201
|
Xing Y, Luo P, Hu R, Wang D, Zhou G, Jiang J. TRIB3 Promotes Lung Adenocarcinoma Progression via an Enhanced Warburg Effect. Cancer Manag Res 2020; 12:13195-13206. [PMID: 33380827 PMCID: PMC7767749 DOI: 10.2147/cmar.s287956] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/10/2020] [Indexed: 02/05/2023] Open
Abstract
Background The pseudokinase Tribbles 3 (TRIB3) is involved in many cellular processes and various cancers. In recent years, the importance of metabolic transformation in the maintenance of malignant tumors has become increasingly prominent. Abnormal metabolism of cancer cells is considered a hallmark of cancer. However, the exact role and molecular mechanism of TRIB3 in lung adenocarcinoma (LUAD) cell reprogramming is largely unknown. Methods The oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) of cells were examined with a Seahorse XF Extracellular Flux Analyzer. In vitro and in vivo RT-qPCR, Western blotting, and functional assays were performed to explore the functional roles of TRIB3 in LUAD. Results In the present study, we demonstrated that TRIB3 is remarkably upregulated in LUAD cell lines as well as tissues. TRIB3 knockdown significantly inhibited LUAD cell growth and suppressed LUAD cell invasion, while TRIB3 overexpression conferred the opposite effects. Moreover, silencing TRIB3 suppressed the tumorigenesis and metastatic ability of LUAD cells. Mechanistically, we demonstrated that silencing TRIB3 significantly impaired aerobic glycolysis ability in LUAD cells. Furthermore, our data indicated that TRIB3 knockdown decreased hypoxia-inducible factor (HIF)1α levels and targeted the glycolytic genes regulated by HIF1α. Conclusion Together, our findings revealed a previously unappreciated function of TRIB3 in cancer cell metabolism and tumor progression, illustrating that TRIB3 could be considered a valuable therapeutic target for LUAD patients.
Collapse
Affiliation(s)
- Yutong Xing
- Department of Cardiothoracic Surgery, The Fifth Hospital of Xiamen, Xiamen, People's Republic of China.,Department of Cardiothoracic Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, People's Republic of China
| | - Peng Luo
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, People's Republic of China
| | - Rui Hu
- Department of Cardiothoracic Surgery, The Fifth Hospital of Xiamen, Xiamen, People's Republic of China
| | - Duanduan Wang
- Department of Cardiothoracic Surgery, The Fifth Hospital of Xiamen, Xiamen, People's Republic of China
| | - Gang Zhou
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, People's Republic of China
| | - Jie Jiang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| |
Collapse
|
202
|
Chaudhary A, Bag S, Arora N, Radhakrishnan VS, Mishra D, Mukherjee G. Hypoxic Transformation of Immune Cell Metabolism Within the Microenvironment of Oral Cancers. FRONTIERS IN ORAL HEALTH 2020; 1:585710. [PMID: 35047983 PMCID: PMC8757756 DOI: 10.3389/froh.2020.585710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/13/2020] [Indexed: 01/01/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) includes tumors of the lips, tongue, gingivobuccal complex, and floor of the mouth. Prognosis for OSCC is highly heterogeneous, with overall 5-year survival of ~50%, but median survival of just 8-10 months for patients with locoregional recurrence or metastatic disease. A key feature of OSCC is microenvironmental oxygen depletion due to rapid growth of constituent tumor cells, which triggers hypoxia-associated signaling events and metabolic adaptations that influence subsequent tumor progression. Better understanding of leukocyte responses to tissue hypoxia and onco-metabolite expression under low-oxygen conditions will therefore be essential to develop more effective methods of diagnosing and treating patients with OSCC. This review assesses recent literature on metabolic reprogramming, redox homeostasis, and associated signaling pathways that mediate crosstalk of OSCC with immune cells in the hypoxic tumor microenvironment. The likely functional consequences of this metabolic interface between oxygen-starved OSCC and infiltrating leukocytes are also discussed. The hypoxic microenvironment of OSCC modifies redox signaling and alters the metabolic profile of tumor-infiltrating immune cells. Improved understanding of heterotypic interactions between host leukocytes, tumor cells, and hypoxia-induced onco-metabolites will inform the development of novel theranostic strategies for OSCC.
