1
|
Hu Y, Xu W, Chen L. Post-translational modifications and the reprogramming of tumor metabolism. Discov Oncol 2025; 16:929. [PMID: 40418495 DOI: 10.1007/s12672-025-02674-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 05/12/2025] [Indexed: 05/27/2025] Open
Abstract
Metabolic reprogramming occurs alongside tumor development. As cancers advance from precancerous lesions to locally invasive tumors and then to metastatic tumors, metabolic patterns exhibit distinct changes, including mutations in metabolic enzymes and modifications in the activity of metabolic regulatory proteins. Alterations in metabolic patterns can influence tumor evolution, either establishing or alleviating metabolic burdens and facilitating cancer growth. To fully understand how metabolic reprogramming helps tumors grow and find the metabolic activities that are most useful for treating tumors, we need to have a deeper understanding of how metabolic patterns are controlled as tumors grow. Post-translational modifications (PTMs), a critical mechanism in the regulation of protein function, can influence protein activity, stability, and interactions in several ways. In tumor cells, PTMs-mediated metabolic reprogramming is a crucial mechanism for adapting to the challenging microenvironment and sustaining fast growth. This article will deeply explore the intricate regulatory mechanism of PTMs on metabolic reprogramming and its role in tumor progression, with the expectation of providing new theoretical basis and potential targets for tumor treatment.
Collapse
Affiliation(s)
- Yuqing Hu
- Central Laboratory and Precision Medicine Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang Province, China
- Jinhua Key Laboratory of Cancer Nutrition and Metabolism Research, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang Province, China
| | - Wenxia Xu
- Central Laboratory and Precision Medicine Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang Province, China.
- Jinhua Key Laboratory of Cancer Nutrition and Metabolism Research, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang Province, China.
| | - Lin Chen
- Central Laboratory and Precision Medicine Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang Province, China.
- Jinhua Key Laboratory of Cancer Nutrition and Metabolism Research, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang Province, China.
| |
Collapse
|
2
|
Hansman DS, Du J, Casson RJ, Peet DJ. Eye on the horizon: The metabolic landscape of the RPE in aging and disease. Prog Retin Eye Res 2025; 104:101306. [PMID: 39433211 PMCID: PMC11833275 DOI: 10.1016/j.preteyeres.2024.101306] [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: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
To meet the prodigious bioenergetic demands of the photoreceptors, glucose and other nutrients must traverse the retinal pigment epithelium (RPE), a polarised monolayer of cells that lie at the interface between the outer retina and the choroid, the principal vascular layer of the eye. Recent investigations have revealed a metabolic ecosystem in the outer retina where the photoreceptors and RPE engage in a complex exchange of sugars, amino acids, and other metabolites. Perturbation of this delicate metabolic balance has been identified in the aging retina, as well as in age-related macular degeneration (AMD), the leading cause of blindness in the Western world. Also common in the aging and diseased retina are elevated levels of cytokines, oxidative stress, advanced glycation end-products, increased growth factor signalling, and biomechanical stress - all of which have been associated with metabolic dysregulation in non-retinal cell types and tissues. Herein, we outline the role of these factors in retinal homeostasis, aging, and disease. We discuss their effects on glucose, mitochondrial, lipid, and amino acid metabolism in tissues and cell types outside the retina, highlighting the signalling pathways through which they induce these changes. Lastly, we discuss promising avenues for future research investigating the roles of these pathological conditions on retinal metabolism, potentially offering novel therapeutic approaches to combat age-related retinal disease.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
3
|
Bhowmick R, Campit S, Katkam SK, Keshamouni VG, Chandrasekaran S. Genome-scale modeling identifies dynamic metabolic vulnerabilities during the epithelial to mesenchymal transition. Commun Biol 2024; 7:1704. [PMID: 39730911 DOI: 10.1038/s42003-024-07408-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/17/2024] [Indexed: 12/29/2024] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a conserved cellular process critical for embryogenesis, wound healing, and cancer metastasis. During EMT, cells undergo large-scale metabolic reprogramming that supports multiple functional phenotypes including migration, invasion, survival, chemo-resistance and stemness. However, the extent of metabolic network rewiring during EMT is unclear. In this work, using genome-scale metabolic modeling, we perform a meta-analysis of time-course transcriptomics, time-course proteomics, and single-cell transcriptomics EMT datasets from cell culture models stimulated with TGF-β. We uncovered temporal metabolic dependencies in glycolysis and glutamine metabolism, and experimentally validated isoform-specific dependency on Enolase3 for cell survival during EMT. We derived a prioritized list of metabolic dependencies based on model predictions, literature mining, and CRISPR-Cas9 essentiality screens. Notably, enolase and triose phosphate isomerase reaction fluxes significantly correlate with survival of lung adenocarcinoma patients. Our study illustrates how integration of heterogeneous datasets using a mechanistic computational model can uncover temporal and cell-state-specific metabolic dependencies.
Collapse
Affiliation(s)
- Rupa Bhowmick
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Scott Campit
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI, USA
| | - Shiva Krishna Katkam
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Venkateshwar G Keshamouni
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
- LTC Charles S. Kettles VA Medical Center, Research Service (151), Ann Arbor, MI, USA.
| | - Sriram Chandrasekaran
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
- Center for Bioinformatics and Computational Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
4
|
Balakrishnan K, Xiao Y, Chen Y, Dong J. Elevated Expression of Cell Adhesion, Metabolic, and Mucus Secretion Gene Clusters Associated with Tumorigenesis, Metastasis, and Poor Survival in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:4049. [PMID: 39682235 DOI: 10.3390/cancers16234049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
OBJECTIVES Technological advances in identifying gene expression profiles are being applied to study an array of cancers. The goal of this study was to identify differentially expressed genes in pancreatic ductal adenocarcinoma (PDAC) and examine their potential role in tumorigenesis and metastasis. METHODS The transcriptomic profiles of PDAC and non-tumorous tissue samples were derived from the gene expression omnibus (GEO), which is a public repository. The GEO2R tool was used to further derive differentially expressed genes from those profiles. RESULTS In this study, a total of 68 genes were derived from upregulated PDAC genes in three or more transcriptomic profiles and were considered PDAC gene sets. The identified PDAC gene sets were examined in the molecular signatures database (MSigDB) for ontological investigation, which revealed that these genes were involved in the extracellular matrix and associated with the cell adhesion process in PDAC tumorigenesis. The gene set enrichment analysis showed greater enrichment scores for the gene sets. Moreover, the identified gene sets were examined for protein-protein interaction using the STRING database. Based on functional k-means clustering, the following three functional cluster groups were identified in this study: extracellular matrix/cell adhesion, metabolic, and mucus secretion-related protein groups. The receiver operating characteristic (ROC) curve revealed greater specificity and sensitivity for these cluster genes in predicting PDAC tumorigenesis and metastases. In addition, the expression of the cluster genes affects the overall survival rate of PDAC patients. Using the cancer genome atlas (TCGA) database, the associations between expression levels and clinicopathological features were validated. CONCLUSIONS Overall, the genes identified in this study appear to be critical in PDAC development and can serve as potential diagnostic and prognostic targets for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Karthik Balakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yi Xiao
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yuanhong Chen
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
5
|
Youssef KK, Nieto MA. Epithelial-mesenchymal transition in tissue repair and degeneration. Nat Rev Mol Cell Biol 2024; 25:720-739. [PMID: 38684869 DOI: 10.1038/s41580-024-00733-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/02/2024]
Abstract
Epithelial-mesenchymal transitions (EMTs) are the epitome of cell plasticity in embryonic development and cancer; during EMT, epithelial cells undergo dramatic phenotypic changes and become able to migrate to form different tissues or give rise to metastases, respectively. The importance of EMTs in other contexts, such as tissue repair and fibrosis in the adult, has become increasingly recognized and studied. In this Review, we discuss the function of EMT in the adult after tissue damage and compare features of embryonic and adult EMT. Whereas sustained EMT leads to adult tissue degeneration, fibrosis and organ failure, its transient activation, which confers phenotypic and functional plasticity on somatic cells, promotes tissue repair after damage. Understanding the mechanisms and temporal regulation of different EMTs provides insight into how some tissues heal and has the potential to open new therapeutic avenues to promote repair or regeneration of tissue damage that is currently irreversible. We also discuss therapeutic strategies that modulate EMT that hold clinical promise in ameliorating fibrosis, and how precise EMT activation could be harnessed to enhance tissue repair.
Collapse
Affiliation(s)
| | - M Angela Nieto
- Instituto de Neurociencias (CSIC-UMH), Sant Joan d'Alacant, Spain.
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain.
| |
Collapse
|
6
|
Shi X, Xiao B, Feng R. Identification of a glycolysis-related miRNA Signature for Predicting Breast cancer Survival. Mol Biotechnol 2024; 66:1988-2006. [PMID: 37535159 DOI: 10.1007/s12033-023-00837-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
Breast cancer (BC) is a common type of cancer and has a poor prognosis. In this study, we collected the mRNA and miRNA expression profiles of BC patients were obtained from The Cancer Genome Atlas (TCGA) to explore a novel prognostic strategy for BC patients using bioinformatics tools. We found that six glycolysis-related miRNAs (GRmiRs, including hsa-mir-1247, hsa-mir148b, hsa-mir-133a-2, has-mir-1307, hsa-mir-195 and hsa-mir-1258) were correlated with prognosis of BC samples. The risk score model was established based on 6 prognosis-associated GRmiRs. The outcome of high risk group was significantly poorer. Cox regression analysis showed that risk score was an independent prognostic factor. Differentially expressed genes identified between high and low risk groups were mainly enriched in inflammation and immune-related signaling pathways. The proportion of infiltration of 12 kinds of immune cells in high and low risk groups were significantly different. Risk score was closely associated with many immune indexes. Multiple DEGRGs and miRNAs were associated with drugs. In conclusion, glycolysis-related miRNA signature effectively predicts BC prognosis.
Collapse
Affiliation(s)
- Xuejing Shi
- Department of Galactophore, Tianjin Central Hospital of Gynecology and Obstetrics, No. 156 Nankai Sanma Road, Tianjin, Nankai District, 300100, P.R. China
| | - Baoqiang Xiao
- Department of General Surgery, Tianjin Hospital, Tianjin, Hexi District, 300211, P.R. China
| | - Rui Feng
- Department of Galactophore, Tianjin Central Hospital of Gynecology and Obstetrics, No. 156 Nankai Sanma Road, Tianjin, Nankai District, 300100, P.R. China.
| |
Collapse
|
7
|
Farina A, Viggiani V, Cortese F, Moretti M, Tartaglione S, Angeloni A, Anastasi E. Combined PIVKA II and Vimentin-Guided EMT Tracking in Pancreatic Adenocarcinoma Combined Biomarker-Guided EMT Tracking in PDAC. Cancers (Basel) 2024; 16:2362. [PMID: 39001424 PMCID: PMC11240554 DOI: 10.3390/cancers16132362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
"Background/Aim": the current inability to diagnose Pancreatic Cancer Adenocarcinoma (PDAC) at an early stage strongly influences therapeutic strategies. Protein Induced by Vitamin K Absence (PIVKA II) showed an accurate diagnostic performance for PDAC. Since circulating PIVKA II has been recently associated with pancreatic origin cells with Vimentin, an epithelial-to-mesenchymal transition (EMT) early activation marker, the aim of this study was to investigate in vivo the combination between the two proteins. "Materials and Methods": we assayed the presence of PIVKA II and Vimentin proteins by using different diagnostic methods. A total of 20 PDAC patients and 10 healthy donors were tested by Western Blot analysis; 74 PDAC patient and 46 healthy donors were assayed by ECLIA and Elisa. "Results": Western Blot analysis showed the concomitant expression of PIVKA II and Vimentin in PDAC patient sera. Immunometric assay performed on a larger cohort of patients demonstrated that 72% of PIVKA II-positive PDAC patients were Vimentin-positive. Additionally, in a group of PDAC patients with PIVKA II levels ≥2070 ng/mL, the percentage of Vimentin-positive subjects reached 84%. "Conclusion": the association between PIVKA II protein and the EMT suggests that this molecule could be considered a marker of the acquisition of an aggressive phenotype.
