1
|
Wang M, Qu L, Du X, Song P, Ng JPL, Wong VKW, Law BYK, Fu X. Natural Products and Derivatives Targeting Metabolic Reprogramming in Colorectal Cancer: A Comprehensive Review. Metabolites 2024; 14:490. [PMID: 39330497 PMCID: PMC11433951 DOI: 10.3390/metabo14090490] [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: 07/17/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Metabolic reprogramming is a critical pathogenesis of colorectal cancer (CRC), referring to metabolic disorders that cancer cells make in response to the stimulating pressure. Metabolic reprogramming induces changes in genetic material and promotes CRC progression and has been proven to be an efficient target of CRC. As natural products have garnered interest due to notable pharmacological effects and potential in counteracting chemoresistance, an increasing body of research is delving into the impact of these natural products on the metabolic reprogramming associated with CRC. In this review, we collected published data from the Web of Science and PubMed, covering the period from January 1980 to October 2023. This article focuses on five central facets of metabolic alterations in cancer cells, glucose metabolism, mitochondrial oxidative phosphorylation (OXPHOS), amino acid metabolism, fatty acid synthesis, and nucleotide metabolism, to provide an overview of recent advancements in natural product interventions targeting metabolic reprogramming in CRC. Our analysis underscores the potential of natural products in disrupting the metabolic pathways of CRC, suggesting promising therapeutic targets for CRC and expanding treatment options for metabolic-associated ailments.
Collapse
Affiliation(s)
- Mengyu Wang
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Liqun Qu
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medical Sciences Shandong University of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine’s Deep Development and Industrialization, Qingdao 266114, China
| | - Xinying Du
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medical Sciences Shandong University of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine’s Deep Development and Industrialization, Qingdao 266114, China
| | - Peng Song
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jerome P. L. Ng
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
| | - Vincent Kam Wai Wong
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
| | - Betty Yuen Kwan Law
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
| | - Xianjun Fu
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medical Sciences Shandong University of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine’s Deep Development and Industrialization, Qingdao 266114, China
| |
Collapse
|
2
|
Das C, Bhattacharya A, Adhikari S, Mondal A, Mondal P, Adhikary S, Roy S, Ramos K, Yadav KK, Tainer JA, Pandita TK. A prismatic view of the epigenetic-metabolic regulatory axis in breast cancer therapy resistance. Oncogene 2024; 43:1727-1741. [PMID: 38719949 PMCID: PMC11161412 DOI: 10.1038/s41388-024-03054-9] [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: 12/15/2023] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Abstract
Epigenetic regulation established during development to maintain patterns of transcriptional expression and silencing for metabolism and other fundamental cell processes can be reprogrammed in cancer, providing a molecular mechanism for persistent alterations in phenotype. Metabolic deregulation and reprogramming are thus an emerging hallmark of cancer with opportunities for molecular classification as a critical preliminary step for precision therapeutic intervention. Yet, acquisition of therapy resistance against most conventional treatment regimens coupled with tumor relapse, continue to pose unsolved problems for precision healthcare, as exemplified in breast cancer where existing data informs both cancer genotype and phenotype. Furthermore, epigenetic reprograming of the metabolic milieu of cancer cells is among the most crucial determinants of therapeutic resistance and cancer relapse. Importantly, subtype-specific epigenetic-metabolic interplay profoundly affects malignant transformation, resistance to chemotherapy, and response to targeted therapies. In this review, we therefore prismatically dissect interconnected epigenetic and metabolic regulatory pathways and then integrate them into an observable cancer metabolism-therapy-resistance axis that may inform clinical intervention. Optimally coupling genome-wide analysis with an understanding of metabolic elements, epigenetic reprogramming, and their integration by metabolic profiling may decode missing molecular mechanisms at the level of individual tumors. The proposed approach of linking metabolic biochemistry back to genotype, epigenetics, and phenotype for specific tumors and their microenvironment may thus enable successful mechanistic targeting of epigenetic modifiers and oncometabolites despite tumor metabolic heterogeneity.
Collapse
Affiliation(s)
- Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India.
- Homi Bhabha National Institute, Mumbai, 400094, India.
| | - Apoorva Bhattacharya
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
| | - Swagata Adhikari
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Atanu Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Payel Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Santanu Adhikary
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, 700032, India
| | - Siddhartha Roy
- Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, 700032, India
| | - Kenneth Ramos
- Center for Genomics and Precision Medicine, Texas A&M University, School of Medicine, Houston, TX, 77030, USA
| | - Kamlesh K Yadav
- Center for Genomics and Precision Medicine, Texas A&M University, School of Medicine, Houston, TX, 77030, USA
- School of Engineering Medicine, Texas A&M University, School of Medicine, Houston, TX, 77030, USA
| | - John A Tainer
- The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Tej K Pandita
- Center for Genomics and Precision Medicine, Texas A&M University, School of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
3
|
Abu Nayem SM, Islam S, Shah SS, Awal A, Ghann W, Anand D, Ahmad I, Uddin J, Aziz MA, Saleh Ahammad AJ. Biocompatible Gold Nanoparticles-Modified Fluorine Doped Tin Oxide Electrode for the Fabrication of Enzyme-Free Glucose Sensor. Chem Asian J 2024; 19:e202400074. [PMID: 38545693 DOI: 10.1002/asia.202400074] [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: 01/22/2024] [Revised: 03/13/2024] [Indexed: 04/24/2024]
Abstract
This work demonstrates the use of jute stick extract as a reducing and stabilizing agent for the synthesis of spherical gold nanoparticles (AuNPs). In UV-Vis spectroscopy, peak at 550 nm was used to confirm the formation of AuNPs. The spherical surface morphology of AuNPs was determined through SEM and TEM analysis. While XRD investigation revealed the crystallinity of the prepared AuNPs. To ensure the biocompatibility of synthesized AuNPs, a bacterial investigation was conducted with negative results towards bacterial strain. The, modified FTO with AuNPs were able to detect glucose in CV analysis and the constructed sensor displayed a wide linear range of 50 μM to 40 mM with a detection limit of 20 μM. Scan rate analysis was performed to determine the charge transfer coefficient (0.42) and Tafel slope (102 mV/decade). Furthermore, the interfacial surface mechanism is illustrated to understand the interaction of glucose with the electrode surface in an alkaline medium and the product formation through the dehydrogenation and hydrolysis process. The prepared sensor also showed good stability, reproducibility, and anti-interference capabilities. In the case of real sample analysis, we used a blood serum sample. A low RSD value (<10 %) suggests the practical use of AuNPs/FTO in real-life applications.
Collapse
Affiliation(s)
- S M Abu Nayem
- Department of Chemistry, Jagannath University, Dhaka, 1100, Bangladesh Tel
| | - Santa Islam
- Department of Chemistry, Jagannath University, Dhaka, 1100, Bangladesh Tel
| | - Syed Shaheen Shah
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto, 615-8520, Japan
| | - Abdul Awal
- Department of Chemistry, Jagannath University, Dhaka, 1100, Bangladesh Tel
| | - William Ghann
- Center for Nanotechnology, Department of Natural Sciences, Coppin State University, 2500 W. North Ave, Baltimore, MD, USA
| | - Deepak Anand
- Department of Bioengineering, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia
| | - Irshad Ahmad
- Department of Bioengineering, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia
- Interdisciplinary Research Center for Membranes and Water Security, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia
| | - Jamal Uddin
- Center for Nanotechnology, Department of Natural Sciences, Coppin State University, 2500 W. North Ave, Baltimore, MD, USA
| | - Md Abdul Aziz
- Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management (IRC- HTCM), King Fahd University of Petroleum & Minerals, KFUPM Box 5040, Dhahran, 31261, Saudi Arabia Tel
| | - A J Saleh Ahammad
- Department of Chemistry, Jagannath University, Dhaka, 1100, Bangladesh Tel
| |
Collapse
|
4
|
Zhang Y, Li L, Chu F, Wu H, Xiao X, Ye J, Li K. Itraconazole inhibits tumor growth via CEBPB-mediated glycolysis in colorectal cancer. Cancer Sci 2024; 115:1154-1169. [PMID: 38278779 PMCID: PMC11007002 DOI: 10.1111/cas.16082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/20/2023] [Accepted: 01/08/2024] [Indexed: 01/28/2024] Open
Abstract
Advanced colorectal cancer (CRC) is characterized by a high recurrence and metastasis rate, which is the primary cause of patient mortality. Unfortunately, effective anti-cancer drugs for CRC are still lacking in clinical practice. We screened FDA-approved drugs by utilizing targeted organoid sequencing data and found that the antifungal drug itraconazole had a potential therapeutic effect on CRC tumors. However, the effect and mechanism of itraconazole on CRC tumors have not been investigated. A cell line-derived xenograft model in tumor-bearing mice was established and single-cell RNA sequencing was performed on tumor samples from four mice with or without itraconazole treatment. The proportion of cell populations and gene expression profiles was significantly different between the two groups. We found that itraconazole could inhibit tumor growth and glycolysis. We revealed that CEBPB was a new target for itraconazole, and that silencing CEBPB could repress CRC glycolysis and tumor growth by inhibiting ENO1 expression. Clinical analysis showed that CEBPB expression was obviously elevated in CRC patients, and was associated with poor survival. In summary, itraconazole treatment remodeled cell composition and gene expression profiles. Itraconazole inhibited cell glycolysis and tumor growth via the CEBPB-ENO1 axis. In this study, we illustrate a new energy metabolism mechanism for itraconazole on tumor growth in CRC that will provide a theoretical basis for CRC targeting/combination therapy.
