | - Ming Lei
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
- Department of Nephrology, Guangzhou Eighth People's Hospital, Guangzhou, Guangdong 510440, China
| | - Piao Luo
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, TCM Precision Medicine Research Department, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, Hunan 410007, China
| | - Jigang Wang
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Ningbo Municipal Hospital of TCM, Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo 310060, China
| | - Xinzhou Zhang
- Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
- Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| |
Collapse
|
4
|
Hao X, Zhu X, Tian H, Lai G, Zhang W, Zhou H, Liu S. Pharmacological effect and mechanism of orlistat in anti-tumor therapy: A review. Medicine (Baltimore) 2023; 102:e34671. [PMID: 37682175 PMCID: PMC10489489 DOI: 10.1097/md.0000000000034671] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/19/2023] [Indexed: 09/09/2023] Open
Abstract
Research has demonstrated that obesity is an important risk factor for cancer progression. Orlistat is a lipase inhibitor with promising therapeutic effects on obesity. In addition to being regarded as a slimming drug, a growing number of studies in recent years have suggested that orlistat has anti-tumor activities, while the underlying mechanism is still not well elucidated. This paper reviewed recent pharmacological effects and mechanisms of orlistat against tumors and found that orlistat can target cancer cells through activation or suppression of multiple signaling pathways. It can induce tumor cells apoptosis or death, interfere with tumor cells' cycles controlling, suppress fatty acid synthase activity, increase ferroptosis, inhibit tumor angiogenesis, and improve tumor cells glycolytic. Thus, this review may shed new light on anti-tumor mechanism and drug repurposing of orlistat, and anti-tumor drug development.
Collapse
Affiliation(s)
- Xiaoqing Hao
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
- School of Pharmacy & Clinical Pharmacy (School of Integrative Pharmacy), Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Xiaodi Zhu
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Huiqun Tian
- The Second People’s Hospital of China Three Gorges University, Yichang, People’s Republic of China
| | - Guanxi Lai
- School of Pharmacy & Clinical Pharmacy (School of Integrative Pharmacy), Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Wei Zhang
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Honghao Zhou
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Song Liu
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
- School of Pharmacy & Clinical Pharmacy (School of Integrative Pharmacy), Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| |
Collapse
|
5
|
Cuttica CM, Briata IM, DeCensi A. Novel Treatments for Obesity: Implications for Cancer Prevention and Treatment. Nutrients 2023; 15:3737. [PMID: 37686769 PMCID: PMC10490004 DOI: 10.3390/nu15173737] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
It is now established that obesity is related to a higher incidence of cancer during a lifespan. The effective treatment of obesity opens up new perspectives in the treatment of a relevant modifiable cancer risk factor. The present narrative review summarizes the correlations between weight loss in obesity and cancer. The current knowledge between obesity treatment and cancer was explored, highlighting the greatest potential for its use in the treatment of cancer in the clinical setting. Evidence for the effects of obesity therapy on proliferation, apoptosis, and response to chemotherapy is summarized. While more studies, including large, long-term clinical trials, are needed to adequately evaluate the relationship and durability between anti-obesity treatment and cancer, collaboration between oncologists and obesity treatment experts is increasingly important.
Collapse
Affiliation(s)
| | - Irene Maria Briata
- Division of Medical Oncology, E.O. Ospedali Galliera, 16128 Genoa, Italy; (I.M.B.); (A.D.)
| | - Andrea DeCensi
- Division of Medical Oncology, E.O. Ospedali Galliera, 16128 Genoa, Italy; (I.M.B.); (A.D.)
- Wolfson Institute of Population Health, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AD, UK
| |
Collapse
|
6
|
Zhao X, Li K, Chen M, Liu L. Metabolic codependencies in the tumor microenvironment and gastric cancer: Difficulties and opportunities. Biomed Pharmacother 2023; 162:114601. [PMID: 36989719 DOI: 10.1016/j.biopha.2023.114601] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Oncogenesis and the development of tumors affect metabolism throughout the body. Metabolic reprogramming (also known as metabolic remodeling) is a feature of malignant tumors that is driven by oncogenic changes in the cancer cells themselves as well as by cytokines in the tumor microenvironment. These include endothelial cells, matrix fibroblasts, immune cells, and malignant tumor cells. The heterogeneity of mutant clones is affected by the actions of other cells in the tumor and by metabolites and cytokines in the microenvironment. Metabolism can also influence immune cell phenotype and function. Metabolic reprogramming of cancer cells is the result of a convergence of both internal and external signals. The basal metabolic state is maintained by internal signaling, while external signaling fine-tunes the metabolic process based on metabolite availability and cellular needs. This paper reviews the metabolic characteristics of gastric cancer, focusing on the intrinsic and extrinsic mechanisms that drive cancer metabolism in the tumor microenvironment, and interactions between tumor cell metabolic changes and microenvironment metabolic changes. This information will be helpful for the individualized metabolic treatment of gastric cancers.