Collapse
Affiliation(s)
- Antonella Farina
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| | - Valentina Viggiani
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| | - Francesca Cortese
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| | - Marta Moretti
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| | - Sara Tartaglione
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| | - Antonio Angeloni
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| | - Emanuela Anastasi
- Department of Experimental Medicine, "La Sapienza" University of Rome, V. Le Regina Elena 324, 00161 Rome, Italy
| |
Collapse
|
8
|
Ba Q, Wang X, Hu H, Lu Y. Single-Cell RNA Sequencing Analysis Reveals Metabolic Changes in Epithelial Glycosphingolipids and Establishes a Prognostic Risk Model for Pancreatic Cancer. Diagnostics (Basel) 2024; 14:1094. [PMID: 38893622 PMCID: PMC11171987 DOI: 10.3390/diagnostics14111094] [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: 04/15/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
OBJECTIVE Metabolic reprogramming serves as a distinctive feature of cancer, impacting proliferation and metastasis, with aberrant glycosphingolipid expression playing a crucial role in malignancy. Nevertheless, limited research has investigated the connection between glycosphingolipid metabolism and pancreatic cancer. METHODS This study utilized a single-cell sequencing dataset to analyze the cell composition in pancreatic cancer tissues and quantified single-cell metabolism using a newly developed computational pipeline called scMetabolism. A gene signature developed from the differential expressed genes (DEGs), related to epithelial cell glycosphingolipid metabolism, was established to forecast patient survival, immune response, mutation status, and reaction to chemotherapy with pancreatic adenocarcinoma (PAAD). RESULTS The single-cell sequencing analysis revealed a significant increase in epithelial cell proportions in PAAD, with high glycosphingolipid metabolism occurring in the cancerous tissue. A six-gene signature prognostic model based on abnormal epithelial glycosphingolipid metabolism was created and confirmed using publicly available databases. Patients with PAAD were divided into high- and low-risk categories according to the median risk score, with those in the high-risk group demonstrating a more unfavorable survival outcome in all three cohorts, with higher rates of gene mutations (e.g., KRAS, CDKN2A), increased levels of immunosuppressive cells (macrophages, Th2 cells, regulatory T cells), and heightened sensitivity to Acetalax and Selumetinlb. CONCLUSIONS Abnormal metabolism of glycosphingolipids in epithelial cells may promote the development of PAAD. A model utilizing a gene signature associated with epithelial glycosphingolipids metabolism has been established, serving as a valuable indicator for the prognostic stratification of patients with PAAD.
Collapse
Affiliation(s)
| | | | | | - Yanjun Lu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
9
|
Li F, Si W, Xia L, Yin D, Wei T, Tao M, Cui X, Yang J, Hong T, Wei R. Positive feedback regulation between glycolysis and histone lactylation drives oncogenesis in pancreatic ductal adenocarcinoma. Mol Cancer 2024; 23:90. [PMID: 38711083 PMCID: PMC11071201 DOI: 10.1186/s12943-024-02008-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/24/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Metabolic reprogramming and epigenetic alterations contribute to the aggressiveness of pancreatic ductal adenocarcinoma (PDAC). Lactate-dependent histone modification is a new type of histone mark, which links glycolysis metabolite to the epigenetic process of lactylation. However, the role of histone lactylation in PDAC remains unclear. METHODS The level of histone lactylation in PDAC was identified by western blot and immunohistochemistry, and its relationship with the overall survival was evaluated using a Kaplan-Meier survival plot. The participation of histone lactylation in the growth and progression of PDAC was confirmed through inhibition of histone lactylation by glycolysis inhibitors or lactate dehydrogenase A (LDHA) knockdown both in vitro and in vivo. The potential writers and erasers of histone lactylation in PDAC were identified by western blot and functional experiments. The potential target genes of H3K18 lactylation (H3K18la) were screened by CUT&Tag and RNA-seq analyses. The candidate target genes TTK protein kinase (TTK) and BUB1 mitotic checkpoint serine/threonine kinase B (BUB1B) were validated through ChIP-qPCR, RT-qPCR and western blot analyses. Next, the effects of these two genes in PDAC were confirmed by knockdown or overexpression. The interaction between TTK and LDHA was identified by Co-IP assay. RESULTS Histone lactylation, especially H3K18la level was elevated in PDAC, and the high level of H3K18la was associated with poor prognosis. The suppression of glycolytic activity by different kinds of inhibitors or LDHA knockdown contributed to the anti-tumor effects of PDAC in vitro and in vivo. E1A binding protein p300 (P300) and histone deacetylase 2 were the potential writer and eraser of histone lactylation in PDAC cells, respectively. H3K18la was enriched at the promoters and activated the transcription of mitotic checkpoint regulators TTK and BUB1B. Interestingly, TTK and BUB1B could elevate the expression of P300 which in turn increased glycolysis. Moreover, TTK phosphorylated LDHA at tyrosine 239 (Y239) and activated LDHA, and subsequently upregulated lactate and H3K18la levels. CONCLUSIONS The glycolysis-H3K18la-TTK/BUB1B positive feedback loop exacerbates dysfunction in PDAC. These findings delivered a new exploration and significant inter-relationship between lactate metabolic reprogramming and epigenetic regulation, which might pave the way toward novel lactylation treatment strategies in PDAC therapy.
Collapse
Affiliation(s)
- Fei Li
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Wenzhe Si
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Li Xia
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Deshan Yin
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Tianjiao Wei
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Ming Tao
- Department of General Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaona Cui
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Jin Yang
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China.
| | - Rui Wei
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
10
|
Hansman D, Ma Y, Thomas D, Smith J, Casson R, Peet D. Metabolic reprogramming of the retinal pigment epithelium by cytokines associated with age-related macular degeneration. Biosci Rep 2024; 44:BSR20231904. [PMID: 38567515 PMCID: PMC11043024 DOI: 10.1042/bsr20231904] [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: 11/22/2023] [Revised: 03/17/2024] [Accepted: 04/02/2024] [Indexed: 04/04/2024] Open
Abstract
The complex metabolic relationship between the retinal pigment epithelium (RPE) and photoreceptors is essential for maintaining retinal health. Recent evidence indicates the RPE acts as an adjacent lactate sink, suppressing glycolysis in the epithelium in order to maximize glycolysis in the photoreceptors. Dysregulated metabolism within the RPE has been implicated in the pathogenesis of age-related macular degeneration (AMD), a leading cause of vision loss. In the present study, we investigate the effects of four cytokines associated with AMD, TNFα, TGF-β2, IL-6, and IL-1β, as well as a cocktail containing all four cytokines, on RPE metabolism using ARPE-19 cells, primary human RPE cells, and ex vivo rat eyecups. Strikingly, we found cytokine-specific changes in numerous metabolic markers including lactate production, glucose consumption, extracellular acidification rate, and oxygen consumption rate accompanied by increases in total mitochondrial volume and ATP production. Together, all four cytokines could potently override the constitutive suppression of glycolysis in the RPE, through a mechanism independent of PI3K/AKT, MEK/ERK, or NF-κB. Finally, we observed changes in glycolytic gene expression with cytokine treatment, including in lactate dehydrogenase subunit and glucose transporter expression. Our findings provide new insights into the metabolic changes in the RPE under inflammatory conditions and highlight potential therapeutic targets for AMD.
Collapse
Affiliation(s)
- David S. Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Yuefang Ma
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Daniel Thomas
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Justine R. Smith
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Robert J. Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J. Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
11
|
Wang Y, Chen X, Yang Y. CircRNA-regulated glucose metabolism in ovarian cancer: an emerging landscape for therapeutic intervention. Clin Transl Oncol 2024; 26:584-596. [PMID: 37578652 DOI: 10.1007/s12094-023-03285-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/11/2023] [Indexed: 08/15/2023]
Abstract
Ovarian cancer (OC) has the highest mortality rate among female reproductive system tumours, with limited efficacy of traditional treatments and 5-year survival rates that rarely exceed 40%. Circular RNA (circRNA) is a stable endogenous circular RNA that typically regulates protein expression by binding to downstream miRNA. It has been demonstrated that circRNAs play an important role in the proliferation, migration, and glucose metabolism (such as the Warburg effect) of OC and can regulate the expression of glucose metabolism-related proteins such as GLUT1 and HK2, promoting anaerobic glycolysis of cancer cells, increasing glucose uptake and ATP production, and affecting energy supply and biosynthetic substances to support tumour growth and invasion. This review summarises the formation and characteristics of circRNAs and focuses on their role in regulating glucose metabolism in OC cells and their potential therapeutic value, providing insights for identifying new therapeutic targets.
Collapse
Affiliation(s)
- Yaolong Wang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Gynecological Oncology of Gansu Province, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Xi Chen
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Gynecological Oncology of Gansu Province, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yongxiu Yang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
- Key Laboratory of Gynecological Oncology of Gansu Province, Lanzhou, Gansu, China.
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
12
|
Han X, Song X, Xiao Z, Zhu G, Gao R, Ni B, Li J. Study on the mechanism of MDSC-platelets and their role in the breast cancer microenvironment. Front Cell Dev Biol 2024; 12:1310442. [PMID: 38404689 PMCID: PMC10884319 DOI: 10.3389/fcell.2024.1310442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are key immunosuppressive cells in the tumor microenvironment (TME) that play critical roles in promoting tumor growth and metastasis. Tumor-associated platelets (TAPs) help cancer cells evade the immune system and promote metastasis. In this paper, we describe the interaction between MDSCs and TAPs, including their generation, secretion, activation, and recruitment, as well as the effects of MDSCs and platelets on the generation and changes in the immune, metabolic, and angiogenic breast cancer (BC) microenvironments. In addition, we summarize preclinical and clinical studies, traditional Chinese medicine (TCM) therapeutic approaches, and new technologies related to targeting and preventing MDSCs from interacting with TAPs to modulate the BC TME, discuss the potential mechanisms, and provide perspectives for future development. The therapeutic strategies discussed in this review may have implications in promoting the normalization of the BC TME, reducing primary tumor growth and distant lung metastasis, and improving the efficiency of anti-tumor therapy, thereby improving the overall survival (OS) and progression-free survival (PFS) of patients. However, despite the significant advances in understanding these mechanisms and therapeutic strategies, the complexity and heterogeneity of MDSCs and side effects of antiplatelet agents remain challenging. This requires further investigation in future prospective cohort studies.
Collapse
Affiliation(s)
- Xinpu Han
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Hematology-Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaotong Song
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhigang Xiao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guanghui Zhu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruike Gao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baoyi Ni
- Department of Oncology, First Hospital of Heilongjiang University of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jie Li
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Atiya HI, Gorecki G, Garcia GL, Frisbie LG, Baruwal R, Coffman L. Stromal-Modulated Epithelial-to-Mesenchymal Transition in Cancer Cells. Biomolecules 2023; 13:1604. [PMID: 38002286 PMCID: PMC10669774 DOI: 10.3390/biom13111604] [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: 09/21/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
The ability of cancer cells to detach from the primary site and metastasize is the main cause of cancer- related death among all cancer types. Epithelial-to-mesenchymal transition (EMT) is the first event of the metastatic cascade, resulting in the loss of cell-cell adhesion and the acquisition of motile and stem-like phenotypes. A critical modulator of EMT in cancer cells is the stromal tumor microenvironment (TME), which can promote the acquisition of a mesenchymal phenotype through direct interaction with cancer cells or changes to the broader microenvironment. In this review, we will explore the role of stromal cells in modulating cancer cell EMT, with particular emphasis on the function of mesenchymal stromal/stem cells (MSCs) through the activation of EMT-inducing pathways, extra cellular matrix (ECM) remodeling, immune cell alteration, and metabolic rewiring.