Collapse
Affiliation(s)
- Yong Zhang
- Branch Center of Advanced Medical Research CenterZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
- Department of GastroenterologyZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
- Medical Key Laboratory for Diagnosis and Treatment of Colorectal Cancer in Henan ProvinceZhengzhouChina
- Zhengzhou Key Laboratory for Diagnosis, Treatment and Research of Colorectal CancerZhengzhouChina
| | - Lu Li
- Department of GastroenterologyZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
- Medical Key Laboratory for Diagnosis and Treatment of Colorectal Cancer in Henan ProvinceZhengzhouChina
- Zhengzhou Key Laboratory for Diagnosis, Treatment and Research of Colorectal CancerZhengzhouChina
| | - Feifei Chu
- Department of GastroenterologyZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
- Medical Key Laboratory for Diagnosis and Treatment of Colorectal Cancer in Henan ProvinceZhengzhouChina
- Zhengzhou Key Laboratory for Diagnosis, Treatment and Research of Colorectal CancerZhengzhouChina
| | - Huili Wu
- Department of GastroenterologyZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
- Medical Key Laboratory for Diagnosis and Treatment of Colorectal Cancer in Henan ProvinceZhengzhouChina
- Zhengzhou Key Laboratory for Diagnosis, Treatment and Research of Colorectal CancerZhengzhouChina
| | - Xingguo Xiao
- Department of GastroenterologyZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
- Medical Key Laboratory for Diagnosis and Treatment of Colorectal Cancer in Henan ProvinceZhengzhouChina
- Zhengzhou Key Laboratory for Diagnosis, Treatment and Research of Colorectal CancerZhengzhouChina
| | - Jianping Ye
- Branch Center of Advanced Medical Research CenterZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
| | - Kunkun Li
- Department of GastroenterologyZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
- Medical Key Laboratory for Diagnosis and Treatment of Colorectal Cancer in Henan ProvinceZhengzhouChina
- Zhengzhou Key Laboratory for Diagnosis, Treatment and Research of Colorectal CancerZhengzhouChina
| |
Collapse
|
5
|
Li S, Fang W, Zheng J, Peng Z, Yu B, Chen C, Zhang Y, Jiang W, Yuan S, Zhang L, Zhang X. Whole-transcriptome defines novel glucose metabolic subtypes in colorectal cancer. J Cell Mol Med 2024; 28:e18065. [PMID: 38116696 PMCID: PMC10902307 DOI: 10.1111/jcmm.18065] [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: 06/13/2023] [Revised: 11/11/2023] [Accepted: 11/17/2023] [Indexed: 12/21/2023] Open
Abstract
Colorectal cancer (CRC) is the most prevalent malignancy of the digestive system. Glucose metabolism plays a crucial role in CRC development. However, the heterogeneity of glucose metabolic patterns in CRC is not well characterized. Here, we classified CRC into specific glucose metabolic subtypes and identified the key regulators. 2228 carbohydrate metabolism-related genes were screened out from the GeneCards database, 202 of them were identified as prognosis genes in the TCGA database. Based on the expression patterns of the 202 genes, three metabolic subtypes were obtained by the non-negative matrix factorization clustering method. The C1 subtype had the worst survival outcome and was characterized with higher immune cell infiltration and more activation in extracellular matrix pathways than the other two subtypes. The C2 subtype was the most prevalent in CRC and was characterized by low immune cell infiltration. The C3 subtype had the smallest number of individuals and had a better prognosis, with higher levels of NRF2 and TP53 pathway expression. Secreted frizzled-related protein 2 (SFRP2) and thrombospondin-2 (THBS2) were confirmed as biomarkers for the C1 subtype. Their expression levels were elevated in high glucose condition, while their knockdown inhibited migration and invasion of HCT 116 cells. The analysis of therapeutic potential found that the C1 subtype was more sensitive to immune and PI3K-Akt pathway inhibitors than the other subtypes. To sum up, this study revealed a novel glucose-related CRC subtype, characterized by SFRP2 and THBS2, with poor prognosis but possible therapeutic benefits from immune and targeted therapies.
Collapse
Affiliation(s)
- Shaohua Li
- The Third School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
- Department of General SurgerySouthern Medical University Affiliated Fengxian Central HospitalShanghaiChina
| | - Wei Fang
- The Third School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
- Department of General SurgerySouthern Medical University Affiliated Fengxian Central HospitalShanghaiChina
| | - Jianfeng Zheng
- The Third School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
- Department of General SurgerySouthern Medical University Affiliated Fengxian Central HospitalShanghaiChina
| | - Zhiqiang Peng
- State Key Laboratory of ProteomicsNational Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijingChina
| | - Biyue Yu
- School of Life SciencesHebei UniversityBaodingChina
| | - Chunhui Chen
- State Key Laboratory of ProteomicsNational Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijingChina
| | - Yuting Zhang
- School of Life SciencesHebei UniversityBaodingChina
| | - Wenli Jiang
- School of Life SciencesHebei UniversityBaodingChina
| | - Shuhui Yuan
- State Key Laboratory of ProteomicsNational Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijingChina
| | - Lingqiang Zhang
- State Key Laboratory of ProteomicsNational Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijingChina
| | - Xueli Zhang
- The Third School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
- Department of General SurgerySouthern Medical University Affiliated Fengxian Central HospitalShanghaiChina
| |
Collapse
|
6
|
Jin Y, Jiang A, Sun L, Lu Y. Long noncoding RNA TMPO-AS1 accelerates glycolysis by regulating the miR-1270/PKM2 axis in colorectal cancer. BMC Cancer 2024; 24:238. [PMID: 38383342 PMCID: PMC10880273 DOI: 10.1186/s12885-024-11964-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: 09/25/2023] [Accepted: 02/06/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Long noncoding RNA thymopoietin-antisense RNA 1 (TMPO-AS1) is recognized as a participant in cancer progression. Nevertheless, its biological function in colorectal cancer remains obscure and needs further elucidation. METHODS AND RESULTS First, we discovered enriched TMPO-AS1 in the tumor tissues that were related to poor prognosis. TMPO-AS1 knockdown enhanced SW480 cell apoptosis but inhibited invasion, proliferation, migration, and glucose metabolism. Further, MiR-1270 is directly bound with TMPO-AS1. MiR-1270 mimics were confirmed to inhibit cell proliferation, invasion, and glucose metabolism in our study. Mechanistically, miR-1270 directly is bound with the 3' untranslated regions (3'UTR) of PKM2 to downregulate PKM2. MiR-1270 inhibitors reversed the TMPO-AS1 knockdown's effect on suppressing the tumor cell proliferation, invasion, and glycolysis, while the knockdown of PKM2 further inverted the function of miR-1270 inhibitors on the TMPO-AS1 knockdown. CONCLUSIONS This study illustrated that TMPO-AS1 advanced the development and the glycolysis of colorectal cancer by modulating the miR-1270/PKM2 axis, which provided a new insight into the colorectal cancer therapeutic strategy.
Collapse
Affiliation(s)
- Yingmin Jin
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Str, Harbin, 150001, People's Republic of China.
| | - Aimin Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Str, Harbin, 150001, People's Republic of China
| | - Liying Sun
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Str, Harbin, 150001, People's Republic of China
| | - Yue Lu
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Str, Harbin, 150001, People's Republic of China
| |
Collapse
|
7
|
Xiao H, Wu GL, Tan S, Tan X, Yang Q. Recent Progress on Tumor Microenvironment-Activated NIR-II Phototheranostic Agents with Simultaneous Activation for Diagnosis and Treatment. Chem Asian J 2024; 19:e202301036. [PMID: 38230541 DOI: 10.1002/asia.202301036] [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: 11/23/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/18/2024]
Abstract
Malignant tumors seriously threaten human life and well-being. Emerging Near-infrared II (NIR-II, 1000-1700 nm) phototheranostic nanotechnology integrates diagnostic and treatment modalities, offering merits including improved tissue penetration and enhanced spatiotemporal resolution. This remarkable progress has opened promising avenues for advancing tumor theranostic research. The tumor microenvironment (TME) differs from normal tissues, exhibiting distinct attributes such as hypoxia, acidosis, overexpressed hydrogen peroxide, excess glutathione, and other factors. Capitalizing on these attributes, researchers have developed TME-activatable NIR-II phototheranostic agents with diagnostic and therapeutic attributes concurrently. Therefore, developing TME-activatable NIR-II phototheranostic agents with diagnostic and therapeutic activation holds significant research importance. Currently, research on TME-activatable NIR-II phototheranostic agents is still in its preliminary stages. This review examines the recent advances in developing dual-functional NIR-II activatable phototheranostic agents over the past years. It systematically presents NIR-II phototheranostic agents activated by various TME factors such as acidity (pH), hydrogen peroxide (H2 O2 ), glutathione (GSH), hydrogen sulfide (H2 S), enzymes, and their hybrid. This encompasses NIR-II fluorescence and photoacoustic imaging diagnostics, along with therapeutic modalities, including photothermal, photodynamic, chemodynamic, and gas therapies triggered by these TME factors. Lastly, the difficulties and opportunities confronting NIR-II activatable phototheranostic agents in the simultaneous diagnosis and treatment field are highlighted.