Collapse
Affiliation(s)
- Amrita Chaudhary
- Department of Histopathology, Tata Medical Center, Kolkata, India
| | - Swarnendu Bag
- Department of Histopathology, Tata Medical Center, Kolkata, India
| | - Neeraj Arora
- Department of Laboratory Hematology and Molecular Genetics, Tata Medical Center, Kolkata, India
| | | | - Deepak Mishra
- Department of Laboratory Hematology and Molecular Genetics, Tata Medical Center, Kolkata, India
| | | |
Collapse
|
203
|
Zhu GX, Gao D, Shao ZZ, Chen L, Ding WJ, Yu QF. Wnt/β‑catenin signaling: Causes and treatment targets of drug resistance in colorectal cancer (Review). Mol Med Rep 2020; 23:105. [PMID: 33300082 PMCID: PMC7723170 DOI: 10.3892/mmr.2020.11744] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common malignant tumor in humans. Chemotherapy is used for the treatment of CRC. However, the effect of chemotherapy remains unsatisfactory due to drug resistance. Growing evidence has shown that the presence of highly metastatic tumor stem cells, regulation of non-coding RNAs and the tumor microenvironment contributes to drug resistance mechanisms in CRC. Wnt/β-catenin signaling mediates the chemoresistance of CRC in these three aspects. Therefore, the present study analyzed the abundant evidence of the contribution of Wnt/β-catenin signaling to the development of drug resistance in CRC and discussed its possible role in improving the chemosensitivity of CRC, which may provide guidelines for its clinical treatment.
Collapse
Affiliation(s)
- Gui-Xian Zhu
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Dian Gao
- Department of Pathogen Biology and Immunology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhao-Zhao Shao
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Li Chen
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wen-Jie Ding
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qiong-Fang Yu
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
204
|
Wang H, Ma Z, Cheng X, Tuo B, Liu X, Li T. Physiological and Pathophysiological Roles of Ion Transporter-Mediated Metabolism in the Thyroid Gland and in Thyroid Cancer. Onco Targets Ther 2020; 13:12427-12441. [PMID: 33299328 PMCID: PMC7721308 DOI: 10.2147/ott.s280797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/17/2020] [Indexed: 12/21/2022] Open
Abstract
Thyroid cancer is the most common type of endocrine tumor and has shown an increasing annual incidence, especially among women. Patients with thyroid cancer have a good prognosis, with a high five-year survival rate; however, the recurrence rate and disease status of thyroid cancer remain a burden for patients, which compels us to further elucidate the pathogenesis of this disease. Recently, ion transporters have gradually become a hot topic in the field of thyroid gland biology and cancer research. Additionally, alterations in the metabolic state of tumor cells and protein molecules have gradually become the focus of scientific research. This review focuses on the progress in understanding the physiological and pathophysiological roles of ion transporter-mediated metabolism in both the thyroid gland and thyroid cancer. We also hope to shed light on new targets for the treatment and prognosis of thyroid cancer.
Collapse
Affiliation(s)
- Hu Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Zhiyuan Ma
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Xiaoming Cheng
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Digestive Disease Institute of Guizhou Province, Zunyi, People’s Republic of China
| | - Xuemei Liu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Digestive Disease Institute of Guizhou Province, Zunyi, People’s Republic of China
| | - Taolang Li
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| |
Collapse
|
205
|
Feldt M, Menard J, Rosendahl AH, Lettiero B, Bendahl PO, Belting M, Borgquist S. The effect of statin treatment on intratumoral cholesterol levels and LDL receptor expression: a window-of-opportunity breast cancer trial. Cancer Metab 2020; 8:25. [PMID: 33292612 PMCID: PMC7682108 DOI: 10.1186/s40170-020-00231-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 10/27/2020] [Indexed: 12/23/2022] Open
Abstract