Collapse
Affiliation(s)
- Huda I. Atiya
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Grace Gorecki
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Geyon L. Garcia
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Leonard G. Frisbie
- Department of Integrative Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Roja Baruwal
- Molecular Pharmacology Graduate Program, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lan Coffman
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee Women’s Research Institute, Pittsburgh, PA15213, USA
| |
Collapse
|
14
|
Mathur D, Liao C, Lin W, La Ferlita A, Alaimo S, Taylor S, Zhong Y, Iacobuzio-Donahue C, Ferro A, Xavier JB. The Ratio of Key Metabolic Transcripts Is a Predictive Biomarker of Breast Cancer Metastasis to the Lung. Cancer Res 2023; 83:3478-3491. [PMID: 37526524 PMCID: PMC10570685 DOI: 10.1158/0008-5472.can-23-0153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/12/2023] [Accepted: 07/27/2023] [Indexed: 08/02/2023]
Abstract
Understanding the rewired metabolism underlying organ-specific metastasis in breast cancer could help identify strategies to improve the treatment and prevention of metastatic disease. Here, we used a systems biology approach to compare metabolic fluxes used by parental breast cancer cells and their brain- and lung-homing derivatives. Divergent lineages had distinct, heritable metabolic fluxes. Lung-homing cells maintained high glycolytic flux despite low levels of glycolytic intermediates, constitutively activating a pathway sink into lactate. This strong Warburg effect was associated with a high ratio of lactate dehydrogenase (LDH) to pyruvate dehydrogenase (PDH) expression, which correlated with lung metastasis in patients with breast cancer. Although feature classification models trained on clinical characteristics alone were unable to predict tropism, the LDH/PDH ratio was a significant predictor of metastasis to the lung but not to other organs, independent of other transcriptomic signatures. High lactate efflux was also a trait in lung-homing metastatic pancreatic cancer cells, suggesting that lactate production may be a convergent phenotype in lung metastasis. Together, these analyses highlight the essential role that metabolism plays in organ-specific cancer metastasis and identify a putative biomarker for predicting lung metastasis in patients with breast cancer. SIGNIFICANCE Lung-homing metastatic breast cancer cells express an elevated ratio of lactate dehydrogenase to pyruvate dehydrogenase, indicating that ratios of specific metabolic gene transcripts have potential as metabolic biomarkers for predicting organ-specific metastasis.
Collapse
Affiliation(s)
- Deepti Mathur
- Program for Computational and Systems Biology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Chen Liao
- Program for Computational and Systems Biology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Wendy Lin
- Program for Computational and Systems Biology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Alessandro La Ferlita
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Salvatore Alaimo
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| | - Samuel Taylor
- Weill Cornell–Rockefeller–Sloan Kettering Tri-Institutional MD–PhD Program, New York, New York
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Yi Zhong
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, New Jersey
| | | | - Alfredo Ferro
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| | - Joao B. Xavier
- Program for Computational and Systems Biology, Memorial Sloan-Kettering Cancer Center, New York, New York
| |
Collapse
|
15
|
Zhang W, Zhou H, Li H, Mou H, Yinwang E, Xue Y, Wang S, Zhang Y, Wang Z, Chen T, Sun H, Wang F, Zhang J, Chai X, Chen S, Li B, Zhang C, Gao J, Ye Z. Cancer cells reprogram to metastatic state through the acquisition of platelet mitochondria. Cell Rep 2023; 42:113147. [PMID: 37756158 DOI: 10.1016/j.celrep.2023.113147] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/30/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Metastasis is the major cause of cancer deaths, and cancer cells evolve to adapt to various tumor microenvironments, which hinders the treatment of tumor metastasis. Platelets play critical roles in tumor development, especially during metastasis. Here, we elucidate the role of platelet mitochondria in tumor metastasis. Cancer cells are reprogrammed to a metastatic state through the acquisition of platelet mitochondria via the PINK1/Parkin-Mfn2 pathway. Furthermore, platelet mitochondria regulate the GSH/GSSG ratio and reactive oxygen species (ROS) in cancer cells to promote lung metastasis of osteosarcoma. Impairing platelet mitochondrial function has proven to be an efficient approach to impair metastasis, providing a direction for osteosarcoma therapy. Our findings demonstrate mitochondrial transfer between platelets and cancer cells and suggest a role for platelet mitochondria in tumor metastasis.
Collapse
Affiliation(s)
- Wenkan Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hao Zhou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hengyuan Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Haochen Mou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Eloy Yinwang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yucheng Xue
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Shengdong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yongxing Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zenan Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Tao Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hangxiang Sun
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Fangqian Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jiahao Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xupeng Chai
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Shixin Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Binghao Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Zhaoming Ye
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, People's Republic of China; Institute of Orthopedic Research, Zhejiang University, Hangzhou 310009, People's Republic of China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
16
|
Kar S, Maji N, Sen K, Roy S, Maity A, Ghosh Dastidar S, Nath S, Basu G, Basu M. Reprogramming of glucose metabolism via PFKFB4 is critical in FGF16-driven invasion of breast cancer cells. Biosci Rep 2023; 43:BSR20230677. [PMID: 37222403 PMCID: PMC10407156 DOI: 10.1042/bsr20230677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/25/2023] Open
Abstract
Fibroblast growth factors (FGFs) are expressed in both developing and adult tissues and play important roles in embryogenesis, tissue homeostasis, angiogenesis, and neoplastic transformation. Here, we report the elevated expression of FGF16 in human breast tumor and investigate its potential involvement in breast cancer progression. The onset of epithelial-mesenchymal transition (EMT), a prerequisite for cancer metastasis, was observed in human mammary epithelial cell-line MCF10A by FGF16. Further study unveiled that FGF16 alters mRNA expression of a set of extracellular matrix genes to promote cellular invasion. Cancer cells undergoing EMT often show metabolic alteration to sustain their continuous proliferation and energy-intensive migration. Similarly, FGF16 induced a significant metabolic shift toward aerobic glycolysis. At the molecular level, FGF16 enhanced GLUT3 expression to facilitate glucose transport into cells, which through aerobic glycolysis generates lactate. The bi-functional protein, 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase 4 (PFKFB4) was found to be a mediator in FGF16-driven glycolysis and subsequent invasion. Furthermore, PFKFB4 was found to play a critical role in promoting lactate-induced cell invasion since silencing PFKFB4 decreased lactate level and rendered the cells less invasive. These findings support potential clinical intervention of any of the members of FGF16-GLUT3-PFKFB4 axis to control the invasion of breast cancer cells.
Collapse
Affiliation(s)
- Swarnali Kar
- Department of Biophysics, Bose Institute, P 1/12, C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Nilanjana Maji
- Department of Biophysics, Bose Institute, P 1/12, C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Kamalika Sen
- Bioinformatics Centre, Bose Institute, P 1/12, C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Stuti Roy
- Basic and Translational Research Division, Saroj Gupta Cancer Centre and Research Institute (SGCC & RI), Kolkata 700063, India
| | - Atanu Maity
- Bioinformatics Centre, Bose Institute, P 1/12, C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Shubhra Ghosh Dastidar
- Bioinformatics Centre, Bose Institute, P 1/12, C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Somsubhra Nath
- Basic and Translational Research Division, Saroj Gupta Cancer Centre and Research Institute (SGCC & RI), Kolkata 700063, India
| | - Gautam Basu
- Department of Biophysics, Bose Institute, P 1/12, C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Moitri Basu
- Department of Biophysics, Bose Institute, P 1/12, C.I.T. Scheme VIIM, Kolkata 700054, India
| |
Collapse
|
17
|
Xing W, Li X, Zhou Y, Li M, Zhu M. Lactate metabolic pathway regulates tumor cell metastasis and its use as a new therapeutic target. EXPLORATION OF MEDICINE 2023:541-559. [DOI: https:/doi.org/10.37349/emed.2023.00160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/17/2023] [Indexed: 09/04/2023] Open
Abstract
Abnormal energy metabolism is one of the ten hallmarks of tumors, and tumor cell metabolism provides energy and a suitable microenvironment for tumorigenesis and metastasis. Tumor cells can consume large amounts of glucose and produce large amounts of lactate through glycolysis even in the presence of oxygen, a process called aerobic glycolysis, also known as the Warburg effect. Lactate is the end product of the aerobic glycolysis. Lactate dehydrogenase A (LDHA), which is highly expressed in cancer cells, promotes lactate production and transports lactate to the tumor microenvironment and is taken up by surrounding stromal cells under the action of monocarboxylate transporter 1/4 (MCT1/4), which in turn influences the immune response and enhances the invasion and metastasis of cancer cells. Therapeutic strategies targeting lactate metabolism have been intensively investigated, focusing on its metastasis-promoting properties and various target inhibitors; AZD3965, an MCT1 inhibitor, has entered phase I clinical trials, and the LDHA inhibitor N-hydroxyindole (NHI) has shown cancer therapeutic activity in pre-clinical studies. Interventions targeting lactate metabolism are emerging as a promising option for cancer therapy, with chemotherapy or radiotherapy combined with lactate-metabolism-targeted drugs adding to the effectiveness of cancer treatment. Based on current research, this article outlines the role of lactate metabolism in tumor metastasis and the potential value of inhibitors targeting lactate metabolism in cancer therapy.
Collapse
Affiliation(s)
- Weimei Xing
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China
| | - Xiaowei Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China
| | - Yuli Zhou
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China
| | - Mengsen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China; Department of Medical Oncology, Second Affiliated Hospital, Hainan Medical University, Haikou 570311, Hainan, China; Institution of Tumour, First Affiliated Hospital, Hainan Medical University, Haikou 570102, Hainan, China
| | - Mingyue Zhu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China
| |
Collapse
|
18
|
Fan JR, Chang SN, Chu CT, Chen HC. AKT2-mediated nuclear deformation leads to genome instability during epithelial-mesenchymal transition. iScience 2023; 26:106992. [PMID: 37378334 PMCID: PMC10291577 DOI: 10.1016/j.isci.2023.106992] [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: 02/03/2023] [Revised: 05/04/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Nuclear deformation has been observed in some cancer cells for decades, but its underlying mechanism and biological significance remain elusive. To address these questions, we employed human lung cancer A549 cell line as a model in context with transforming growth factor β (TGFβ)-induced epithelial-mesenchymal transition. Here, we report that nuclear deformation induced by TGFβ is concomitant with increased phosphorylation of lamin A at Ser390, defective nuclear lamina and genome instability. AKT2 and Smad3 serve as the downstream effectors for TGFβ to induce nuclear deformation. AKT2 directly phosphorylates lamin A at Ser390, whereas Smad3 is required for AKT2 activation upon TGFβ stimulation. Expression of the lamin A mutant with a substitution of Ser390 to Ala or suppression of AKT2 or Smad3 prevents nuclear deformation and genome instability induced by TGFβ. These findings reveal a molecular mechanism for TGFβ-induced nuclear deformation and establish a role of nuclear deformation in genome instability during epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Jia-Rong Fan
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Sung-Nian Chang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Ching-Tung Chu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Hong-Chen Chen
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| |
Collapse
|
19
|
Tong Y, Qi Y, Xiong G, Li J, Scott TL, Chen J, He D, Li L, Wang C, Lane AN, Xu R. The PLOD2/succinate axis regulates the epithelial-mesenchymal plasticity and cancer cell stemness. Proc Natl Acad Sci U S A 2023; 120:e2214942120. [PMID: 37155842 PMCID: PMC10194013 DOI: 10.1073/pnas.2214942120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 03/08/2023] [Indexed: 05/10/2023] Open
Abstract
Aberrant accumulation of succinate has been detected in many cancers. However, the cellular function and regulation of succinate in cancer progression is not completely understood. Using stable isotope-resolved metabolomics analysis, we showed that the epithelial mesenchymal transition (EMT) was associated with profound changes in metabolites, including elevation of cytoplasmic succinate levels. The treatment with cell-permeable succinate induced mesenchymal phenotypes in mammary epithelial cells and enhanced cancer cell stemness. Chromatin immunoprecipitation and sequence analysis showed that elevated cytoplasmic succinate levels were sufficient to reduce global 5-hydroxymethylcytosinene (5hmC) accumulation and induce transcriptional repression of EMT-related genes. We showed that expression of procollagen-lysine,2-oxoglutarate 5-dioxygenase 2 (PLOD2) was associated with elevation of cytoplasmic succinate during the EMT process. Silencing of PLOD2 expression in breast cancer cells reduced succinate levels and inhibited cancer cell mesenchymal phenotypes and stemness, which was accompanied by elevated 5hmC levels in chromatin. Importantly, exogenous succinate rescued cancer cell stemness and 5hmC levels in PLOD2-silenced cells, suggesting that PLOD2 promotes cancer progression at least partially through succinate. These results reveal the previously unidentified function of succinate in enhancing cancer cell plasticity and stemness.