Collapse
Affiliation(s)
- Hao Xiao
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28, West Changsheng Road, Hengyang City, Hunan Province, 421001, China
- MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, 28, West Changsheng Road, Hengyang City, Hunan Province, 421001, China
| | - Gui-Long Wu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28, West Changsheng Road, Hengyang City, Hunan Province, 421001, China
- MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, 28, West Changsheng Road, Hengyang City, Hunan Province, 421001, China
| | - Senyou Tan
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28, West Changsheng Road, Hengyang City, Hunan Province, 421001, China
- MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, 28, West Changsheng Road, Hengyang City, Hunan Province, 421001, China
| | - Xiaofeng Tan
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28, West Changsheng Road, Hengyang City, Hunan Province, 421001, China
- MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, 28, West Changsheng Road, Hengyang City, Hunan Province, 421001, China
- National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, 53 Xiangchun Road, Changsha City, Hunan Province, 410008, China
| | - Qinglai Yang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28, West Changsheng Road, Hengyang City, Hunan Province, 421001, China
- MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, 28, West Changsheng Road, Hengyang City, Hunan Province, 421001, China
- National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, 53 Xiangchun Road, Changsha City, Hunan Province, 410008, China
| |
Collapse
|
8
|
Li J, Yang C, Zheng Y. A novel disulfidptosis and glycolysis related risk score signature for prediction of prognosis and ICI therapeutic responsiveness in colorectal cancer. Sci Rep 2023; 13:13344. [PMID: 37587262 PMCID: PMC10432503 DOI: 10.1038/s41598-023-40381-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023] Open
Abstract
Disulfidptosis is a newly-identified non-programmed cell death mode with tight associations with glucose metabolism. Elevated glycolysis is an important metabolic feature of tumor cells, which fulfills the energy requirement for their rapid growth and progression. Our present study determined to develop a disulfidptosis and glycolysis related gene (DGRG) risk score signature to predict the prognosis and ICI therapeutic responsiveness for CRC patients. First, the gene expression and clinical profiles for CRC patients were obtained from TCGA and GEO database. Using weighted gene co-expression network analysis, we identified hub genes showing the strongest correlations with both disulfidptosis and glycolysis activities. Next, a DGRG risk score signature was successfully developed through univariate and least absolute shrinkage and selection operator method Cox regression method. A DGRG risk score-based nomogram could further enhance the predictive performance. In addition, an array of systemic analysis was performed to unravel the correlation of DGRG risk score with tumor microenvironment. The results showed that CRC patients with low DGRG risk level had up-regulated immune cell infiltrations, enhanced metabolic activities and heightened gene mutation frequencies, while high risk patients was the opposite. Moreover, our present study identified low risk CRC patients as potential beneficiaries from immune checkpoint inhibitor (ICI) therapies. Our present work highlighted the potential utility of DGRG risk score signature in prognosis prediction and ICI responsiveness determination for CRC patients, which demonstrated promising clinical application value.
Collapse
Affiliation(s)
- Jiazheng Li
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chao Yang
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yongbin Zheng
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
9
|
Liu Y, Jiang C, Liu Q, Huang R, Wang M, Guo X. CircRNAs: emerging factors for regulating glucose metabolism in colorectal cancer. Clin Transl Oncol 2023:10.1007/s12094-023-03131-7. [PMID: 36944731 DOI: 10.1007/s12094-023-03131-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/19/2023] [Indexed: 03/23/2023]
Abstract
Colorectal cancer is a malignant disease with a high incidence and low survival rate, and the effectiveness of traditional treatments, such as surgery and radiotherapy, is very limited. CircRNAs, a kind of stable endogenous circular RNA, generally function by sponging miRNAs and binding or translating proteins. CircRNAs have been identified to play an important role in regulating the proliferation and metabolism of CRC. In recent years, many reports have indicated that by regulating the expression of glycolysis-related proteins, such as GLUT1 and HK2, or directly translating proteins, circRNAs can promote the Warburg effect in cancer cells, thereby driving CRC metabolism. Moreover, the Warburg effect increases lactate production in cancer cells and promotes acidification of the TME, which further drives cancer progression. In this review, we summarized the remarkable role of circRNAs in regulating glucose metabolism in CRC in recent years, which might be useful for finding new targets for the clinical treatment of CRC.
Collapse
Affiliation(s)
- Yulin Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, The People's Republic of China
| | - Chenjun Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, The People's Republic of China
| | - Qianqian Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, The People's Republic of China
| | - Runchun Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, The People's Republic of China
| | - Mancai Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, The People's Republic of China
- General Surgery Department, The Second Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Xiaohu Guo
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, The People's Republic of China.
- General Surgery Department, The Second Hospital of Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
10
|
Adamová B, Říhová K, Pokludová J, Beneš P, Šmarda J, Navrátilová J. Synergistic cytotoxicity of perifosine and ABT-737 to colon cancer cells. J Cell Mol Med 2022; 27:76-88. [PMID: 36523175 PMCID: PMC9806293 DOI: 10.1111/jcmm.17636] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
An acidic environment and hypoxia within the tumour are hallmarks of cancer that contribute to cell resistance to therapy. Deregulation of the PI3K/Akt pathway is common in colon cancer. Numerous Akt-targeted therapies are being developed, the activity of Akt-inhibitors is, however, strongly pH-dependent. Combination therapy thus represents an opportunity to increase their efficacy. In this study, the cytotoxicity of the Akt inhibitor perifosine and the Bcl-2/Bcl-xL inhibitor ABT-737 was tested in colon cancer HT-29 and HCT-116 cells cultured in monolayer or in the form of spheroids. The efficacy of single drugs and their combination was analysed in different tumour-specific environments including acidosis and hypoxia using a series of viability assays. Changes in protein content and distribution were determined by immunoblotting and a "peeling analysis" of immunohistochemical signals. While the cytotoxicity of single agents was influenced by the tumour-specific microenvironment, perifosine and ABT-737 in combination synergistically induced apoptosis in cells cultured in both 2D and 3D independently on pH and oxygen level. Thus, the combined therapy of perifosine and ABT-737 could be considered as a potential treatment strategy for colon cancer.
Collapse
Affiliation(s)
- Barbora Adamová
- Department of Experimental Biology, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Kamila Říhová
- Department of Experimental Biology, Faculty of ScienceMasaryk UniversityBrnoCzech Republic,International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
| | - Jana Pokludová
- Department of Experimental Biology, Faculty of ScienceMasaryk UniversityBrnoCzech Republic,International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
| | - Petr Beneš
- Department of Experimental Biology, Faculty of ScienceMasaryk UniversityBrnoCzech Republic,International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
| | - Jan Šmarda
- Department of Experimental Biology, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Jarmila Navrátilová
- Department of Experimental Biology, Faculty of ScienceMasaryk UniversityBrnoCzech Republic,International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
| |
Collapse
|
11
|
Fan S, Xing J, Jiang Z, Zhang Z, Zhang H, Wang D, Tang D. Effects of Long Non-Coding RNAs Induced by the Gut Microbiome on Regulating the Development of Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14235813. [PMID: 36497293 PMCID: PMC9735521 DOI: 10.3390/cancers14235813] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 11/29/2022] Open
Abstract
Although an imbalanced gut microbiome is closely associated with colorectal cancer (CRC), how the gut microbiome affects CRC is not known. Long non-coding RNAs (lncRNAs) can affect important cellular functions such as cell division, proliferation, and apoptosis. The abnormal expression of lncRNAs can promote CRC cell growth, proliferation, and metastasis, mediating the effects of the gut microbiome on CRC. Generally, the gut microbiome regulates the lncRNAs expression, which subsequently impacts the host transcriptome to change the expression of downstream target molecules, ultimately resulting in the development and progression of CRC. We focused on the important role of the microbiome in CRC and their effects on CRC-related lncRNAs. We also reviewed the impact of the two main pathogenic bacteria, Fusobacterium nucleatum and enterotoxigenic Bacteroides fragilis, and metabolites of the gut microbiome, butyrate, and lipopolysaccharide, on lncRNAs. Finally, available therapies that target the gut microbiome and lncRNAs to prevent and treat CRC were proposed.
Collapse
Affiliation(s)
- Shiying Fan
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Juan Xing
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Zhengting Jiang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Zhilin Zhang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Huan Zhang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People’s Hospital, Yangzhou University, Yangzhou 225000, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People’s Hospital, Yangzhou University, Yangzhou 225000, China
- Correspondence: ; Tel.: +86-18952783556
| |
Collapse
|
12
|
Shan J, Han D, Shen C, Lei Q, Zhang Y. Mechanism and strategies of immunotherapy resistance in colorectal cancer. Front Immunol 2022; 13:1016646. [PMID: 36238278 PMCID: PMC9550896 DOI: 10.3389/fimmu.2022.1016646] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/05/2022] [Indexed: 11/15/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world. Although there are standard treatment options for CRC, most patients respond poorly to these treatments. Immunotherapies have gradually emerged due to the increasing awareness and understanding of tumor immunity, exhibiting good therapeutic efficacy in various cancers. Immunotherapies include cytokines, immune checkpoint inhibitors (ICIs), and adoptive cell therapies. In particular, ICIs, which are antibodies against cytotoxic T lymphocyte-associated protein 4 (CTLA-4), programmed cell death 1 (PD-1), or its ligand PD-L1, have been successfully applied clinically for solid tumors, relieving the inhibitory effect of the tumor microenvironment on T cells. However, only a minority of patients with cancer achieve a durable clinical response during immunotherapy. Several factors restrict the efficacy of immunotherapy, leading to the development of drug resistance. In this review, we aimed to discuss the current status of immunotherapy for CRC and elaborate on the mechanisms that mediate resistance to immunotherapy and other potential therapeutic strategies.