Collapse
|
7
|
Liu S, Deng Y, Yu Y, Xia X. Knock-down of PGM1 inhibits cell viability, glycolysis, and oxidative phosphorylation in glioma under low glucose condition via the Myc signaling pathway. Biochem Biophys Res Commun 2023; 656:38-45. [PMID: 36947965 DOI: 10.1016/j.bbrc.2023.03.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
PGM1 is an essential enzyme for glucose metabolism and is involved in cell viability, proliferation, and metabolism. However, the regulatory role of PGMI in glioma progression and the relation between gliomas and PGM1 expression are still unclear. This study aimed to explore the role of PGM1 in glycolysis and oxidative phosphorylation in glioma. Correlation and enrichment analyses of PGM1 in glioma cells were explored in TCGA database and two hospital cohorts. The cell viability, glycolysis, and oxidative phosphorylation were investigated in PGM1 knock-down and overexpression situations. Higher PGM1 expression in glioma patients was associated with a poor survival rate. However, knock-down of PGM1 reduced glioma cell viability, glycolysis, and oxidative phosphorylation under low glucose condition. Moreover, it suppressed tumor growth in vivo. On the other hand, PGM1 overexpression promoted glioma cell viability, glycolysis, and oxidative phosphorylation under low glucose condition by a Myc positive feedback loop. Glioma patients with higher PGM1 expression were associated with poor survival rates. Additionally, PGM1 could promote glioma cell viability, glycolysis, and oxidative phosphorylation under low glucose condition via a myc-positive feedback loop, suggesting PGM1 could be a potential therapeutic target for gliomas.
Collapse
Affiliation(s)
- Shenghua Liu
- Department of Neurosurgery, Santai Affiliated Hospital of North Sichuan Medical College, Mianyang, 621100, China
| | - Yuanyin Deng
- Department of Clinical Medicine, Zhejiang University City College School of Medicine, Hangzhou, 310015, China
| | - Yunhu Yu
- Department of Neurosurgery, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Xiangping Xia
- Department of Neurosurgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
8
|
Song HC, Xie CY, Kong Q, Wei L, Wang XT. Daylight ultraviolet B radiation ruptured the cell membrane, promoted nucleotide metabolism and inhibited energy metabolism in the plasma of Pacific oyster. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 862:160729. [PMID: 36496017 DOI: 10.1016/j.scitotenv.2022.160729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/28/2022] [Accepted: 12/02/2022] [Indexed: 06/17/2023]
Abstract
The increasing and intensifying ultraviolet B (UVB) radiation in sunlight is an environmental threat to aquatic ecosystems, potentially affecting the entire life cycle of wild or aquacultural Pacific oyster Crassostrea gigas with photoreception. Due to its complex composition, plasma is an important biological specimen for investigating the degree of disturbance from its steady state caused by the external environment in the open-pipe-type hemolymph of mollusks. We performed a multi-omic analysis of C. gigas plasma exposed to daylight UVB radiation. Hub differentially expressed genes (DEGs) and differentially expressed proteins (DEPs) were identified using the functional classification of Clusters of Orthologous Groups of proteins (COGs) through the protein-protein interaction (PPI)-based maximal clique centrality (MCC) algorithm. Our results summarize three types of UVB influences (disruption of the cell membrane, promotion of nucleotide metabolism, and inhibition of energy metabolism) on C. gigas based on transcriptomic, proteomic, and metabolomic analyses. The associated hub DEGs, DEPs (e.g., nucleoside diphosphate kinase, malate dehydrogenase, and hydroxyacyl-coenzyme A dehydrogenase), and metabolites (e.g., uridine, adenine, deoxyguanosine, guanosine, and xylitol) in the plasma were identified as biomarkers of mollusk response to UVB radiation, and could be used to evaluate the influence of environmental UVB on mollusks in future studies.