Collapse
Affiliation(s)
- Yuxin Tong
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| | - Yifei Qi
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| | - Gaofeng Xiong
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| | - Junyan Li
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| | - Timothy L. Scott
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY40536
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY40536
| | - Jie Chen
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| | - Daheng He
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
| | - Linzhang Li
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
| | - Andrew N. Lane
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY40536
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY40536
| | - Ren Xu
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| |
Collapse
|
20
|
Tian X, Wang Y, Xu W, Tang H, Zhu S, Anwaier A, Liu W, Wang W, Zhu W, Su J, Qu Y, Zhang H, Ye D. Special issue "The advance of solid tumor research in China": Multi-omics analysis based on 1311 clear cell renal cell carcinoma samples identifies a glycolysis signature associated with prognosis and treatment response. Int J Cancer 2023; 152:66-78. [PMID: 35579992 PMCID: PMC9796246 DOI: 10.1002/ijc.34121] [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: 12/18/2021] [Revised: 04/10/2022] [Accepted: 04/29/2022] [Indexed: 01/01/2023]
Abstract
In clear cell renal cell carcinoma (ccRCC), glycolysis is enhanced mainly because of the increased expression of key enzymes in glycolysis. Hence, the discovery of new molecular biomarkers for glycolysis may help guide and establish a precise system of diagnosis and treatment for ccRCC. Expression profiles of 1079 tumor samples of ccRCC patients (including 311 patients treated with everolimus or nivolumab) were downloaded from public databases. Proteomic profiles of 232 ccRCC samples were obtained from Fudan University Shanghai Cancer Center (FUSCC). Biological changes, tumor microenvironment and prognostic differences were explored between samples with various glycolysis characteristics. There were significant differences in CD8+ effector T cells, epithelial-to-mesenchymal transition and pan-fibroblast TGFb between the Low and High glyScore groups. The tumor mutation burden of the Low glyScore group was lower than that of the High glyScore group. And higher glyScore was significantly associated with worse overall survival (OS) in 768 ccRCC patients (P < .0001). External validation in FUSCC cohort also indicated that glyScore was of strong ability for predicting OS (P < .05). GlyScore may serve as a biomarker for predicting everolimus response in ccRCC patients due to its significant associations with progression-free survival (PFS). And glyScore may also predict overall survival in patients treated with nivolumab. We calculated the glyScore in ccRCC and the defined glyScore was of strong ability for predicting OS. In addition, glyScore may also serve as a biomarker for predicting PFS in patients treated with everolimus and could predict OS in patients treated with nivolumab.
Collapse
Affiliation(s)
- Xi Tian
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yue Wang
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Wenhao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Haidan Tang
- Affiliated Hospital of Youjiang Medical University for NationalitiesGuangxiChina
| | - Shuxuan Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Aihetaimujiang Anwaier
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Wangrui Liu
- Affiliated Hospital of Youjiang Medical University for NationalitiesGuangxiChina,Department of Interventional Oncology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenfeng Wang
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Wenkai Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jiaqi Su
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yuanyuan Qu
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
21
|
Huang Y. Targeting glycolysis for cancer therapy using drug delivery systems. J Control Release 2023; 353:650-662. [PMID: 36493949 DOI: 10.1016/j.jconrel.2022.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/03/2022] [Indexed: 12/15/2022]
Abstract
There is close crosstalk between cancer metabolism and immunity. Cancer metabolism regulation is a promising therapeutic target for cancer immunotherapy. Warburg effect is characterized by abnormal glucose metabolism that includes common features of increased glucose uptake and lactate production. The aerobic glycolysis can reprogram the cancer cells and promote the formation of a suppressive immune microenvironment. As a case in point, lactate plays an essential role in tumorigenesis, which is the end product of glycolysis as well as serves as a fuel supporting cancer cell survival. Meanwhile, it is also an important immune regulator that drives immunosuppression in tumors. Immunometabolic therapy is to intervene tumor metabolism and regulate the related metabolites that participate in the innate and acquired immunity, thereby reinstalling the immune balance and eliciting anticancer immune responses. In this contribution to the Orations - New Horizons of the Journal of controlled Release I will provide an overview of glucose metabolism in tumors and its effects on drug resistance and tumor metastasis, and present the advance of glycolysis-targeting therapy strategies with drug delivery techniques, as well as discuss the challenges in glycolysis-targeting immunometabolic therapy.
Collapse
Affiliation(s)
- Yongzhuo Huang
- Zhongshan Institute for Drug Discovery, SIMM, CAS, China; Shanghai Institute of Materia Medica Chinese Academy of Science, China.
| |
Collapse
|
22
|
Chen L, Zhu C, Pan F, Chen Y, Xiong L, Li Y, Chu X, Huang G. Platelets in the tumor microenvironment and their biological effects on cancer hallmarks. Front Oncol 2023; 13:1121401. [PMID: 36937386 PMCID: PMC10022734 DOI: 10.3389/fonc.2023.1121401] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
The interplay between platelets and tumors has long been studied. It has been widely accepted that platelets could promote tumor metastasis. However, the precise interactions between platelets and tumor cells have not been thoroughly investigated. Although platelets may play complex roles in multiple steps of tumor development, most studies focus on the platelets in the circulation of tumor patients. Platelets in the primary tumor microenvironment, in addition to platelets in the circulation during tumor cell dissemination, have recently been studied. Their effects on tumor biology are gradually figured out. According to updated cancer hallmarks, we reviewed the biological effects of platelets on tumors, including regulating tumor proliferation and growth, promoting cancer invasion and metastasis, inducing vasculature, avoiding immune destruction, and mediating tumor metabolism and inflammation.
Collapse
Affiliation(s)
- Lilan Chen
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Chunyan Zhu
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Fan Pan
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Ying Chen
- Division of Immunology, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Lei Xiong
- Department of Cardio-Thoracic Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Yan Li
- Department of Respiratory Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
- *Correspondence: Guichun Huang, ; Yan Li, ; Xiaoyuan Chu,
| | - Xiaoyuan Chu
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
- *Correspondence: Guichun Huang, ; Yan Li, ; Xiaoyuan Chu,
| | - Guichun Huang
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
- *Correspondence: Guichun Huang, ; Yan Li, ; Xiaoyuan Chu,
| |
Collapse
|
23
|
The Molecular and Cellular Strategies of Glioblastoma and Non-Small-Cell Lung Cancer Cells Conferring Radioresistance. Int J Mol Sci 2022; 23:ijms232113577. [PMID: 36362359 PMCID: PMC9656305 DOI: 10.3390/ijms232113577] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Ionizing radiation (IR) has been shown to play a crucial role in the treatment of glioblastoma (GBM; grade IV) and non-small-cell lung cancer (NSCLC). Nevertheless, recent studies have indicated that radiotherapy can offer only palliation owing to the radioresistance of GBM and NSCLC. Therefore, delineating the major radioresistance mechanisms may provide novel therapeutic approaches to sensitize these diseases to IR and improve patient outcomes. This review provides insights into the molecular and cellular mechanisms underlying GBM and NSCLC radioresistance, where it sheds light on the role played by cancer stem cells (CSCs), as well as discusses comprehensively how the cellular dormancy/non-proliferating state and polyploidy impact on their survival and relapse post-IR exposure.
Collapse
|
24
|
Shi X, Yang J, Deng S, Xu H, Wu D, Zeng Q, Wang S, Hu T, Wu F, Zhou H. TGF-β signaling in the tumor metabolic microenvironment and targeted therapies. J Hematol Oncol 2022; 15:135. [PMID: 36115986 PMCID: PMC9482317 DOI: 10.1186/s13045-022-01349-6] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/24/2022] [Indexed: 12/30/2022] Open
Abstract
AbstractTransforming growth factor-β (TGF-β) signaling has a paradoxical role in cancer progression, and it acts as a tumor suppressor in the early stages but a tumor promoter in the late stages of cancer. Once cancer cells are generated, TGF-β signaling is responsible for the orchestration of the immunosuppressive tumor microenvironment (TME) and supports cancer growth, invasion, metastasis, recurrence, and therapy resistance. These progressive behaviors are driven by an “engine” of the metabolic reprogramming in cancer. Recent studies have revealed that TGF-β signaling regulates cancer metabolic reprogramming and is a metabolic driver in the tumor metabolic microenvironment (TMME). Intriguingly, TGF-β ligands act as an “endocrine” cytokine and influence host metabolism. Therefore, having insight into the role of TGF-β signaling in the TMME is instrumental for acknowledging its wide range of effects and designing new cancer treatment strategies. Herein, we try to illustrate the concise definition of TMME based on the published literature. Then, we review the metabolic reprogramming in the TMME and elaborate on the contribution of TGF-β to metabolic rewiring at the cellular (intracellular), tissular (intercellular), and organismal (cancer-host) levels. Furthermore, we propose three potential applications of targeting TGF-β-dependent mechanism reprogramming, paving the way for TGF-β-related antitumor therapy from the perspective of metabolism.
Collapse
|
25
|
Fan N, Fu H, Feng X, Chen Y, Wang J, Wu Y, Bian Y, Li Y. Long non-coding RNAs play an important regulatory role in tumorigenesis and tumor progression through aerobic glycolysis. Front Mol Biosci 2022; 9:941653. [PMID: 36072431 PMCID: PMC9441491 DOI: 10.3389/fmolb.2022.941653] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Compared to normal cells, cancer cells generate ATP mainly through aerobic glycolysis, which promotes tumorigenesis and tumor progression. Long non-coding RNAs (LncRNAs) are a class of transcripts longer than 200 nucleotides with little or without evident protein-encoding function. LncRNAs are involved in the ten hallmarks of cancer, interestingly, they are also closely associated with aerobic glycolysis. However, the mechanism of this process is non-transparent to date. Demonstrating the mechanism of lncRNAs regulating tumorigenesis and tumor progression through aerobic glycolysis is particularly critical for cancer therapy, and may provide novel therapeutic targets or strategies in cancer treatment. In this review, we discuss the role of lncRNAs and aerobic glycolysis in tumorigenesis and tumor progression, and further explore their interaction, in hope to provide a novel therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Ni Fan
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hui Fu
- College of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuchen Feng
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yatong Chen
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jingyu Wang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuqi Wu
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhong Bian
- College of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Yuhong Bian, ; Yingpeng Li,
| | - Yingpeng Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Yuhong Bian, ; Yingpeng Li,
| |
Collapse
|
26
|
Immunometabolic rewiring of tubular epithelial cells in kidney disease. Nat Rev Nephrol 2022; 18:588-603. [PMID: 35798902 DOI: 10.1038/s41581-022-00592-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2022] [Indexed: 12/20/2022]
Abstract
Kidney tubular epithelial cells (TECs) have a crucial role in the damage and repair response to acute and chronic injury. To adequately respond to constant changes in the environment, TECs have considerable bioenergetic needs, which are supported by metabolic pathways. Although little is known about TEC metabolism, a number of ground-breaking studies have shown that defective glucose metabolism or fatty acid oxidation in the kidney has a key role in the response to kidney injury. Imbalanced use of these metabolic pathways can predispose TECs to apoptosis and dedifferentiation, and contribute to lipotoxicity and kidney injury. The accumulation of lipids and aberrant metabolic adaptations of TECs during kidney disease can also be driven by receptors of the innate immune system. Similar to their actions in innate immune cells, pattern recognition receptors regulate the metabolic rewiring of TECs, causing cellular dysfunction and lipid accumulation. TECs should therefore be considered a specialized cell type - like cells of the innate immune system - that is subject to regulation by immunometabolism. Targeting energy metabolism in TECs could represent a strategy for metabolically reprogramming the kidney and promoting kidney repair.