Collapse
Affiliation(s)
- Jiqi Shan
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dong Han
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunyi Shen
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingyang Lei
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
- *Correspondence: Yi Zhang,
| |
Collapse
|
13
|
Lin W, Lu X, Yang H, Huang L, Huang W, Tang Y, Liu S, Wang H, Zhang Y. Metabolic heterogeneity protects metastatic mucosal melanomas cells from ferroptosis. Int J Mol Med 2022; 50:124. [PMID: 36004461 PMCID: PMC9448297 DOI: 10.3892/ijmm.2022.5180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/18/2022] [Indexed: 11/08/2022] Open
Abstract
Cancer heterogeneity has been proposed to be one of the main causes of metastatic dissemination and therapy failure. However, the underlying mechanisms of this phenomenon remain poorly understood. Melanoma is an aggressive malignancy with a high heterogeneity and metastatic potential. Therefore, the present study investigated the possible association between cancer heterogeneity and metastasis in melanoma. In total, two novel Chinese oral mucosal melanoma (COMM) cell lines, namely COMM-1 and COMM-2, were established for exploring methods into preventing the loss of cellular heterogeneity caused by long-term cell culture. Each cell line was grown under two different models of culture, which yielded two subtypes, one exhibited an adhesive morphology (COMM-AD), whereas the other was grown in suspension (COMM-SUS). Compared with the COMM-AD cells, the COMM-SUS cells exhibited higher metastatic capacities and autofluorescence. Further investigations indicated that the COMM-SUS cells exhibited metabolic reprogramming by taking up lactate produced by COMM-AD cells at increased levels to accumulate NADH through monocarboxylate transporter 1, whilst also increasing NADPH levels through the pentose phosphate pathway (PPP). Additionally, increased NADH and NADPH levels in the COMM-SUS cells, coupled with the upregulation of the anti-ferroptotic proteins, glutathione peroxidase 4 and ferroptosis suppressor protein 1, enabled them to resist ferroptotic cell death induced by oxidative stress during hematogenous dissemination. The inhibition of ferroptosis was found to substantially increase the metastatic capacity of COMM-AD cells. Furthermore, suppressing lactate uptake and impairing PPP activation significantly decreased the metastatic potential of the COMM-SUS cells. Thus, the present study on metabolic heterogeneity in COMM cells potentially provides a novel perspective for exploring this mechanism underlying cancer metastasis.
Collapse
Affiliation(s)
- Weifan Lin
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat‑sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Xiangwan Lu
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat‑sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Hang Yang
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat‑sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Linxuan Huang
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat‑sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Wuheng Huang
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat‑sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Yuluan Tang
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat‑sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Situn Liu
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat‑sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Hua Wang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guanghua Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Yan Zhang
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat‑sen University, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|
14
|
Wang H, Wang Q, Dong J, Jiang W, Kong L, Zhang Q, Liu H. New perspective of ceria nanodots for precise tumor therapy via oxidative stress pathway. Heliyon 2022; 8:e10370. [PMID: 36061010 PMCID: PMC9429517 DOI: 10.1016/j.heliyon.2022.e10370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/29/2022] [Accepted: 08/15/2022] [Indexed: 11/26/2022] Open
Abstract
Ceria-based nanomaterials have aroused major attentions among the biomedical application research field in recent years. Most of the researches have mainly focused on promoting the functional healing therapies of normal cells/organs with cerium oxide compounds, while the applications of ceria-based materials employed on cancer curing processes have been merely mentioned. To explore the possible capabilities of cerium oxide nanomaterials exterminating tumor cells, innovatively, we synthesized the eco-friendly pure cerium oxide nanodots (CNDs), proving the prominent ability of CNDs used in tumor chemotherapy (CDT) via Fenton reaction with the highly presence of H2O2 (acidic pH) in tumor tissues. CNDs reacted with the self-produced H2O2 of tumor cells, which generated piled up toxic hydroxyl radical (·OH). The accumulated virulent ·OH restrained the growth of cancer cells intensively. This peroxidase-like activity, provided a distinguished paradigm for effective cancer curing treatment. We also verified the biosafety of CNDs applied on normal cells. Notably, not only did CNDs be harmless to normal cells, but also it protected them from the damages of reactive oxygen species (ROS). In normal cells/tissues, under the microenvironment of neutral pH and low level of H2O2, the CNDs could effectively function as an annihilator inhibiting ROS. They reduced the damages caused by ROS, exhibiting catalase-like activity. The research we studied, which estimated CNDs thoroughly, has provided a new perspective to the future researches of the cerium oxide biomaterial applications.
Collapse
Affiliation(s)
- Hui Wang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Qi Wang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Jianyue Dong
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Weiwei Jiang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Linghong Kong
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Qiong Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Hanping Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
15
|
The Antitumor Effect of Cinnamaldehyde Derivative CB-PIC in Hepatocellular Carcinoma Cells via Inhibition of Pyruvate and STAT3 Signaling. Int J Mol Sci 2022; 23:ijms23126461. [PMID: 35742904 PMCID: PMC9223629 DOI: 10.3390/ijms23126461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
Though cinnamaldehyde derivative (CB-PIC), a major compound of cinnamon, is known to have anticancer activity, its underlying mechanism is not fully understood. In the present study, the anticancer mechanism of CB-PIC was investigated in human hepatocellular carcinoma cells (HCCs) in association with signal transducer and activator of transcription 3 (STAT3) signaling. CB-PIC exerted cytotoxicity in HepG2 and Huh7 cells. CB-PIC increased the sub G1 population and attenuated the expression of pro-poly (ADP-ribose) polymerase (PARP) and pro-Caspase3 in HepG2 and Huh7 cells. Interestingly, CB-PIC significantly abrogated the expression of a glycolytic enzyme pyruvate kinase M2 (PKM2) in HepG2 cells more than in LNCaP, A549, and HCT-116 cells. Consistently, CB-PIC reduced the expression of hexokinase 2 (HK2) and PKM2, along with a reduced production of lactate in HepG2 and Huh7 cells. Notably, CB-PIC suppressed the phosphorylation of STAT3 in HepG2 and Huh7 cells and conversely STAT3 depletion enhanced the capacity of CB-PIC to suppress the expression of HK2, PKM2, and pro-caspase3 and to reduce the viability in Huh7 cells. Furthermore, CB-PIC activated the phosphorylation of AMPK and ERK and suppressed expression of IL-6 as STAT3-related genes in HepG2 and Huh7 cells. Conversely, pyruvate treatment reversed the inhibitory effect of CB-PIC on p-STAT3, HK2, PKM2, and pro-PARP in Huh7 cells. Overall, there findings suggest that CB-PIC exerts an apoptotic effect via inhibition of the Warburg effect mediated by p-STAT3 and pyruvate signaling.
Collapse
|
16
|
Liu Z, Liu Z, Zhou X, Lu Y, Yao Y, Wang W, Lu S, Wang B, Li F, Fu W. A glycolysis-related two-gene risk model that can effectively predict the prognosis of patients with rectal cancer. Hum Genomics 2022; 16:5. [PMID: 35109912 PMCID: PMC8812245 DOI: 10.1186/s40246-022-00377-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Background Aerobic glycolysis is an emerging hallmark of cancer. Although some studies have constructed glycolysis-related prognostic models of colon adenocarcinoma (COAD) based on The Cancer Genome Atlas (TCGA) database, whether the COAD glycolysis-related prognostic model is appropriate for distinguishing the prognosis of rectal adenocarcinoma (READ) patients remains unknown. Exploring critical and specific glycolytic genes related to READ prognosis may help us discover new potential therapeutic targets for READ patients. Results Three gene sets, HALLMARK_GLYCOLYSIS, REACTOME_GLYCOLYSIS and REACTOME_REGULATION_OF_GLYCOLYSIS_BY_FRUCTOSE_2_6_BISPHOSPHATE_METABOLISM, were both significantly enriched in both COAD and READ through glycolysis-related gene set enrichment analysis (GSEA). We found that six genes (ANKZF1, STC2, SUCLG2P2, P4HA1, GPC1 and PCK1) were independent prognostic genes in COAD, while TSTA3 and PKP2 were independent prognostic genes in READ. Glycolysis-related prognostic model of COAD and READ was, respectively, constructed and assessed in COAD and READ. We found that the glycolysis-related prognostic model of COAD was not appropriate for READ, while glycolysis-related prognostic model of READ was more appropriate for READ than for COAD. PCA and t-SNE analysis confirmed that READ patients in two groups (high and low risk score groups) were distributed in discrete directions based on the glycolysis-related prognostic model of READ. We found that this model was an independent prognostic indicator through multivariate Cox analysis, and it still showed robust effectiveness in different age, gender, M stage, and TNM stage. A nomogram combining the risk model of READ with clinicopathological characteristics was established to provide oncologists with a practical tool to evaluate the rectal cancer outcomes. GO enrichment and KEGG analyses confirmed that differentially expressed genes (DEGs) were enriched in several glycolysis-related molecular functions or pathways based on glycolysis-related prognostic model of READ. Conclusions We found that a glycolysis-related prognostic model of COAD was not appropriate for READ, and we established a novel glycolysis-related two-gene risk model to effectively predict the prognosis of rectal cancer patients.