Background Deregulated lipid metabolism is common in cancer cells and the mevalonate pathway, which synthesizes cholesterol, is central in lipid metabolism. This study aimed to assess statin-induced changes of the intratumoral levels of cholesterol and the expression of the low-density lipoprotein receptor (LDLR) to enhance our understanding of the role of the mevalonate pathway in cancer cholesterol metabolism. Methods This study is based on a phase II clinical trial designed as a window-of-opportunity trial including 50 breast cancer patients treated with 80 mg of atorvastatin/day for 2 weeks, between the time of diagnosis and breast surgery. Lipids were extracted from frozen tumor tissue sampled pre- and post-atorvastatin treatment. Intratumoral cholesterol levels were measured using a fluorometric quantitation assay. LDLR expression was evaluated by immunohistochemistry on formalin-fixed paraffin-embedded tumor tissue. Paired blood samples pre- and post-atorvastatin were analyzed for circulating low-density lipoprotein (LDL), high-density lipoprotein (HDL), apolipoprotein A1, and apolipoprotein B. In vitro experiments on MCF-7 breast cancer cells treated with atorvastatin were performed for comparison on the cellular level. Results In the trial, 42 patients completed all study parts. From the paired tumor tissue samples, assessment of the cholesterol levels was achievable for 14 tumors, and for the LDLR expression in 24 tumors. Following atorvastatin treatment, the expression of LDLR was significantly increased (P = 0.004), while the intratumoral levels of total cholesterol remained stable. A positive association between intratumoral cholesterol levels and tumor proliferation measured by Ki-67 expression was found. In agreement with the clinical findings, results from in vitro experiments showed no significant changes of the intracellular cholesterol levels after atorvastatin treatment while increased expression of the LDLR was found, although not reaching statistical significance. Conclusions This study shows an upregulation of LDLR and preserved intratumoral cholesterol levels in breast cancer patients treated with statins. Together with previous findings on the anti-proliferative effect of statins in breast cancer, the present data suggest a potential role for LDLR in the statin-induced regulation of breast cancer cell proliferation. Trial registration The study has been registered at ClinicalTrials.gov (i.e., ID number: NCT00816244, NIH), December 30, 2008. Supplementary Information The online version contains supplementary material available at 10.1186/s40170-020-00231-8.
Collapse
Affiliation(s)
- Maria Feldt
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden. .,Department of Oncology, Skåne University Hospital, Lund, Sweden.
| | - Julien Menard
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Ann H Rosendahl
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Barbara Lettiero
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Pär-Ola Bendahl
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Mattias Belting
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Oncology, Skåne University Hospital, Lund, Sweden.,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Signe Borgquist
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
206
|
Wang L, Wang H, Wu B, Zhang C, Yu H, Li X, Wang Q, Shi X, Fan C, Wang D, Luo J, Yang J. Long Noncoding RNA LINC00551 Suppresses Glycolysis and Tumor Progression by Regulating c-Myc-Mediated PKM2 Expression in Lung Adenocarcinoma. Onco Targets Ther 2020; 13:11459-11470. [PMID: 33204101 PMCID: PMC7665500 DOI: 10.2147/ott.s273797] [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: 08/19/2020] [Accepted: 10/07/2020] [Indexed: 01/26/2023] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a leading cause of mortality associated with cancer globally. Thus, it is essential to elucidate its tumorigenesis and prognosis. Accumulating evidence shows that long noncoding RNAs (lncRNAs) play important roles in the occurrence and progression of tumors by regulating their glucose metabolism. Methods Bioinformatics analysis was performed to explore the expression of LINC00551 in LUAD. The level of LINC00551 in LUAD cells and tissues was detected by RT-qPCR. CCK-8, colony formation, EDU and transwell assays were conducted to evaluate the cell growth and migration of LUAD cells (A549 and PC9). High throughput sequencing was used to discover the downstream genes of LINC00551. The metabolic function of LUAD cells was identified by glucose uptake and lactate production assays. Furthermore, tumor xenografts were established to investigate the effects of LINC00551 on tumor growth in vivo. Results Herein, we found that LINC00551 was low-expressed in LUAD, and its level correlated with clinical prognosis. Ectopic expression of LINC00551 inhibited the proliferation and migration of LUAD cells (A549 and PC9). High throughput sequencing and gene enrichment analysis revealed that LINC0551 may be involved in metabolic pathway. Glucose uptake and lactate production assays suggested that LINC00551 suppressed glycolysis of LUAD cells. Mechanistically, our work revealed that LINC00551 inhibited glycolysis in LUAD cells by impairing c-Myc-mediated transcription of an important glycolysis-related enzyme PKM2. Conclusion In summary, our study identifies LINC00551 as a tumor suppressor in LUAD and implicates the LINC00551/c-Myc/PKM2 axis in the glycolytic remodeling of LUAD.