Collapse
Affiliation(s)
- Hong-Ce Song
- School of Agriculture, Ludong University, Yantai City, Shandong Province 264025, China
| | - Chao-Yi Xie
- School of Agriculture, Ludong University, Yantai City, Shandong Province 264025, China
| | - Qing Kong
- School of Agriculture, Ludong University, Yantai City, Shandong Province 264025, China
| | - Lei Wei
- School of Agriculture, Ludong University, Yantai City, Shandong Province 264025, China.
| | - Xiao-Tong Wang
- School of Agriculture, Ludong University, Yantai City, Shandong Province 264025, China.
| |
Collapse
|
9
|
Cao B, Zhao R, Li H, Xu X, Gao J, Chen L, Wei B. Inhibition of androgen receptor enhanced the anticancer effects of everolimus through targeting glucose transporter 12. Int J Biol Sci 2023; 19:104-119. [PMID: 36594084 PMCID: PMC9760431 DOI: 10.7150/ijbs.75106] [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: 05/14/2022] [Accepted: 10/08/2022] [Indexed: 11/24/2022] Open
Abstract
Everolimus was designed as a mammalian target of rapamycin (mTOR) inhibitor. It has been proven as a targeted drug for gastric cancer (GC) therapy. However, long-term treatment with everolimus may cause severe side effects for recipients. Decreasing the dosage and attenuating the associated risks are feasible to promote clinical translation of everolimus. This study aimed to identify the underlying mechanisms of responses to everolimus and develop novel regimens for GC treatment. Our findings proved that there was a significant dose-dependent relationship of everolimus-induced GC cell apoptosis and glycolysis inhibition. Then, we found that a member of glucose transporter (GLUT12) family, GLUT12, was actively upregulated to counteract the anticancer effects of everolimus. GLUT12 might be overexpressed in GC. High expression of GLUT12 might be correlated with tumor progression and short survival time of GC patients. Bioinformatic analysis suggested that GLUT12 might be involved in regulating cancer development and metabolism. The experiments proved that GLUT12 significantly promoted GC growth, glycolysis and impaired the anticancer effects of everolimus. Androgen receptor (AR) is a classical oncogenic factor in many types of cancer. Everolimus elevated GLUT12 expression in an AR-dependent manner. Inhibition of AR activity abrogated the promotive effects on GLUT12 expression. Both in-vitro and in-vivo experiments demonstrated that GLUT12 knockdown augmented anticancer effects of everolimus. Enzalutamide, an AR inhibitor, or AR knockdown was comparable to GLUT12 suppression. This study identified the role of the AR/GLUT12 pathway in the development of poor responses to everolimus. Interference with AR/GLUT12 pathway may serve as a promising approach to promoting the translational application of everolimus in GC therapy.
Collapse
Affiliation(s)
- Bo Cao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Chinese PLA, Beijing 100853, China
| | - Ruiyang Zhao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Chinese PLA, Beijing 100853, China
| | - Hanghang Li
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Chinese PLA, Beijing 100853, China
| | - Xingming Xu
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Jingwang Gao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Chinese PLA, Beijing 100853, China
| | - Lin Chen
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Chinese PLA, Beijing 100853, China.,✉ Corresponding authors: Bo Wei, MD, PhD, Chief Doctor, Professor, Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Tel: +86-10-66938071; E-mail: ; Lin Chen, MD, PhD, Chief Doctor, Professor, Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Tel: +86-10-66938066; E-mail:
| | - Bo Wei
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Chinese PLA, Beijing 100853, China.,✉ Corresponding authors: Bo Wei, MD, PhD, Chief Doctor, Professor, Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Tel: +86-10-66938071; E-mail: ; Lin Chen, MD, PhD, Chief Doctor, Professor, Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Tel: +86-10-66938066; E-mail:
| |
Collapse
|
10
|
Wang W, Mu M, Zou Y, Deng S, Lu Y, Li Q, Li Z, Tao H, Wang Y, Tao X. Glycogen metabolism reprogramming promotes inflammation in coal dust-exposed lung. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 242:113913. [PMID: 35907323 DOI: 10.1016/j.ecoenv.2022.113913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 06/15/2023]
Abstract
Long-term coal dust exposure triggers complex inflammatory processes in the coal workers' pneumoconiosis (CWP) lungs. The progress of the inflammation is reported to be affected by disordered cell metabolism. However, the changes in the metabolic reprogramming associated with the pulmonary inflammation induced by the coal dust particles are unknown. Herein, we show that coal dust exposure causes glycogen accumulation and the reprogramming of glucose metabolism in the CWP lung. The glycogen accumulation caused by coal dust is mainly due to macrophages, which reprogram glycogen metabolism and trigger an inflammatory response. In addition, 2-deoxy-D-glucose (2-DG) reduced glycogen content in macrophages, which was accompanied by mitigated inflammation and restrained NF-κB activation. Accordingly, we have pinpointed a novel and crucial metabolic pathway that is an essential regulator of the inflammatory phenotype of coal dust-exposed macrophages. These results shed light on new ways to regulate CWP inflammation.