Collapse
|
27
|
Ohara Y, Valenzuela P, Hussain SP. The interactive role of inflammatory mediators and metabolic reprogramming in pancreatic cancer. Trends Cancer 2022; 8:556-569. [PMID: 35525794 PMCID: PMC9233125 DOI: 10.1016/j.trecan.2022.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 10/18/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by its highly reactive inflammatory desmoplastic stroma with evidence of an extensive tumor stromal interaction largely mediated by inflammatory factors. KRAS mutation and inflammatory signaling promote protumorigenic events, including metabolic reprogramming with several inter-regulatory crosstalks to fulfill the high demand of energy and regulate oxidative stress for tumor growth and progression. Notably, the more aggressive molecular subtype of PDAC enhances influx of glycolytic intermediates. This review focuses on the interactive role of inflammatory signaling and metabolic reprogramming with emerging evidence of crosstalk, which supports the development, progression, and therapeutic resistance of PDAC. Understanding the emerging crosstalk between inflammation and metabolic adaptations may identify potential targets and develop novel therapeutic approaches for PDAC.
Collapse
Affiliation(s)
- Yuuki Ohara
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paloma Valenzuela
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - S Perwez Hussain
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
28
|
Afzaal A, Rehman K, Kamal S, Akash MSH. Versatile role of sirtuins in metabolic disorders: From modulation of mitochondrial function to therapeutic interventions. J Biochem Mol Toxicol 2022; 36:e23047. [PMID: 35297126 DOI: 10.1002/jbt.23047] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 01/11/2022] [Accepted: 03/02/2022] [Indexed: 12/17/2022]
Abstract
Sirtuins (SIRT1-7) are distinct histone deacetylases (HDACs) whose activity is determined by cellular metabolic status andnicotinamide adenine dinucleotide (NAD+ ) levels. HDACs of class III are the members of the SIRT's protein family. SIRTs are the enzymes that modulate mitochondrial activity and energy metabolism. SIRTs have been linked to a number of clinical and physiological operations, such as energy responses to low-calorie availability, aging, stress resistance, inflammation, and apoptosis. Mammalian SIRT2 orthologs have been identified as SIRT1-7 that are found in several subcellular sections, including the cytoplasm (SIRT1, 2), mitochondrial matrix (SIRT3, 4, 5), and the core (SIRT1, 2, 6, 7). For their deacetylase or ADP-ribosyl transferase action, all SIRTs require NAD+ and are linked to cellular energy levels. Evolutionarily, SIRT1 is related to yeast's SIRT2 as well as received primary attention in the circulatory system. An endogenous protein, SIRT1 is involved in the development of heart failure and plays a key role in cell death and survival. SIRT2 downregulation protects against ischemic-reperfusion damage. Increase in human longevity is caused by an increase in SIRT3 expression. Cardiomyocytes are also protected by SIRT3 from oxidative damage and aging, as well as suppressing cardiac hypertrophy. SIRT4 and SIRT5 perform their roles in the heart. SIRT6 has also been linked to a reduction in heart hypertrophy. SIRT7 is known to be involved in the regulation of stress responses and apoptosis in the heart.
Collapse
Affiliation(s)
- Ammara Afzaal
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | - Shagufta Kamal
- Department of Biochemistry, Government College University, Faisalabad, Pakistan
| | | |
Collapse
|
29
|
Gao M, Mei C, Guo Y, Xia P, Zhang H, Liu Y, Yao Y, Jiang X, Yuan Y, Qian Y. Loss of FBP1 by aPKC-ι/Snail Pathway-Mediated Repression Promotes Invasion and Aerobic Glycolysis of Intrahepatic Cholangiocarcinoma. Front Oncol 2021. [DOI: 10.3389/fonc.2021.756419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is one of the most commonly diagnosed malignancies worldwide, and the second most common primary liver tumor. The lack of effective diagnostic and treatment methods results in poor patient prognosis and high mortality rate. Atypical protein kinase C-ι (aPKC-ι) is highly expressed in primary and metastatic ICC tissues, and regulates epithelial mesenchymal transition (EMT) through the aPKC-ι/P-Sp1/Snail signaling pathway. Recent studies have correlated aberrant glucose metabolism with EMT. Given the vital role of FBP1 in regulating glucose metabolism in cancer cells, we hypothesized that aPKC-ι downregulates FBP1 in ICC cells through the Snai1 pathway, and enhances glycolysis and metastasis. We confirmed the ability of aPKC-ι promotes glycolysis, invasion and metastasis of cancer cells, and further demonstrated that FBP1 inhibits the malignant properties of ICC cells by antagonizing aPKC-ι. Our findings provide novel insights into the molecular mechanisms of ICC progression and metastasis, as well as a theoretical basis for exploring new treatment strategies.
Collapse
|
30
|
Nam H, Kundu A, Karki S, Brinkley GJ, Chandrashekar DS, Kirkman RL, Liu J, Liberti MV, Locasale JW, Mitchell T, Varambally S, Sudarshan S. The TGF-β/HDAC7 axis suppresses TCA cycle metabolism in renal cancer. JCI Insight 2021; 6:148438. [PMID: 34609963 PMCID: PMC8663777 DOI: 10.1172/jci.insight.148438] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 09/30/2021] [Indexed: 01/06/2023] Open
Abstract
Mounting evidence points to alterations in mitochondrial metabolism in renal cell carcinoma (RCC). However, the mechanisms that regulate the TCA cycle in RCC remain uncharacterized. Here, we demonstrate that loss of TCA cycle enzyme expression is retained in RCC metastatic tissues. Moreover, proteomic analysis demonstrates that reduced TCA cycle enzyme expression is far more pronounced in RCC relative to other tumor types. Loss of TCA cycle enzyme expression is correlated with reduced expression of the transcription factor PGC-1α, which is also lost in RCC tissues. PGC-1α reexpression in RCC cells restores the expression of TCA cycle enzymes in vitro and in vivo and leads to enhanced glucose carbon incorporation into TCA cycle intermediates. Mechanistically, TGF-β signaling, in concert with histone deacetylase 7 (HDAC7), suppresses TCA cycle enzyme expression. Our studies show that pharmacologic inhibition of TGF-β restores the expression of TCA cycle enzymes and suppresses tumor growth in an orthotopic model of RCC. Taken together, this investigation reveals a potentially novel role for the TGF-β/HDAC7 axis in global suppression of TCA cycle enzymes in RCC and provides insight into the molecular basis of altered mitochondrial metabolism in this malignancy.
Collapse
Affiliation(s)
| | | | | | | | - Darshan S Chandrashekar
- Department of Pathology, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | | | - Juan Liu
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Maria V Liberti
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | | | - Sooryanarayana Varambally
- Department of Pathology, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA.,O'Neal Comprehensive Cancer Center, UAB, Birmingham, Alabama, USA
| | - Sunil Sudarshan
- Department of Urology and.,O'Neal Comprehensive Cancer Center, UAB, Birmingham, Alabama, USA.,Birmingham Veterans Affairs Medical Center, Birmingham, Alabama, USA
| |
Collapse
|
31
|
Zhou MY, Cheng ML, Huang T, Hu RH, Zou GL, Li H, Zhang BF, Zhu JJ, Liu YM, Liu Y, Zhao XK. Transforming growth factor beta-1 upregulates glucose transporter 1 and glycolysis through canonical and noncanonical pathways in hepatic stellate cells. World J Gastroenterol 2021; 27:6908-6926. [PMID: 34790014 PMCID: PMC8567474 DOI: 10.3748/wjg.v27.i40.6908] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/19/2021] [Accepted: 09/08/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatic stellate cells (HSCs) are the key effector cells mediating the occurrence and development of liver fibrosis, while aerobic glycolysis is an important metabolic characteristic of HSC activation. Transforming growth factor-β1 (TGF-β1) induces aerobic glycolysis and is a driving factor for metabolic reprogramming. The occurrence of glycolysis depends on a high glucose uptake level. Glucose transporter 1 (GLUT1) is the most widely distributed glucose transporter in the body and mainly participates in the regulation of carbohydrate metabolism, thus affecting cell proliferation and growth. However, little is known about the relationship between TGF-β1 and GLUT1 in the process of liver fibrosis and the molecular mechanism underlying the promotion of aerobic glycolysis in HSCs.
AIM To investigate the mechanisms of action of GLUT1, TGF-β1 and aerobic glycolysis in the process of HSC activation during liver fibrosis.
METHODS Immunohistochemical staining and immunofluorescence assays were used to examine GLUT1 expression in fibrotic liver tissue. A Seahorse extracellular flux (XF) analyzer was used to examine changes in aerobic glycolytic flux, lactate production levels and glucose consumption levels in HSCs upon TGF-β1 stimulation. The mechanism by which TGF-β1 induces GLUT1 protein expression in HSCs was further explored by inhibiting/promoting the TGF-β1/mothers-against-decapentaplegic-homolog 2/3 (Smad2/3) signaling pathway and inhibiting the p38 and phosphoinositide 3-kinase (PI3K)/AKT signaling pathways. In addition, GLUT1 expression was silenced to observe changes in the growth and proliferation of HSCs. Finally, a GLUT1 inhibitor was used to verify the in vivo effects of GLUT1 on a mouse model of liver fibrosis.
RESULTS GLUT1 protein expression was increased in both mouse and human fibrotic liver tissues. In addition, immunofluorescence staining revealed colocalization of GLUT1 and alpha-smooth muscle actin proteins, indicating that GLUT1 expression was related to the development of liver fibrosis. TGF-β1 caused an increase in aerobic glycolysis in HSCs and induced GLUT1 expression in HSCs by activating the Smad, p38 MAPK and P13K/AKT signaling pathways. The p38 MAPK and Smad pathways synergistically affected the induction of GLUT1 expression. GLUT1 inhibition eliminated the effect of TGF-β1 on HSC proliferation and migration. A GLUT1 inhibitor was administered in a mouse model of liver fibrosis, and GLUT1 inhibition reduced the degree of liver inflammation and liver fibrosis.
CONCLUSION TGF-β1 induces GLUT1 expression in HSCs, a process related to liver fibrosis progression. In vitro experiments revealed that TGF-β1-induced GLUT1 expression might be one of the mechanisms mediating the metabolic reprogramming of HSCs. In addition, in vivo experiments also indicated that the GLUT1 protein promotes the occurrence and development of liver fibrosis.
Collapse
Affiliation(s)
- Ming-Yu Zhou
- Department of Internal Medicine, Guizhou Medical University, Guiyang 550001, Guizhou Province, China
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Ming-Liang Cheng
- Department of Internal Medicine, Guizhou Medical University, Guiyang 550001, Guizhou Province, China
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Tao Huang
- Department of Internal Medicine, Guizhou Medical University, Guiyang 550001, Guizhou Province, China
| | - Rui-Han Hu
- Department of Internal Medicine, Guizhou Medical University, Guiyang 550001, Guizhou Province, China
| | - Gao-Liang Zou
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Hong Li
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Bao-Fang Zhang
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Juan-Juan Zhu
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Yong-Mei Liu
- Clinical Laboratory Center, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Yang Liu
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Xue-Ke Zhao
- Department of Internal Medicine, Guizhou Medical University, Guiyang 550001, Guizhou Province, China
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| |
Collapse
|
32
|
Bisht VS, Giri K, Kumar D, Ambatipudi K. Oxygen and metabolic reprogramming in the tumor microenvironment influences metastasis homing. Cancer Biol Ther 2021; 22:493-512. [PMID: 34696706 DOI: 10.1080/15384047.2021.1992233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Tumor metastasis is the leading cause of cancer mortality, often characterized by abnormal cell growth and invasion to distant organs. The cancer invasion due to epithelial to mesenchymal transition is affected by metabolic and oxygen availability in the tumor-associated micro-environment. A precise alteration in oxygen and metabolic signaling between healthy and metastatic cells is a substantial probe for understanding tumor progression and metastasis. Molecular heterogeneity in the tumor microenvironment help to sustain the metastatic cell growth during their survival shift from low to high metabolic-oxygen-rich sites and reinforces the metastatic events. This review highlighted the crucial role of oxygen and metabolites in metastatic progression and exemplified the role of metabolic rewiring and oxygen availability in cancer cell adaptation. Furthermore, we have also addressed potential applications of altered oxygen and metabolic networking with tumor type that could be a signature pattern to assess tumor growth and chemotherapeutics efficacy in managing cancer metastasis.