Supplementary Information The online version contains supplementary material available at 10.1186/s40246-022-00377-0.
Collapse
Affiliation(s)
- Zhenzhen Liu
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, People's Republic of China
| | - Zhentao Liu
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, People's Republic of China
| | - Xin Zhou
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, People's Republic of China
| | - Yongqu Lu
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, People's Republic of China
| | - Yanhong Yao
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, People's Republic of China
| | - Wendong Wang
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, People's Republic of China
| | - Siyi Lu
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, People's Republic of China
| | - Bingyan Wang
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, People's Republic of China
| | - Fei Li
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, People's Republic of China
| | - Wei Fu
- Department of General Surgery, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing, People's Republic of China.
| |
Collapse
|
17
|
Zhu Y, Liu Z, Lv D, Cheng X, Wang J, Liu B, Han Z, Wang Y, Liu R, Gao Y. Identification of PYGL as a key prognostic gene of glioma by integrated bioinformatics analysis. Future Oncol 2022; 18:579-596. [PMID: 35037470 DOI: 10.2217/fon-2021-0759] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: PYGL has been reported to have carcinogenic effects in a variety of tumors. This study is the first to reveal the relationship between PYGL and the prognosis of glioma. Materials & methods: Analyzing the Chinese Glioma Genome Atlas database, the authors revealed the expression status and prognostic value of PYGL in gliomas and used RT-qPCR to verify PYGL expression again. Subsequently, they used Gene Set Enrichment Analysis to explore the biological pathways that PYGL may participate in. The authors also used the tumor immune estimation resource database to explore the relationship between PYGL and tumor immune cells. Results: PYGL is involved in the malignant progression of glioma. Conclusions: PYGL can be used as a new biomarker and molecular target for evaluating the prognosis and immunotherapy of glioma.
Collapse
Affiliation(s)
- Yongjie Zhu
- Henan University People's Hospital, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450003, China
| | - Zhendong Liu
- Department Of Orthopaedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, China
| | - Dongbo Lv
- Department Of Orthopaedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, China
| | - Xingbo Cheng
- Department of Neurosurgery of the First affiliate Hospital of Harbin Medical University, Harbin 150000, China
| | - Jialin Wang
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, No. 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450003, China
| | - Binfeng Liu
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, No. 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450003, China
| | - Zhibin Han
- Department of Neurosurgery of the First affiliate Hospital of Harbin Medical University, Harbin 150000, China
| | - Yanbiao Wang
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, No. 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450003, China
| | - Runze Liu
- Henan University People's Hospital, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450003, China
| | - Yanzheng Gao
- Department of Surgery of Spine & Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation & Transformation, Henan Key Laboratory for intelligent precision orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Henan, Zhengzhou 450003, China
| |
Collapse
|
18
|
Sun Q, Wu J, Zhu G, Li T, Zhu X, Ni B, Xu B, Ma X, Li J. Lactate-related metabolic reprogramming and immune regulation in colorectal cancer. Front Endocrinol (Lausanne) 2022; 13:1089918. [PMID: 36778600 PMCID: PMC9909490 DOI: 10.3389/fendo.2022.1089918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/27/2022] [Indexed: 01/27/2023] Open
Abstract
Changes in cellular metabolism involving fuel sources are well-known mechanisms of cancer cell differentiation in the context of carcinogenesis. Metabolic reprogramming is regulated by oncogenic signaling and transcriptional networks and has been identified as an essential component of malignant transformation. Hypoxic and acidified tumor microenvironment contributes mainly to the production of glycolytic products known as lactate. Mounting evidence suggests that lactate in the tumor microenvironment of colorectal cancer(CRC) contributes to cancer therapeutic resistance and metastasis. The contents related to the regulatory effects of lactate on metabolism, immune response, and intercellular communication in the tumor microenvironment of CRC are also constantly updated. Here we summarize the latest studies about the pleiotropic effects of lactate in CRC and the clinical value of targeting lactate metabolism as treatment. Different effects of lactate on various immune cell types, microenvironment characteristics, and pathophysiological processes have also emerged. Potential specific therapeutic targeting of CRC lactate metabolism is also discussed. With increased knowledge, effective druggable targets might be identified, with the aim of improving treatment outcomes by reducing chemoresistance.
Collapse
Affiliation(s)
- Qianhui Sun
- Oncology Department, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingyuan Wu
- Oncology Department, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Guanghui Zhu
- Oncology Department, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Tingting Li
- Graduate College, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Xiaoyu Zhu
- Oncology Department, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baoyi Ni
- Oncology Department, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bowen Xu
- Oncology Department, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Xinyi Ma
- Oncology Department, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Li
- Oncology Department, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jie Li,
| |
Collapse
|
19
|
Hong J, Guo F, Lu SY, Shen C, Ma D, Zhang X, Xie Y, Yan T, Yu T, Sun T, Qian Y, Zhong M, Chen J, Peng Y, Wang C, Zhou X, Liu J, Liu Q, Ma X, Chen YX, Chen H, Fang JY. F. nucleatum targets lncRNA ENO1-IT1 to promote glycolysis and oncogenesis in colorectal cancer. Gut 2021; 70:2123-2137. [PMID: 33318144 DOI: 10.1136/gutjnl-2020-322780] [Citation(s) in RCA: 141] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/22/2020] [Accepted: 11/29/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Microbiota disorder promotes chronic inflammation and carcinogenesis. High glycolysis is associated with poor prognosis in patients with colorectal cancer (CRC). However, the potential correlation between the gut microbiota and glucose metabolism is unknown in CRC. DESIGN 18F-FDG (18F-fluorodeoxyglucose) PET (positron emission tomography)/CT image scanning data and microbiota PCR analysis were performed to measure the correlation between metabolic alterations and microbiota disorder in 33 patients with CRC. Multiple colorectal cancer models, metabolic analysis and Seahorse assay were established to assess the role of long non-coding RNA (lncRNA) enolase1-intronic transcript 1 (ENO1-IT1) in Fusobacterium (F.) nucleatum-induced glucose metabolism and colorectal carcinogenesis. RNA immunoprecipitation and chromatin immunoprecipitation sequencing were conducted to identify potential targets of lncRNA ENO1-IT1. RESULTS We have found F. nucleatum abundance correlated with high glucose metabolism in patients with CRC. Furthermore, F. nucleatum supported carcinogenesis via increasing CRC cell glucose metabolism. Mechanistically, F. nucleatum activated lncRNA ENO1-IT1 transcription via upregulating the binding efficiency of transcription factor SP1 to the promoter region of lncRNA ENO1-IT1. Elevated ENO1-IT behaved as a guider modular for KAT7 histone acetyltransferase, specifying the histone modification pattern on its target genes, including ENO1, and consequently altering CRC biological function. CONCLUSION F. nucleatum and glucose metabolism are mechanistically, biologically and clinically connected to CRC. Targeting ENO1 pathway may be meaningful in treating patients with CRC with elevated F. nucleatum.
Collapse
Affiliation(s)
- Jie Hong
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fangfang Guo
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shi-Yuan Lu
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chaoqin Shen
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dan Ma
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyu Zhang
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yile Xie
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tingting Yan
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - TaChung Yu
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tiantian Sun
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Qian
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Zhong
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinxian Chen
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanshen Peng
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng Wang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiang Zhou
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Liu
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiong Ma
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Xuan Chen
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haoyan Chen
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing-Yuan Fang
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
20
|
Huang P, Zhu S, Liang X, Zhang Q, Luo X, Liu C, Song L. Regulatory Mechanisms of LncRNAs in Cancer Glycolysis: Facts and Perspectives. Cancer Manag Res 2021; 13:5317-5336. [PMID: 34262341 PMCID: PMC8275123 DOI: 10.2147/cmar.s314502] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/19/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer cells exhibit distinct metabolic characteristics that employ glycolysis to provide energy and intermediary metabolites. This aberrant metabolic phenotype favors cancer progression. LncRNAs are transcripts longer than 200 nucleotides that do not encode proteins. LncRNAs contribute to cancer progression and therapeutic resistance and affect aerobic glycolysis via multiple mechanisms, including modulating glycolytic transporters and enzymes. Further, dysregulated signaling pathways are vital for glycolysis. In this review, we highlight regulatory mechanisms for lncRNAs in aerobic glycolysis that provide novel insights into cancer development. Moreover, a comprehensive understanding of the regulatory mechanisms of lncRNAs in aerobic glycolysis can provide new strategies for clinical cancer management.