Collapse
Affiliation(s)
- Wenyang Wang
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Min Mu
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, China
| | - Yuanjie Zou
- School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Songsong Deng
- Department of Clinical Laboratory, Chaoyang Hospital, Huainan, China
| | - Yuting Lu
- School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Qinglong Li
- School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Zeyu Li
- School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Huihui Tao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, China
| | - Yun Wang
- School of Bioengineering, Huainan Normal University, Huainan 232038, China
| | - Xinrong Tao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, China.
| |
Collapse
|
11
|
Key Molecules of Fatty Acid Metabolism in Gastric Cancer. Biomolecules 2022; 12:biom12050706. [PMID: 35625633 PMCID: PMC9138239 DOI: 10.3390/biom12050706] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 02/05/2023] Open
Abstract
Fatty acid metabolism is closely linked to the progression of gastric cancer (GC), a very aggressive and life-threatening tumor. This study examines linked molecules, such as Sterol Regulatory Element-Binding Protein 1 (SREBP1), ATP Citrate Lyase (ACLY), Acetyl-CoA Synthases (ACSs), Acetyl-CoA Carboxylase (ACC), Fatty Acid Synthase (FASN), Stearoyl-CoA Desaturase 1 (SCD1), CD36, Fatty Acid Binding Proteins (FABPs), and Carnitine palmitoyltransferase 1 (CPT1), as well as their latest studies and findings in gastric cancer to unveil its core mechanism. The major enzymes of fatty acid de novo synthesis are ACLY, ACSs, ACC, FASN, and SCD1, while SREBP1 is the upstream molecule of fatty acid anabolism. Fatty acid absorption is mediated by CD36 and FABPs, and fatty acid catabolism is mediated by CPT1. If at all possible, we will discover novel links between fatty acid metabolism and a prospective gastric cancer target.
Collapse
|
12
|
Fang Y, Pei S, Huang K, Xu F, Xiang G, Lan L, Zheng X. Single-Cell Transcriptome Reveals the Metabolic and Clinical Features of a Highly Malignant Cell Subpopulation in Pancreatic Ductal Adenocarcinoma. Front Cell Dev Biol 2022; 10:798165. [PMID: 35252177 PMCID: PMC8894596 DOI: 10.3389/fcell.2022.798165] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/17/2022] [Indexed: 12/13/2022] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is a malignant tumor with a high mortality rate. PDAC exhibits significant heterogeneity as well as alterations in metabolic pathways that are associated with its malignant progression. In this study, we explored the metabolic and clinical features of a highly malignant subgroup of PDAC based on single-cell transcriptome technology.Methods: A highly malignant cell subpopulation was identified at single-cell resolution based on the expression of malignant genes. The metabolic landscape of different cell types was analyzed based on metabolic pathway gene sets. In vitro experiments to verify the biological functions of the marker genes were performed. PDAC patient subgroups with highly malignant cell subpopulations were distinguished according to five glycolytic marker genes. Five glycolytic highly malignant-related gene signatures were used to construct the glycolytic highly malignant-related genes signature (GHS) scores.Results: This study identified a highly malignant tumor cell subpopulation from the single-cell RNA sequencing (scRNA-seq) data. The analysis of the metabolic pathway revealed that highly malignant cells had an abnormally active metabolism, and enhanced glycolysis was a major metabolic feature. Five glycolytic marker genes that accounted for the highly malignant cell subpopulations were identified, namely, EN O 1, LDHA, PKM, PGK1, and PGM1. An in vitro cell experiment showed that proliferation rates of PANC-1 and CFPAC-1 cell lines decreased after knockdown of these five genes. Patients with metabolic profiles of highly malignant cell subpopulations exhibit clinical features of higher mortality, higher mutational burden, and immune deserts. The GHS score evaluated using the five marker genes was an independent prognostic factor for patients with PDAC.Conclusion: We revealed a subpopulation of highly malignant cells in PDAC with enhanced glycolysis as the main metabolic feature. We obtained five glycolytic marker gene signatures, which could be used to identify PDAC patient subgroups with highly malignant cell subpopulations, and proposed a GHS prognostic score.
Collapse
Affiliation(s)
- Yangyang Fang
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- School of Laboratory Medical and Life Science, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Shunjie Pei
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- School of Laboratory Medical and Life Science, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Kaizhao Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Feng Xu
- School of Laboratory Medical and Life Science, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Guangxin Xiang
- School of Laboratory Medical and Life Science, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Linhua Lan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Linhua Lan, ; Xiaoqun Zheng,
| | - Xiaoqun Zheng
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- School of Laboratory Medical and Life Science, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Linhua Lan, ; Xiaoqun Zheng,
| |
Collapse
|