Collapse
Affiliation(s)
- Vinod S Bisht
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Kuldeep Giri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Deepak Kumar
- Department of Cancer Biology, Central Drug Research Institute, Lucknow, India.,Academy of Scientific & Innovative Research, New Delhi, India
| | - Kiran Ambatipudi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| |
Collapse
|
33
|
Chen Y, Bei J, Liu M, Huang J, Xie L, Huang W, Cai M, Guo Y, Lin L, Zhu K. Sublethal heat stress-induced O-GlcNAcylation coordinates the Warburg effect to promote hepatocellular carcinoma recurrence and metastasis after thermal ablation. Cancer Lett 2021; 518:23-34. [PMID: 34126196 DOI: 10.1016/j.canlet.2021.06.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/15/2021] [Accepted: 06/05/2021] [Indexed: 02/07/2023]
Abstract
The malignant transformation of residual hepatocellular carcinoma (HCC) cells after thermal ablation is considered as the main factor promoting postoperative HCC progression, which greatly limits the improvement of long-term survival, and at present there is no effective targeted therapeutic strategies. The Warburg effect is a metabolic feature correlated highly with malignant transformation (e.g. epithelial-to-mesenchymal transition [EMT]). Here, we showed that sublethal heat stress triggered a stronger Warburg effect of HCC cells, which contributed to the thermotolerance and invasion of HCC cells. Sublethal heat stress-induced O-GlcNAcylation was involved in this process. Such enhanced Warburg effect in HCC cells may be eliminated through O-GlcNAcylation inhibition, resulting in impaired thermotolerance and EMT, and thereby preventing tumor recurrence and metastasis of HCC-bearing mice after insufficient thermal ablation. Finally, we present evidence that sublethal heat stress-induced O-GlcNAcylation regulates the Warburg effect in HCC cells by promoting hypoxia-inducible factor 1α (HIF-1α) stability. In conclusion, the present study suggests that O-GlcNAcylation coordinates the Warburg effect to promote HCC progression after thermal ablation, which may serve as a novel potential target for controlling postoperative HCC recurrence and metastasis.
Collapse
MESH Headings
- Acylation/physiology
- Animals
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Epithelial-Mesenchymal Transition/physiology
- Heat-Shock Response/physiology
- Humans
- Hyperthermia, Induced/methods
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/pathology
- Warburg Effect, Oncologic
Collapse
Affiliation(s)
- Ye Chen
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China
| | - Jiaxin Bei
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China
| | - Mingyu Liu
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China
| | - Jingjun Huang
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China
| | - Lulu Xie
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China
| | - Wensou Huang
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China
| | - Mingyue Cai
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China
| | - Yongjian Guo
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China
| | - Liteng Lin
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China.
| | - Kangshun Zhu
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, 250 East Changgang Road, Guangzhou, Guangdong Province, 510260, China.
| |
Collapse
|
34
|
Metabolic Reprogramming of Mammary Epithelial Cells during TGF-β-Induced Epithelial-to-Mesenchymal Transition. Metabolites 2021; 11:metabo11090626. [PMID: 34564442 PMCID: PMC8464788 DOI: 10.3390/metabo11090626] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/30/2021] [Accepted: 09/04/2021] [Indexed: 02/05/2023] Open
Abstract
The cytokine transforming growth factor-β (TGF-β) can induce normal breast epithelial cells to take on a mesenchymal phenotype, termed epithelial-to-mesenchymal transition (EMT). While the transcriptional and proteomic changes during TGF-β-induced EMT have been described, the metabolic rewiring that occurs in epithelial cells undergoing EMT is not well understood. Here, we quantitively analyzed the TGF-β-induced metabolic reprogramming during EMT of non-transformed NMuMG mouse mammary gland epithelial cells using nuclear magnetic resonance (NMR) spectroscopy. We found that TGF-β elevates glycolytic and tricarboxylic acid (TCA)-cycle activity and increases glutaminolysis. Additionally, TGF-β affects the hexosamine pathway, arginine-proline metabolism, the cellular redox state, and strongly affects choline metabolism during EMT. TGF-β was found to induce phosphocholine production. A kinase inhibitor RSM-93A that inhibits choline kinase α (CHKα) mitigated TGF-β-induced changes associated with EMT, i.e., increased filamentous (F)-actin stress fiber formation and N-Cadherin mesenchymal marker expression.
Collapse
|
35
|
TGF Beta Induces Vitamin D Receptor and Modulates Mitochondrial Activity of Human Pancreatic Cancer Cells. Cancers (Basel) 2021; 13:cancers13122932. [PMID: 34208208 PMCID: PMC8230851 DOI: 10.3390/cancers13122932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/10/2021] [Indexed: 01/13/2023] Open
Abstract
The inflammatory cytokine TGFβ is both a tumor suppressor during cancer initiation and a promoter of metastasis along cancer progression. Inflammation and cancer are strictly linked, and cancer onset often correlates with the insufficiency of vitamin D, known for its anti-inflammatory properties. In this study, we investigated the interplay between TGFβ and vitamin D in two models of human pancreatic cancer, and we analyzed the metabolic effects of a prolonged TGFβ treatment mimicking the inflammatory environment of pancreatic cancer in vivo. We confirmed the induction of the vitamin D receptor previously described in epithelial cells, but the inhibitory effects of vitamin D on epithelial-mesenchymal transition (EMT) were lost when the hormone was given after a long treatment with TGFβ. Moreover, we detected an ROS-mediated toxicity of the acute treatment with TGFβ, whereas a chronic exposure to low doses had a protumorigenic effect. In fact, it boosted the mitochondrial respiration and cancer cell migration without ROS production and cytotoxicity. Our observations shed some light on the multifaceted role of TGFβ in tumor progression, revealing that a sustained exposure to TGFβ at low doses results in an irreversibly increased EMT associated with a metabolic modulation which favors the formation of metastasis.
Collapse
|
36
|
Jia D, Park JH, Kaur H, Jung KH, Yang S, Tripathi S, Galbraith M, Deng Y, Jolly MK, Kaipparettu BA, Onuchic JN, Levine H. Towards decoding the coupled decision-making of metabolism and epithelial-to-mesenchymal transition in cancer. Br J Cancer 2021; 124:1902-1911. [PMID: 33859341 PMCID: PMC8184790 DOI: 10.1038/s41416-021-01385-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/17/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer cells have the plasticity to adjust their metabolic phenotypes for survival and metastasis. A developmental programme known as epithelial-to-mesenchymal transition (EMT) plays a critical role during metastasis, promoting the loss of polarity and cell-cell adhesion and the acquisition of motile, stem-cell characteristics. Cells undergoing EMT or the reverse mesenchymal-to-epithelial transition (MET) are often associated with metabolic changes, as the change in phenotype often correlates with a different balance of proliferation versus energy-intensive migration. Extensive crosstalk occurs between metabolism and EMT, but how this crosstalk leads to coordinated physiological changes is still uncertain. The elusive connection between metabolism and EMT compromises the efficacy of metabolic therapies targeting metastasis. In this review, we aim to clarify the causation between metabolism and EMT on the basis of experimental studies, and propose integrated theoretical-experimental efforts to better understand the coupled decision-making of metabolism and EMT.
Collapse
Affiliation(s)
- Dongya Jia
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA.
| | - Jun Hyoung Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Harsimran Kaur
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, Karnataka, India
| | - Kwang Hwa Jung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Sukjin Yang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Shubham Tripathi
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA
- PhD Program in Systems, Synthetic, and Physical Biology, Rice University, Houston, TX, USA
- Center for Theoretical Biological Physics and Department of Physics, Northeastern University, Boston, MA, USA
| | - Madeline Galbraith
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA
- Department of Physics and Astronomy, Rice University, Houston, TX, USA
| | - Youyuan Deng
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA
- Applied Physics Graduate Program, Rice University, Houston, TX, USA
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, Karnataka, India
| | - Benny Abraham Kaipparettu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| | - José N Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA.
- Department of Physics and Astronomy, Rice University, Houston, TX, USA.
- Department of Chemistry, Rice University, Houston, TX, USA.
- Department of Biosciences, Rice University, Houston, TX, USA.
| | - Herbert Levine
- Center for Theoretical Biological Physics and Department of Physics, Northeastern University, Boston, MA, USA.
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
37
|
Abstract
Many tumors are now understood to be heterogenous cell populations arising from a minority of epithelial-like cancer stem cells (CSCs). CSCs demonstrate distinctive metabolic signatures from the more differentiated surrounding tumor bulk that confer resistance to traditional chemotherapeutic regimens and potential for tumor relapse. Many CSC phenotypes including metabolism, epithelial-to-mesenchymal transition, cellular signaling pathway activity, and others, arise from altered mitochondrial function and turnover, which are regulated by constant cycles of mitochondrial fusion and fission. Further, recycling of mitochondria through mitophagy in CSCs is associated with maintenance of reactive oxygen species levels that dictate gene expression. The protein machinery that drives mitochondrial dynamics is surprisingly simple and may represent attractive new therapeutic avenues to target CSC metabolism and selectively eradicate tumor-generating cells to reduce the risks of metastasis and relapse for a variety of tumor types.
Collapse
Affiliation(s)
- Dane T Sessions
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - David F Kashatus
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA.