Collapse
Affiliation(s)
- Peng Huang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| | - Shaomi Zhu
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| | - Xin Liang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| | - Qinxiu Zhang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| | - Xiaohong Luo
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| | - Chi Liu
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| | - Linjiang Song
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| |
Collapse
|
21
|
Prevention of High Glucose-Mediated EMT by Inhibition of Hsp70 Chaperone. Int J Mol Sci 2021; 22:ijms22136902. [PMID: 34199046 PMCID: PMC8268552 DOI: 10.3390/ijms22136902] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/11/2021] [Accepted: 06/24/2021] [Indexed: 12/29/2022] Open
Abstract
Hyperglycemia may contribute to the progression of carcinomas by triggering epithelial-to-mesenchymal transition (EMT). Some proteostasis systems are involved in metastasis; in this paper, we sought to explore the mechanism of Hsp70 chaperone in EMT. We showed that knockdown of Hsp70 reduced cell migration capacity concomitantly with levels of mRNA of the Slug, Snail, and Twist markers of EMT, in colon cancer cells incubated in high glucose medium. Conversely, treatment of cells with Hsp70 inducer U-133 were found to elevate cell motility, along with the other EMT markers. To prove that inhibiting Hsp70 may reduce EMT efficiency, we treated cells with a CL-43 inhibitor of the HSF1 transcription factor, which lowered Hsp70 and HSF1 content in the control and induced EMT in carcinoma cells. Importantly, CL-43 reduced migration capacity, EMT-linked transcription factors, and increased content of epithelial marker E-cadherin in colon cancer cells of three lines, including one derived from a clinical sample. To prove that Hsp70 chaperone should be targeted when inhibiting the EMT pathway, we treated cancer cells with 2-phenylethynesulfonamide (PES) and demonstrated that the compound inhibited substrate-binding capacity of Hsp70. Furthermore, PES suppressed EMT features, cell motility, and expression of specific transcription factors. In conclusion, the Hsp70 chaperone machine efficiently protects mechanisms of the EMT, and the safe inhibitors of the chaperone are needed to hamper metastasis at its initial stage.
Collapse
|
22
|
Iacobini C, Vitale M, Pesce C, Pugliese G, Menini S. Diabetic Complications and Oxidative Stress: A 20-Year Voyage Back in Time and Back to the Future. Antioxidants (Basel) 2021; 10:727. [PMID: 34063078 PMCID: PMC8147954 DOI: 10.3390/antiox10050727] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
Twenty years have passed since Brownlee and colleagues proposed a single unifying mechanism for diabetic complications, introducing a turning point in this field of research. For the first time, reactive oxygen species (ROS) were identified as the causal link between hyperglycemia and four seemingly independent pathways that are involved in the pathogenesis of diabetes-associated vascular disease. Before and after this milestone in diabetes research, hundreds of articles describe a role for ROS, but the failure of clinical trials to demonstrate antioxidant benefits and some recent experimental studies showing that ROS are dispensable for the pathogenesis of diabetic complications call for time to reflect. This twenty-year journey focuses on the most relevant literature regarding the main sources of ROS generation in diabetes and their role in the pathogenesis of cell dysfunction and diabetic complications. To identify future research directions, this review discusses the evidence in favor and against oxidative stress as an initial event in the cellular biochemical abnormalities induced by hyperglycemia. It also explores possible alternative mechanisms, including carbonyl stress and the Warburg effect, linking glucose and lipid excess, mitochondrial dysfunction, and the activation of alternative pathways of glucose metabolism leading to vascular cell injury and inflammation.
Collapse
Affiliation(s)
- Carla Iacobini
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (S.M.)
| | - Martina Vitale
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (S.M.)
| | - Carlo Pesce
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetic and Maternal Infantile Sciences (DINOGMI), Department of Excellence of MIUR, University of Genoa Medical School, 16132 Genoa, Italy;
| | - Giuseppe Pugliese
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (S.M.)
| | - Stefano Menini
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (S.M.)
| |
Collapse
|
23
|
Marcucci F, Rumio C. Glycolysis-induced drug resistance in tumors-A response to danger signals? Neoplasia 2021; 23:234-245. [PMID: 33418276 PMCID: PMC7804361 DOI: 10.1016/j.neo.2020.12.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022] Open
Abstract
Tumor cells often switch from mitochondrial oxidative metabolism to glycolytic metabolism even under aerobic conditions. Tumor cell glycolysis is accompanied by several nonenzymatic activities among which induction of drug resistance has important therapeutic implications. In this article, we review the main aspects of glycolysis-induced drug resistance. We discuss the classes of antitumor drugs that are affected and the components of the glycolytic pathway (transporters, enzymes, metabolites) that are involved in the induction of drug resistance. Glycolysis-associated drug resistance occurs in response to stimuli, either cell-autonomous (e.g., oncoproteins) or deriving from the tumor microenvironment (e.g., hypoxia or pseudohypoxia, mechanical cues, etc.). Several mechanisms mediate the induction of drug resistance in response to glycolytic metabolism: inhibition of apoptosis, induction of epithelial-mesenchymal transition, induction of autophagy, inhibition of drug influx and increase of drug efflux. We suggest that drug resistance in response to glycolysis comes into play in presence of qualitative (e.g., expression of embryonic enzyme isoforms, post-translational enzyme modifications) or quantitative (e.g., overexpression of enzymes or overproduction of metabolites) alterations of glycolytic metabolism. We also discern similarities between changes occurring in tumor cells in response to stimuli inducing glycolysis-associated drug resistance and those occurring in cells of the innate immune system in response to danger signals and that have been referred to as danger-associated metabolic modifications. Eventually, we briefly address that also mitochondrial oxidative metabolism may induce drug resistance and discuss the therapeutic implications deriving from the fact that the main energy-generating metabolic pathways may be both at the origin of antitumor drug resistance.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| |
Collapse
|
24
|
Zhang T, Jin Y, Jiang X, Li L, Qi X, Mao Y, Hua D. Clinical and Prognostic Relevance of B7-H3 and Indicators of Glucose Metabolism in Colorectal Cancer. Front Oncol 2020; 10:546110. [PMID: 33042836 PMCID: PMC7523031 DOI: 10.3389/fonc.2020.546110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022] Open
Abstract
Objective This study aimed to investigate the clinical and prognostic relevance of B7-H3 expression and indicators of glucose metabolism in patients with colorectal cancer (CRC). Methods Using immunohistochemistry, the expression of B7-H3 was detected in a total of 213 formalin-fixed paraffin-embedded CRC tissue specimens. Furthermore, levels of fasting blood glucose (FBG), lactic dehydrogenase (LDH), and fructosamine (FMN) as indicators of glucose metabolism were analyzed in CRC patients and stratified into high or low expression sub-groups based on Youden Index. The relationship between B7-H3, FBG, LDH, FMN expression, and clinicopathological characteristics were also evaluated to establish their prognostic significance in patients with CRC. Results B7-H3 was highly expressed in CRC tissue. The positive rates of B7-H3 expression was 63.8% (136/213). We found a linear correlation between B7-H3 and FBG in depth of tumor invasion (T3/4) (p = 0.037, r = 0.259), lymph node metastasis (N0) (p = 0.004, r = 0.259), and TNM stage (I/II) (p = 0.009, r = 0.242). High expression of FBG, LDH, FMN [hazard ratio (HR) = 1.916, 95% CI: 1.223–3.00, p = 0.005; HR = 1.801, 95% CI: 1.153–2.813, p = 0.010; HR = 2.154, 95% CI: 1.336–3.472, p = 0.002], respectively, was identified as a significant independent predictor of poor overall survival (OS). Although B7-H3 expression did not affect OS, CRC patients expressing both high B7-H3 and high FMN contributed to a significant decrease in OS (HR = 1.881, 95%CI: 1.059–3.339, p = 0.031). Moreover, with low expression of B7-H3, high expression of FBG, LDH and FMN were also recognized as predictors of inferior OS (HR = 3.393, 95% CI: 1.493-7.709, p = 0.004; HR = 7.107, 95% CI: 2.785–18.138, p = 0.000; HR = 2.800, 95% CI: 1.184–6.625, p = 0.019). Conclusion B7-H3 combined with FBG, LDH, or FMN, could reflect the clinical outcomes of patients with CRC.