| |
Collapse
|
38
|
Zhang J, Rashmi R, Inkman M, Jayachandran K, Ruiz F, Waters MR, Grigsby PW, Markovina S, Schwarz JK. Integrating imaging and RNA-seq improves outcome prediction in cervical cancer. J Clin Invest 2021; 131:139232. [PMID: 33645544 DOI: 10.1172/jci139232] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 12/22/2020] [Indexed: 11/17/2022] Open
Abstract
Approaches using a single type of data have been applied to classify human tumors. Here we integrate imaging features and transcriptomic data using a prospectively collected tumor bank. We demonstrate that increased maximum standardized uptake value on pretreatment 18F-fluorodeoxyglucose-positron emission tomography correlates with epithelial-to-mesenchymal transition (EMT) gene expression. We derived and validated 3 major molecular groups, namely squamous epithelial, squamous mesenchymal, and adenocarcinoma, using prospectively collected institutional (n = 67) and publicly available (n = 304) data sets. Patients with tumors of the squamous mesenchymal subtype showed inferior survival outcomes compared with the other 2 molecular groups. High mesenchymal gene expression in cervical cancer cells positively correlated with the capacity to form spheroids and with resistance to radiation. CaSki organoids were radiation-resistant but sensitive to the glycolysis inhibitor, 2-DG. These experiments provide a strategy for response prediction by integrating large data sets, and highlight the potential for metabolic therapy to influence EMT phenotypes in cervical cancer.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Radiation Oncology.,Institute for Informatics.,Siteman Cancer Center, and
| | | | | | | | | | | | | | | | - Julie K Schwarz
- Department of Radiation Oncology.,Siteman Cancer Center, and.,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
39
|
Ho KH, Huang TW, Shih CM, Lee YT, Liu AJ, Chen PH, Chen KC. Glycolysis-associated lncRNAs identify a subgroup of cancer patients with poor prognoses and a high-infiltration immune microenvironment. BMC Med 2021; 19:59. [PMID: 33627136 PMCID: PMC7905662 DOI: 10.1186/s12916-021-01925-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Long noncoding (lnc)RNAs and glycolysis are both recognized as key regulators of cancers. Some lncRNAs are also reportedly involved in regulating glycolysis metabolism. However, glycolysis-associated lncRNA signatures and their clinical relevance in cancers remain unclear. We investigated the roles of glycolysis-associated lncRNAs in cancers. METHODS Glycolysis scores and glycolysis-associated lncRNA signatures were established using a single-sample gene set enrichment analysis (GSEA) of The Cancer Genome Atlas pan-cancer data. Consensus clustering assays and genomic classifiers were used to stratify patient subtypes and for validation. Fisher's exact test was performed to investigate genomic mutations and molecular subtypes. A differentially expressed gene analysis, with GSEA, transcription factor (TF) activity scoring, cellular distributions, and immune cell infiltration, was conducted to explore the functions of glycolysis-associated lncRNAs. RESULTS Glycolysis-associated lncRNA signatures across 33 cancer types were generated and used to stratify patients into distinct clusters. Patients in cluster 3 had high glycolysis scores and poor survival, especially in bladder carcinoma, low-grade gliomas, mesotheliomas, pancreatic adenocarcinomas, and uveal melanomas. The clinical significance of lncRNA-defined groups was validated using external datasets and genomic classifiers. Gene mutations, molecular subtypes associated with poor prognoses, TFs, oncogenic signaling such as the epithelial-to-mesenchymal transition (EMT), and high immune cell infiltration demonstrated significant associations with cluster 3 patients. Furthermore, five lncRNAs, namely MIR4435-2HG, AC078846.1, AL157392.3, AP001273.1, and RAD51-AS1, exhibited significant correlations with glycolysis across the five cancers. Except MIR4435-2HG, the lncRNAs were distributed in nuclei. MIR4435-2HG was connected to glycolysis, EMT, and immune infiltrations in cancers. CONCLUSIONS We identified a subgroup of cancer patients stratified by glycolysis-associated lncRNAs with poor prognoses, high immune infiltration, and EMT activation, thus providing new directions for cancer therapy.
Collapse
Affiliation(s)
- Kuo-Hao Ho
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Wen Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chwen-Ming Shih
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Ting Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ann-Jeng Liu
- Department of Neurosurgery, Taipei City Hospital Ren-Ai Branch, Taipei, Taiwan
| | - Peng-Hsu Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ku-Chung Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan. .,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
40
|
Shuvalov O, Daks A, Fedorova O, Petukhov A, Barlev N. Linking Metabolic Reprogramming, Plasticity and Tumor Progression. Cancers (Basel) 2021; 13:cancers13040762. [PMID: 33673109 PMCID: PMC7917602 DOI: 10.3390/cancers13040762] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/03/2021] [Accepted: 02/07/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In the present review, we discuss the role of metabolic reprogramming which occurs in malignant cells. The process of metabolic reprogramming is also known as one of the “hallmarks of cancer”. Due to several reasons, including the origin of cancer, tumor microenvironment, and the tumor progression stage, metabolic reprogramming can be heterogeneous and dynamic. In this review, we provide evidence that the usage of metabolic drugs is a promising approach to treat cancer. However, because these drugs can damage not only malignant cells but also normal rapidly dividing cells, it is important to understand the exact metabolic changes which are elicited by particular drivers in concrete tissue and are specific for each stage of cancer development, including metastases. Finally, the review highlights new promising targets for the development of new metabolic drugs. Abstract The specific molecular features of cancer cells that distinguish them from the normal ones are denoted as “hallmarks of cancer”. One of the critical hallmarks of cancer is an altered metabolism which provides tumor cells with energy and structural resources necessary for rapid proliferation. The key feature of a cancer-reprogrammed metabolism is its plasticity, allowing cancer cells to better adapt to various conditions and to oppose different therapies. Furthermore, the alterations of metabolic pathways in malignant cells are heterogeneous and are defined by several factors including the tissue of origin, driving mutations, and microenvironment. In the present review, we discuss the key features of metabolic reprogramming and plasticity associated with different stages of tumor, from primary tumors to metastases. We also provide evidence of the successful usage of metabolic drugs in anticancer therapy. Finally, we highlight new promising targets for the development of new metabolic drugs.
Collapse
Affiliation(s)
- Oleg Shuvalov
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
| | - Alexandra Daks
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
| | - Olga Fedorova
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
| | - Alexey Petukhov
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
- Almazov National Medical Research Center, 197341 St-Petersburg, Russia
| | - Nickolai Barlev
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
- MIPT, 141701 Dolgoprudny, Moscow Region, Russia
- Orekhovich IBMC, 119435 Moscow, Russia
- Correspondence: ; Tel.: +7-812-297-4519
| |
Collapse
|
41
|
Srivastava SP, Kanasaki K, Goodwin JE. Loss of Mitochondrial Control Impacts Renal Health. Front Pharmacol 2020; 11:543973. [PMID: 33362536 PMCID: PMC7756079 DOI: 10.3389/fphar.2020.543973] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
Disruption of mitochondrial biosynthesis or dynamics, or loss of control over mitochondrial regulation leads to a significant alteration in fuel preference and metabolic shifts that potentially affect the health of kidney cells. Mitochondria regulate metabolic networks which affect multiple cellular processes. Indeed, mitochondria have established themselves as therapeutic targets in several diseases. The importance of mitochondria in regulating the pathogenesis of several diseases has been recognized, however, there is limited understanding of mitochondrial biology in the kidney. This review provides an overview of mitochondrial dysfunction in kidney diseases. We describe the importance of mitochondria and mitochondrial sirtuins in the regulation of renal metabolic shifts in diverse cells types, and review this loss of control leads to increased cell-to-cell transdifferentiation processes and myofibroblast-metabolic shifts, which affect the pathophysiology of several kidney diseases. In addition, we examine mitochondrial-targeted therapeutic agents that offer potential leads in combating kidney diseases.
Collapse
Affiliation(s)
- Swayam Prakash Srivastava
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| | - Keizo Kanasaki
- Internal Medicine 1, Shimane University Faculty of Medicine, Izumo, Japan
| | - Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
42
|
Higuchi C, Kuriyama J, Sakura H. Effect of lactate as a peritoneal dialysis fluid buffer on rat peritoneal mesothelial cells. RENAL REPLACEMENT THERAPY 2020. [DOI: 10.1186/s41100-020-00306-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Neutral, low-glucose degradation product (GDP) peritoneal dialysis fluid (PDF) is less damaging to the peritoneum than conventional PDF but is still insufficient for biocompatibility. One remaining issue is the problem of buffering.
Methods
Using cultured rat peritoneal mesothelial cells (PMCs), the present study examined the difference between the effects of neutral low-GDP lactate PDF and neutral low-GDP bicarbonate/lactate PDF on cells. The effects of lactate stimulation on these cells were also examined.
Results
Lactate PDF enhanced mRNA expressions of α-smooth muscle actin (αSMA) and type 1 and type 3 collagens and lowered expression of e-cadherin mRNA in PMCs compared to bicarbonate/lactate PDF. Lactate stimulation increased mRNA expressions of αSMA, matrix metalloproteinase 2 (MMP2), and basic fibroblast growth factor (bFGF) and suppressed e-cadherin mRNA expression. Transforming growth factor (TGF)-β1 and TGF-β2 and collagen type 1 and 3 mRNA expressions were also enhanced by lactate stimulation.
Conclusions
These results suggest that lactate as a PDF buffer may act on PMCs to promote epithelial-mesenchymal transition (EMT) and production of TGF-β, bFGF, and collagen.
Collapse
|
43
|
Sharma N, Bhushan A, He J, Kaushal G, Bhardwaj V. Metabolic plasticity imparts erlotinib-resistance in pancreatic cancer by upregulating glucose-6-phosphate dehydrogenase. Cancer Metab 2020; 8:19. [PMID: 32974013 PMCID: PMC7507640 DOI: 10.1186/s40170-020-00226-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 09/06/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant forms of cancer. Lack of effective treatment options and drug resistance contributes to the low survival among PDAC patients. In this study, we investigated the metabolic alterations in pancreatic cancer cells that do not respond to the EGFR inhibitor erlotinib. We selected erlotinib-resistant pancreatic cancer cells from MiaPaCa2 and AsPC1 cell lines. Metabolic profiling of erlotinib-resistant cells revealed a significant downregulation of glycolytic activity and reduced level of glycolytic metabolites compared to the sensitive cells. The resistant cells displayed elevated expression of the pentose phosphate pathway (PPP) enzymes involved in ROS regulation and nucleotide biosynthesis. The enhanced PPP elevated cellular NADPH/NADP+ ratio and protected the cells from reactive oxygen species (ROS)-induced damage. Inhibition of PPP using 6-aminonicotinamide (6AN) elevated ROS levels, induced G1 cell cycle arrest, and sensitized resistant cells to erlotinib. Genetic studies identified elevated PPP enzyme glucose-6-phosphate dehydrogenase (G6PD) as an important contributor to erlotinib resistance. Mechanistically, our data showed that upregulation of inhibitor of differentiation (ID1) regulates G6PD expression in resistant cells thus contributing to altered metabolic phenotype and reduced response to erlotinib. Together, our results highlight an underlying role of tumor metabolism in PDAC drug response and identify G6PD as a target to overcome drug resistance.
Collapse
Affiliation(s)
- Neha Sharma
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA USA
| | - Alok Bhushan
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA USA
| | - Jun He
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA
| | - Gagan Kaushal
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA USA
| | - Vikas Bhardwaj
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA USA
| |
Collapse
|
44
|
Metabolic Constrains Rule Metastasis Progression. Cells 2020; 9:cells9092081. [PMID: 32932943 PMCID: PMC7563739 DOI: 10.3390/cells9092081] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Metastasis formation accounts for the majority of tumor-associated deaths and consists of different steps, each of them being characterized by a distinctive adaptive phenotype of the cancer cells. Metabolic reprogramming represents one of the main adaptive phenotypes exploited by cancer cells during all the main steps of tumor and metastatic progression. In particular, the metabolism of cancer cells evolves profoundly through all the main phases of metastasis formation, namely the metastatic dissemination, the metastatic colonization of distant organs, the metastatic dormancy, and ultimately the outgrowth into macroscopic lesions. However, the metabolic reprogramming of metastasizing cancer cells has only recently become the subject of intense study. From a clinical point of view, the latter steps of the metastatic process are very important, because patients often undergo surgical removal of the primary tumor when cancer cells have already left the primary tumor site, even though distant metastases are not clinically detectable yet. In this scenario, to precisely elucidate if and how metabolic reprogramming drives acquisition of cancer-specific adaptive phenotypes might pave the way to new therapeutic strategies by combining chemotherapy with metabolic drugs for better cancer eradication. In this review we discuss the latest evidence that claim the importance of metabolic adaptation for cancer progression.