Collapse
Affiliation(s)
- Ting Zhang
- Institue of Cancer, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yufen Jin
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xin Jiang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Longhai Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiaowei Qi
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.,Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yong Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.,Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Dong Hua
- Institue of Cancer, Affiliated Hospital of Jiangnan University, Wuxi, China.,Wuxi School of Medicine, Jiangnan University, Wuxi, China.,Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
25
|
Wang S, Zhang L, Yu Z, Chai K, Chen J. Identification of a Glucose Metabolism-related Signature for prediction of Clinical Prognosis in Clear Cell Renal Cell Carcinoma. J Cancer 2020; 11:4996-5006. [PMID: 32742447 PMCID: PMC7378912 DOI: 10.7150/jca.45296] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Clear cell renal cell carcinoma (ccRCC) is one of the most prevalent and invasive histological subtypes among all renal cell carcinomas (RCC). Cancer cell metabolism, particularly glucose metabolism, has been reported as a hallmark of cancer. However, the characteristics of glucose metabolism-related gene sets in ccRCC have not been systematically profiled. Methods: In this study, we downloaded a gene expression profile and glucose metabolism-related gene set from TCGA (The Cancer Genome Altas) and MSigDB, respectively, to analyze the characteristics of glucose metabolism-related gene sets in ccRCC. We used a multivariable Cox regression analysis to develop a risk signature, which divided patients into low- and high- risk groups. In addition, a nomogram that combined the risk signature and clinical characteristics was created for predicting the 3- and 5-year overall survival (OS) of ccRCC. The accuracy of the nomogram prediction was evaluated using the area under the receiver operating characteristic curve (AUC) and a calibration plot. Results: A total of 231 glucose metabolism-related genes were found, and 68 differentially expressed genes (DEGs) were identified. After screening by univariate regression analysis, LASSO regression analysis and multivariable Cox regression analysis, six glucose metabolism-related DEGs (FBP1, GYG2, KAT2A, LGALS1, PFKP, and RGN) were selected to develop a risk signature. There were significant differences in the clinical features (Fuhrman nuclear grade and TNM stage) between the high- and low-risk groups. The multivariable Cox regression indicated that the risk score was independent of the prognostic factors (training set: HR=3.393, 95% CI [2.025, 5.685], p<0.001; validation set: HR=1.933, 95% CI [1.130, 3.308], p=0.016). The AUCs of the nomograms for the 3-year OS in the training and validation sets were 0.808 and 0.819, respectively, and 0.777 and 0.796, respectively, for the 5- year OS. Conclusion: We demonstrated a novel glucose metabolism-related risk signature for predicting the prognosis of ccRCC. However, additional in vitro and in vivo research is required to validate our findings.
Collapse
Affiliation(s)
- Sheng Wang
- The Second Clinical Medical College, Zhejiang Chinese Medicine University, Hangzhou, Zhejiang.,Department of Oncology, Tongde Hospital of Zhejiang, Hangzhou, Zhejiang 310053, P.R. China
| | - Ling Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medicine University, Hangzhou, Zhejiang.,Department of Oncology, Tongde Hospital of Zhejiang, Hangzhou, Zhejiang 310053, P.R. China
| | - Zhihong Yu
- Department of Oncology, Tongde Hospital of Zhejiang, Hangzhou, Zhejiang 310053, P.R. China
| | - Kequn Chai
- Department of Oncology, Tongde Hospital of Zhejiang, Hangzhou, Zhejiang 310053, P.R. China
| | - Jiabin Chen
- Department of Oncology, Tongde Hospital of Zhejiang, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
26
|
Gupta K, Vuckovic I, Zhang S, Xiong Y, Carlson BL, Jacobs J, Olson I, Petterson XM, Macura SI, Sarkaria J, Burns TC. Radiation Induced Metabolic Alterations Associate With Tumor Aggressiveness and Poor Outcome in Glioblastoma. Front Oncol 2020; 10:535. [PMID: 32432031 PMCID: PMC7214818 DOI: 10.3389/fonc.2020.00535] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/25/2020] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma (GBM) is uniformly fatal with a 1-year median survival, despite best available treatment, including radiotherapy (RT). Impacts of prior RT on tumor recurrence are poorly understood but may increase tumor aggressiveness. Metabolic changes have been investigated in radiation-induced brain injury; however, the tumor-promoting effect following prior radiation is lacking. Since RT is vital to GBM management, we quantified tumor-promoting effects of prior RT on patient-derived intracranial GBM xenografts and characterized metabolic alterations associated with the protumorigenic microenvironment. Human xenografts (GBM143) were implanted into nude mice 24 hrs following 20 Gy cranial radiation vs. sham animals. Tumors in pre-radiated mice were more proliferative and more infiltrative, yielding faster mortality (p < 0.0001). Histologic evaluation of tumor associated macrophage/microglia (TAMs) revealed cells with a more fully activated ameboid morphology in pre-radiated animals. Microdialyzates from radiated brain at the margin of tumor infiltration contralateral to the site of implantation were analyzed by unsupervised liquid chromatography-mass spectrometry (LC-MS). In pre-radiated animals, metabolites known to be associated with tumor progression (i.e., modified nucleotides and polyols) were identified. Whole-tissue metabolomic analysis of pre-radiated brain microenvironment for metabolic alterations in a separate cohort of nude mice using 1H-NMR revealed a significant decrease in levels of antioxidants (glutathione (GSH) and ascorbate (ASC)), NAD+, Tricarboxylic acid cycle (TCA) intermediates, and rise in energy carriers (ATP, GTP). GSH and ASC showed highest Variable Importance on Projection prediction (VIPpred) (1.65) in Orthogonal Partial least square Discriminant Analysis (OPLS-DA); Ascorbate catabolism was identified by GC-MS. To assess longevity of radiation effects, we compared survival with implantation occurring 2 months vs. 24 hrs following radiation, finding worse survival in animals implanted at 2 months. These radiation-induced alterations are consistent with a chronic disease-like microenvironment characterized by reduced levels of antioxidants and NAD+, and elevated extracellular ATP and GTP serving as chemoattractants, promoting cell motility and vesicular secretion with decreased levels of GSH and ASC exacerbating oxidative stress. Taken together, these data suggest IR induces tumor-permissive changes in the microenvironment with metabolomic alterations that may facilitate tumor aggressiveness with important implications for recurrent glioblastoma. Harnessing these metabolomic insights may provide opportunities to attenuate RT-associated aggressiveness of recurrent GBM.
Collapse
Affiliation(s)
- Kshama Gupta
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Ivan Vuckovic
- Metabolomics Core Mayo Clinic, Rochester, MN, United States
| | - Song Zhang
- Metabolomics Core Mayo Clinic, Rochester, MN, United States
| | - Yuning Xiong
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Brett L Carlson
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States
| | - Joshua Jacobs
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Ian Olson
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | | | - Slobodan I Macura
- Metabolomics Core Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Jann Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States
| | - Terry C Burns
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
27
|
Sisakht M, Darabian M, Mahmoodzadeh A, Bazi A, Shafiee SM, Mokarram P, Khoshdel Z. The role of radiation induced oxidative stress as a regulator of radio-adaptive responses. Int J Radiat Biol 2020; 96:561-576. [PMID: 31976798 DOI: 10.1080/09553002.2020.1721597] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Purpose: Various sources of radiation including radiofrequency, electromagnetic radiation (EMR), low- dose X-radiation, low-level microwave radiation and ionizing radiation (IR) are indispensable parts of modern life. In the current review, we discussed the adaptive responses of biological systems to radiation with a focus on the impacts of radiation-induced oxidative stress (RIOS) and its molecular downstream signaling pathways.Materials and methods: A comprehensive search was conducted in Web of Sciences, PubMed, Scopus, Google Scholar, Embase, and Cochrane Library. Keywords included Mesh terms of "radiation," "electromagnetic radiation," "adaptive immunity," "oxidative stress," and "immune checkpoints." Manuscripts published up until December 2019 were included.Results: RIOS induces various molecular adaptors connected with adaptive responses in radiation exposed cells. One of these adaptors includes p53 which promotes various cellular signaling pathways. RIOS also activates the intrinsic apoptotic pathway by depolarization of the mitochondrial membrane potential and activating the caspase apoptotic cascade. RIOS is also involved in radiation-induced proliferative responses through interaction with mitogen-activated protein kinases (MAPks) including p38 MAPK, ERK, and c-Jun N-terminal kinase (JNK). Protein kinase B (Akt)/phosphoinositide 3-kinase (PI3K) signaling pathway has also been reported to be involved in RIOS-induced proliferative responses. Furthermore, RIOS promotes genetic instability by introducing DNA structural and epigenetic alterations, as well as attenuating DNA repair mechanisms. Inflammatory transcription factors including macrophage migration inhibitory factor (MIF), nuclear factor κB (NF-κB), and signal transducer and activator of transcription-3 (STAT-3) paly major role in RIOS-induced inflammation.Conclusion: In conclusion, RIOS considerably contributes to radiation induced adaptive responses. Other possible molecular adaptors modulating RIOS-induced responses are yet to be divulged in future studies.
Collapse
Affiliation(s)
- Mohsen Sisakht
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Darabian
- Department of Radiology, Faculty of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Mahmoodzadeh
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Bazi
- Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Sayed Mohammad Shafiee
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooneh Mokarram
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Khoshdel
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
28
|
Wang G, Wang YZ, Yu Y, Wang JJ, Yin PH, Xu K. Triterpenoids Extracted fromRhus chinensis MillAct Against Colorectal Cancer by Inhibiting Enzymes in Glycolysis and Glutaminolysis: Network Analysis and Experimental Validation. Nutr Cancer 2019; 72:293-319. [PMID: 31267795 DOI: 10.1080/01635581.2019.1631858] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Yu-Zhu Wang
- Department of Medicine, Jiangsu University, Zhenjiang City, China
| | - Yang Yu
- Department of Medicine, Jiangsu University, Zhenjiang City, China
| | - Jun-Jie Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Pei-Hao Yin
- Central laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ke Xu
- Central laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
29
|
Vyas M, Patel N, Celli R, Wajapeyee N, Jain D, Zhang X. Glucose Metabolic Reprogramming and Cell Proliferation Arrest in Colorectal Micropapillary Carcinoma. Gastroenterology Res 2019; 12:128-134. [PMID: 31236153 PMCID: PMC6575135 DOI: 10.14740/gr1145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/30/2019] [Indexed: 12/24/2022] Open
Abstract
Background Micropapillary carcinoma (MPC) has been reported as an aggressive variant of colorectal carcinoma (CRC) associated with frequent lymphovascular invasion and poor outcome. Altered glycogen metabolism by metabolic reprogramming plays a critical role for cancer cell growth and survival. We aimed to investigate glucose metabolic reprogramming in colorectal MPC. Methods Immmunostains for Ki-67 and glucose transporter 1 (GLUT1) were performed on 10 colorectal MPCs. Real-time PCR analysis of expressions of GLUT1 and glycogen metabolizing enzymes: glycogen synthase (GYS1) and glycogen phosphorylase (PYGL) was performed on cultured monolayer and three-dimensional (3D) spheroid HCT116 colon cancer cells. Results GLUT1 was strongly expressed in MPC as compared to adjacent conventional glandular component, and was also significantly increased expression in 3D spheroids. Upregulation of GYS1 and PYGL was markedly increased in 3D spheroids. The proliferation rate (Ki-67) of MPC was significantly lower compared to conventional glandular component. The 3D spheroids showed increased cell cycle arrest. Our results demonstrate altered glycogen metabolism in colorectal MPC. Conclusion The reprogramming of glycogen metabolism in MPC provides a source of energy contributing to tumor cell survival in a low proliferation state. Targeting glucose-regulated metabolism may warrant consideration as possible MPC therapies.
Collapse
Affiliation(s)
- Monika Vyas
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natalie Patel
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Romulo Celli
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Narendra Wajapeyee
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Dhanpat Jain
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Xuchen Zhang
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
30
|
La Vecchia S, Sebastián C. Metabolic pathways regulating colorectal cancer initiation and progression. Semin Cell Dev Biol 2019; 98:63-70. [PMID: 31129171 DOI: 10.1016/j.semcdb.2019.05.018] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/16/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022]
Abstract
Colorectal cancer (CRC) is one of the most common types of cancer worldwide. Despite recent advances in the molecular genetics of CRC, poor treatment outcomes highlight the need for a better understanding of the underlying mechanisms accounting for tumor initiation and progression. Recently, deregulation of cellular metabolism has emerged as a key hallmark of cancer. Reprogramming of core cellular metabolic pathways by cancer cells provides energy, anaplerotic precursors and reducing equivalents required to support tumor growth. Here, we review key findings implicating cancer metabolism as a major contributor of tumor initiation, growth and metastatic dissemination in CRC. We summarize the metabolic pathways governing stem cell fate in the intestine, the metabolic adaptations of proliferating colon cancer cells and their crosstalk with oncogenic signaling, and how they fulfill the energetic demands imposed by the metastatic cascade. Lastly, we discuss how some of these metabolic pathways could represent new vulnerabilities of CRC cells with the potential to be targeted.
Collapse
Affiliation(s)
- Sofia La Vecchia
- Laboratory of Metabolic Dynamics in Cancer, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo TO, Italy
| | - Carlos Sebastián
- Laboratory of Metabolic Dynamics in Cancer, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo TO, Italy.
| |
Collapse
|
31
|
Lin J, Xia L, Liang J, Han Y, Wang H, Oyang L, Tan S, Tian Y, Rao S, Chen X, Tang Y, Su M, Luo X, Wang Y, Wang H, Zhou Y, Liao Q. The roles of glucose metabolic reprogramming in chemo- and radio-resistance. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:218. [PMID: 31122265 PMCID: PMC6533757 DOI: 10.1186/s13046-019-1214-z] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/07/2019] [Indexed: 12/15/2022]
Abstract
Reprogramming of cancer metabolism is a newly recognized hallmark of malignancy. The aberrant glucose metabolism is associated with dramatically increased bioenergetics, biosynthetic, and redox demands, which is vital to maintain rapid cell proliferation, tumor progression, and resistance to chemotherapy and radiation. When the glucose metabolism of cancer is rewiring, the characters of cancer will also occur corresponding changes to regulate the chemo- and radio-resistance of cancer. The procedure is involved in the alteration of many activities, such as the aberrant DNA repairing, enhanced autophagy, oxygen-deficient environment, and increasing exosomes secretions, etc. Targeting altered metabolic pathways related with the glucose metabolism has become a promising anti-cancer strategy. This review summarizes recent progress in our understanding of glucose metabolism in chemo- and radio-resistance malignancy, and highlights potential molecular targets and their inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Jinguan Lin
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Longzheng Xia
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Jiaxin Liang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yaqian Han
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Heran Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Linda Oyang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Shiming Tan
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yutong Tian
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Shan Rao
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xiaoyan Chen
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yanyan Tang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Min Su
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xia Luo
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Ying Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Hui Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yujuan Zhou
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Qianjin Liao
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
32
|
Wang G, Wang YZ, Yu Y, Wang JJ. Inhibitory ASIC2-mediated calcineurin/NFAT against colorectal cancer by triterpenoids extracted from Rhus chinensis Mill. JOURNAL OF ETHNOPHARMACOLOGY 2019; 235:255-267. [PMID: 30772482 DOI: 10.1016/j.jep.2019.02.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Studies have shown that the etiology and pathogenesis of colorectal cancer are closely related to the tumor microenvironment, and the cancer tissue is still in the state of "energy deficit" and has to promote energy generation through high glycolysis. Rhus chinensis Mill is a Chinese herbal medicine used to treat various types of solid tumors in China. Colorectal cancer (CRC) is a heterogeneous disease group caused by abnormal changes in glucose metabolism resulted in lactic acid production, which remodels acidosis. AIM OF THE STUDY Although previous studies have shown that the active compounds of Rhus chinensis Mill. can inhibit the proliferation of tumor cells, whether its triterpenoids could effectively regulate glycolysis involved in CRC have not been systematically investigated. MATERIALS AND METHODS In this study, the extraction of triterpenoids extract from Rhus chinensis Mill. was obtained, and cell viability assay, the percentage of apoptosis for CRC cells were counted, and matrigel invasion assay and production of lactic acid and glucose uptake assay was determined. we further examined the expression of the key glycolytic enzymes and acid-sending ion channel (ASIC) family members of SW620 cells, and some key proteins in the glycolytic pathway were further verified. RESULTS Notably, triterpenoids (TER) of Rhus chinensis Mill. showed effective anti-proliferative activity and significantly altered protein levels associated with CRC cell survival and glycolysis metabolism. TER could down-regulate the expression of ASIC2, in CRC SW620 cell line. Most importantly, the levels of ASIC2 and calcineurin/nuclear factor of activated T cells (NFAT) were also down-regulated by TER. Furthermore, inhibition of activated the ASIC2-mediated calcineurin/NFAT1 pathway and target gene transcript expression of MMP-2 and MMP-9 in parallel to reduce, and resulted in the reduced invasion ability by TER treatment. CONCLUSION The potential pathways and targets that involved in glycolysis to excert the anti-CRC effects of main compounds in triterpenoids of Rhus chinensis Mill. were predicted by network pharmacology methods. Our findings thus provided rational evidence that inhibition of the ASIC2-induced calcineurin/NFAT pathway by triterpenoids in Rhus chinensis Mill. profoundly suppressed cell growth and invasion in CRC, which target alternative glycolysis in colorectal tumor cells, may be a useful adjuvant therapy in the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai 200235, China.
| | - Yu-Zhu Wang
- Department of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province 212001, China
| | - Yang Yu
- Department of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province 212001, China
| | - Jun-Jie Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai 200235, China
| |
Collapse
|
33
|
Im DK, Cheong H, Lee JS, Oh MK, Yang KM. Protein kinase CK2-dependent aerobic glycolysis-induced lactate dehydrogenase A enhances the migration and invasion of cancer cells. Sci Rep 2019; 9:5337. [PMID: 30926903 PMCID: PMC6441004 DOI: 10.1038/s41598-019-41852-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 03/14/2019] [Indexed: 02/06/2023] Open
Abstract
We investigated the intracellular metabolic fluxes of protein kinase CK2-activating (Cα OE) cells and role of lactate dehydrogenase A (LDHA) as a contributor of tumorigenesis after reprogrammed glucose metabolism. Facilitated aerobic glycolysis was confirmed via isotope tracer analysis, in which 13C6-Glc or 13C5-Gln was added to the media, following which metabolites converted from Cα OE cells were identified. We found a greater decrease in cell survival, colony-forming ability, migration, and Cα OE cell invasion under glucose (Glc)-depletion conditions than under glutamine (Gln)-depletion conditions. Cancer cell migration and invasion increased due to LDHA elevation of the altered metabolic axis driven by activated CK2. FX11 treatment and LDHA knockdown suppressed migration and invasion through ROS generation, but this was partially reversed by the antioxidant N-acetylcysteine (NAC). Moreover, LDHA inhibition decreased tumor growth in a mouse xenograft model transplanted with Cα OE cells. Finally, we concluded that LDHA is an excellent metabolic target for tumor therapy, based on CK2α derived aerobic glycolysis.
Collapse
Affiliation(s)
- Dae-Kyun Im
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Korea
| | - Heesun Cheong
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Jong Suk Lee
- Biocenter, Gyeonggido Business and Science Accelerator (GBSA), Suwon, Gyeonggi-do, 16229, Republic of Korea
| | - Min-Kyu Oh
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Korea.
| | - Kyung Mi Yang
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|