Collapse
|
45
|
Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance. Drug Resist Updat 2020; 53:100715. [PMID: 32679188 DOI: 10.1016/j.drup.2020.100715] [Citation(s) in RCA: 332] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 05/29/2020] [Accepted: 06/07/2020] [Indexed: 12/11/2022]
Abstract
It is well established that multifactorial drug resistance hinders successful cancer treatment. Tumor cell interactions with the tumor microenvironment (TME) are crucial in epithelial-mesenchymal transition (EMT) and multidrug resistance (MDR). TME-induced factors secreted by cancer cells and cancer-associated fibroblasts (CAFs) create an inflammatory microenvironment by recruiting immune cells. CD11b+/Gr-1+ myeloid-derived suppressor cells (MDSCs) and inflammatory tumor associated macrophages (TAMs) are main immune cell types which further enhance chronic inflammation. Chronic inflammation nurtures tumor-initiating/cancer stem-like cells (CSCs), induces both EMT and MDR leading to tumor relapses. Pro-thrombotic microenvironment created by inflammatory cytokines and chemokines from TAMs, MDSCs and CAFs is also involved in EMT and MDR. MDSCs are the most common mediators of immunosuppression and are also involved in resistance to targeted therapies, e.g. BRAF inhibitors and oncolytic viruses-based therapies. Expansion of both cancer and stroma cells causes hypoxia by hypoxia-inducible transcription factors (e.g. HIF-1α) resulting in drug resistance. TME factors induce the expression of transcriptional EMT factors, MDR and metabolic adaptation of cancer cells. Promoters of several ATP-binding cassette (ABC) transporter genes contain binding sites for canonical EMT transcription factors, e.g. ZEB, TWIST and SNAIL. Changes in glycolysis, oxidative phosphorylation and autophagy during EMT also promote MDR. Conclusively, EMT signaling simultaneously increases MDR. Owing to the multifactorial nature of MDR, targeting one mechanism seems to be non-sufficient to overcome resistance. Targeting inflammatory processes by immune modulatory compounds such as mTOR inhibitors, demethylating agents, low-dosed histone deacetylase inhibitors may decrease MDR. Targeting EMT and metabolic adaptation by small molecular inhibitors might also reverse MDR. In this review, we summarize evidence for TME components as causative factors of EMT and anticancer drug resistance.
Collapse
|
46
|
Yamaguchi H, Taouk GM. A Potential Role of YAP/TAZ in the Interplay Between Metastasis and Metabolic Alterations. Front Oncol 2020; 10:928. [PMID: 32596154 PMCID: PMC7300268 DOI: 10.3389/fonc.2020.00928] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/12/2020] [Indexed: 12/14/2022] Open
Abstract
Yes-Associated Protein (YAP) and Transcriptional Co-activator with PDZ-binding Motif (TAZ) are the downstream effectors of the Hippo signaling pathway that play a crucial role in various aspects of cancer progression including metastasis. Metastasis is the multistep process of disseminating cancer cells in a body and responsible for the majority of cancer-related death. Emerging evidence has shown that cancer cells reprogram their metabolism to gain proliferation, invasion, migration, and anti-apoptotic abilities and adapt to various environment during metastasis. Moreover, it has increasingly been recognized that YAP/TAZ regulates cellular metabolism that is associated with the phenotypic changes, and recent studies suggest that the YAP/TAZ-mediated metabolic alterations contribute to metastasis. In this review, we will introduce the latest knowledge of YAP/TAZ regulation and function in cancer metastasis and metabolism, and discuss possible links between the YAP/TAZ-mediated metabolic reprogramming and metastasis.
Collapse
Affiliation(s)
- Hirohito Yamaguchi
- Cancer Research Center, College of Health and Life Sciences, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Ghina M Taouk
- Cancer Research Center, College of Health and Life Sciences, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
47
|
Hua W, Ten Dijke P, Kostidis S, Giera M, Hornsveld M. TGFβ-induced metabolic reprogramming during epithelial-to-mesenchymal transition in cancer. Cell Mol Life Sci 2020; 77:2103-2123. [PMID: 31822964 PMCID: PMC7256023 DOI: 10.1007/s00018-019-03398-6] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/10/2019] [Accepted: 11/26/2019] [Indexed: 12/15/2022]
Abstract
Metastasis is the most frequent cause of death in cancer patients. Epithelial-to-mesenchymal transition (EMT) is the process in which cells lose epithelial integrity and become motile, a critical step for cancer cell invasion, drug resistance and immune evasion. The transforming growth factor-β (TGFβ) signaling pathway is a major driver of EMT. Increasing evidence demonstrates that metabolic reprogramming is a hallmark of cancer and extensive metabolic changes are observed during EMT. The aim of this review is to summarize and interconnect recent findings that illustrate how changes in glycolysis, mitochondrial, lipid and choline metabolism coincide and functionally contribute to TGFβ-induced EMT. We describe TGFβ signaling is involved in stimulating both glycolysis and mitochondrial respiration. Interestingly, the subsequent metabolic consequences for the redox state and lipid metabolism in cancer cells are found to be in favor of EMT as well. Combined we illustrate that a better understanding of the mechanistic links between TGFβ signaling, cancer metabolism and EMT holds promising strategies for cancer therapy, some of which are already actively being explored in the clinic.
Collapse
Affiliation(s)
- Wan Hua
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
- National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands.
| | - Sarantos Kostidis
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Marten Hornsveld
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
48
|
Lin Q, He Y, Wang X, Zhang Y, Hu M, Guo W, He Y, Zhang T, Lai L, Sun Z, Yi Z, Liu M, Chen Y. Targeting Pyruvate Carboxylase by a Small Molecule Suppresses Breast Cancer Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903483. [PMID: 32382484 PMCID: PMC7201266 DOI: 10.1002/advs.201903483] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/14/2020] [Accepted: 02/24/2020] [Indexed: 05/29/2023]
Abstract
Rapid metabolism differentiates cancer cells from normal cells and relies on anaplerotic pathways. However, the mechanisms of anaplerosis-associated enzymes are rarely understood. The lack of potent and selective antimetabolism drugs restrains further clinical investigations. A small molecule ZY-444 ((N 4-((5-(4-(benzyloxy)phenyl)-2-thiophenyl)methyl)-N 2-isobutyl-2,4-pyrimidinediamine) is discovered to inhibit cancer cell proliferation specifically, having potent efficacies against tumor growth, metastasis, and recurrence. ZY-444 binds to cellular pyruvate carboxylase (PC), a key anaplerotic enzyme of the tricarboxylic acid cycle, and inactivates its catalytic activity. PC inhibition suppresses breast cancer growth and metastasis through inhibiting the Wnt/β-catenin/Snail signaling pathway. Lower PC expression in patient tumors is correlated with significant survival benefits. Comparative profiles of PC expression in cancer versus normal tissues implicate the tumor selectivity of ZY-444. Overall, ZY-444 holds promise therapeutically as an anti-cancer metabolism agent.
Collapse
Affiliation(s)
- Qingxiang Lin
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational MedicineShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
| | - Yuan He
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational MedicineShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
- Joint Center for Translational MedicineSouthern Medical University Affiliated Fengxian HospitalShanghai201499P. R. China
| | - Xue Wang
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational MedicineShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
| | - Yong Zhang
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational MedicineShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
| | - Meichun Hu
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational MedicineShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
| | - Weikai Guo
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational MedicineShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
| | - Yundong He
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational MedicineShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
| | - Tao Zhang
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational MedicineShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
| | - Li Lai
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational MedicineShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
| | - Zhenliang Sun
- Joint Center for Translational MedicineSouthern Medical University Affiliated Fengxian HospitalShanghai201499P. R. China
| | - Zhengfang Yi
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational MedicineShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
- Joint Center for Translational MedicineSouthern Medical University Affiliated Fengxian HospitalShanghai201499P. R. China
| | - Mingyao Liu
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational MedicineShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
| | - Yihua Chen
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational MedicineShanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
| |
Collapse
|
49
|
Liu X, Feng C, Wei G, Kong W, Meng H, Du Y, Li J. Mitofusin1 Is a Major Mediator in Glucose-Induced Epithelial-to-Mesenchymal Transition in Lung Adenocarcinoma Cells. Onco Targets Ther 2020; 13:3511-3523. [PMID: 32425551 PMCID: PMC7187943 DOI: 10.2147/ott.s238714] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/03/2020] [Indexed: 01/25/2023] Open
Abstract
Background Epithelial-to-mesenchymal transition (EMT) has been considered a latent mediator of diverse biological processes in cancer. However, the mechanisms involved in high glucose-associated EMT in lung adenocarcinoma (LAD) have not been fully clarified. In this study, we aimed to investigate whether mitofusin1 (MFN1) is involved in the EMT of LAD cells induced by glucose and to identify the molecular mechanism involved in this process. Materials and Methods The expression of specific proteins was analysed by Western blotting, immunohistochemistry, co-immunoprecipitation and immunofluorescence analysis. The proliferation, migration and invasion of cells were assessed by Cell Counting Kit-8, bromodeoxyuridine incorporation, wound-healing and transwell assays. Lung tissues of adjacent normal regions and lung tissues from patients with LAD and LAD combined with diabetes mellitus were collected to determine the expression and significance of MFN1. Results Here, we showed that the expression of MFN1 was increased in LAD tissues compared with adjacent normal tissues and expression was even higher in lung tissues from patients with LAD combined with diabetes. In the lung cancer cell line A549, increased cell proliferation, invasion and EMT induced by high glucose were inhibited by MFN1 silencing. Mechanistic studies demonstrated that inhibiting autophagy reversed the abnormal EMT triggered by high glucose conditions. In addition, our data provide novel evidence demonstrating that PTEN-induced kinase (Pink) is a potential regulator involved in MFN1-mediated cell autophagy, which eventually leads to high glucose-induced proliferation, invasion and EMT of A549 cells. Conclusion Taken together, our data show that MFN1 interacts with Pink to induce the autophagic process and that the abnormal occurrence of autophagy ultimately contributes to glucose-induced pathological EMT in LAD.
Collapse
Affiliation(s)
- Xingyuan Liu
- Pathology Department, College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| | - Chuang Feng
- Science and Technology Department, Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| | - Guohua Wei
- Pathology Department, College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| | - Wencong Kong
- Pathology Department, College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| | - Hai Meng
- Clinicopathological Center, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| | - Yaqin Du
- Clinicopathological Center, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| | - Jingyuan Li
- Faculty of Pharmaceutical Sciences, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| |
Collapse
|
50
|
Osumi H, Horiguchi H, Kadomatsu T, Tashiro K, Morinaga J, Takahashi T, Ikeda K, Ito T, Suzuki M, Endo M, Oike Y. Tumor cell-derived angiopoietin-like protein 2 establishes a preference for glycolytic metabolism in lung cancer cells. Cancer Sci 2020; 111:1241-1253. [PMID: 32012400 PMCID: PMC7156862 DOI: 10.1111/cas.14337] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 01/17/2020] [Accepted: 01/22/2020] [Indexed: 01/03/2023] Open
Abstract
We previously revealed that tumor cell‐derived angiopoietin‐like protein 2 (ANGPTL2) accelerates the metastatic capacity of tumors in an autocrine/paracrine manner by activating tumor cell motility and invasiveness and the epithelial‐mesenchymal transition. However, the effects of ANGPTL2 on cancer cell glycolytic metabolism, which is a hallmark of tumor cells, are unknown. Here we report evidence supporting a role for tumor cell‐derived ANGPTL2 in establishing a preference for glycolytic metabolism. We report that a highly metastatic lung cancer cell subline expressing abundant ANGPTL2 showed upregulated expression of the glucose transporter GLUT3 as well as enhanced glycolytic metabolism relative to a less metastatic parental line. Most notably, ANGPTL2 overexpression in the less metastatic line activated glycolytic metabolism by increasing GLUT3 expression. Moreover, ANGPTL2 signaling through integrin α5β1 increased GLUT3 expression by increasing transforming growth factor‐β (TGF‐β) signaling and expression of the downstream transcription factor zinc finger E‐box binding homeobox 1 (ZEB1). Conversely, ANGPTL2 knockdown in the highly metastatic subline decreased TGF‐β1, ZEB1, and GLUT3 expression and antagonized glycolytic metabolism. In primary tumor cells from patients with lung cancer, ANGPTL2 expression levels correlated with GLUT3 expression. Overall, this work suggests that tumor cell‐derived ANGPTL2 accelerates activities associated with glycolytic metabolism in lung cancer cells by activating TGF‐β‐ZEB1‐GLUT3 signaling.
Collapse
Affiliation(s)
- Hironobu Osumi
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.,Department of Thoracic Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Haruki Horiguchi
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.,Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kyosei Tashiro
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Jun Morinaga
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | | | - Koei Ikeda
- Department of Thoracic Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Takaaki Ito
- Department of Pathology and Experimental Medicine, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Makoto Suzuki
- Department of Thoracic Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Motoyoshi Endo
- Department of Molecular Biology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|