Collapse
Affiliation(s)
- Li Wang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Huishan Wang
- Department of Gastroenterology, Shanghai Songjiang District Central Hospital, Shanghai, People's Republic of China
| | - Bining Wu
- Respiratory Department, Nanjing Yuhua Hospital, Yuhua Branch of Nanjing First Hospital, Nanjing, People's Republic of China
| | - Chun Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Hualin Yu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Xueyan Li
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Qinjue Wang
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiaoli Shi
- Department of Hepatology Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Chengfeng Fan
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Dayu Wang
- Department of Obstetrics and Gynecology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Jing Luo
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Jinsong Yang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
207
|
Li Y, Sun XX, Qian DZ, Dai MS. Molecular Crosstalk Between MYC and HIF in Cancer. Front Cell Dev Biol 2020; 8:590576. [PMID: 33251216 PMCID: PMC7676913 DOI: 10.3389/fcell.2020.590576] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 10/21/2020] [Indexed: 12/26/2022] Open
Abstract
The transcription factor c-MYC (MYC thereafter) is a global regulator of gene expression. It is overexpressed or deregulated in human cancers of diverse origins and plays a key role in the development of cancers. Hypoxia-inducible factors (HIFs), a central regulator for cells to adapt to low cellular oxygen levels, is also often overexpressed and activated in many human cancers. HIF mediates the primary transcriptional response of a wide range of genes in response to hypoxia. Earlier studies focused on the inhibition of MYC by HIF during hypoxia, when MYC is expressed at physiological level, to help cells survive under low oxygen conditions. Emerging evidence suggests that MYC and HIF also cooperate to promote cancer cell growth and progression. This review will summarize the current understanding of the complex molecular interplay between MYC and HIF.
Collapse
Affiliation(s)
- Yanping Li
- Department of Molecular and Medical Genetics, School of Medicine, Portland, OR, United States
| | - Xiao-Xin Sun
- Department of Molecular and Medical Genetics, School of Medicine, Portland, OR, United States
| | - David Z Qian
- The OHSU Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| | - Mu-Shui Dai
- Department of Molecular and Medical Genetics, School of Medicine, Portland, OR, United States.,The OHSU Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
208
|
Lactate in the Tumor Microenvironment: An Essential Molecule in Cancer Progression and Treatment. Cancers (Basel) 2020; 12:cancers12113244. [PMID: 33153193 PMCID: PMC7693872 DOI: 10.3390/cancers12113244] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/16/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The role of lactate in cancer described by Otto Warburg in 1927 states that cancer cells uptake high amount of glucose with a marked increase in lactate production, this is known as the “Warburg effect”. Since then lactate turn out to be a major signaling molecule in cancer progression. Its release from tumor cells is accompanied by acidification ranging from 6.3 to 6.9 in the tumor microenvironment (TME) which favors processes such as tumor promotion, angiogenesis, metastasis, tumor resistance and more importantly, immunosuppression which has been associated with a poor outcome. The goal of this review is to examine and discuss in deep detail the recent studies that address the role of lactate in all these cancerous processes. Lastly, we explore the efforts to target the lactate production and its transport as a promising approach for cancer therapeutics. Abstract Cancer is a complex disease that includes the reprogramming of metabolic pathways by malignant proliferating cells, including those affecting the tumor microenvironment (TME). The “TME concept” was introduced in recognition of the roles played by factors other than tumor cells in cancer progression. In response to the hypoxic or semi-hypoxic characteristic of the TME, cancer cells generate a large amount of lactate via the metabolism of glucose and glutamine. Export of this newly generated lactate by the tumor cells together with H+ prevents intracellular acidification but acidifies the TME. In recent years, the importance of lactate and acidosis in carcinogenesis has gained increasing attention, including the role of lactate as a tumor-promoting metabolite. Here we review the existing literature on lactate metabolism in tumor cells and the ability of extracellular lactate to direct the metabolic reprogramming of those cells. Studies demonstrating the roles of lactate in biological processes that drive or sustain carcinogenesis (tumor promotion, angiogenesis, metastasis and tumor resistance) and lactate’s role as an immunosuppressor that contributes to tumor evasion are also considered. Finally, we consider recent therapeutic efforts using available drugs directed at and interfering with lactate production and transport in cancer treatment.
Collapse
|
209
|
Rezinciuc S, Bezavada L, Bahadoran A, Duan S, Wang R, Lopez-Ferrer D, Finkelstein D, McGargill MA, Green DR, Pasa-Tolic L, Smallwood HS. Dynamic metabolic reprogramming in dendritic cells: An early response to influenza infection that is essential for effector function. PLoS Pathog 2020; 16:e1008957. [PMID: 33104753 PMCID: PMC7707590 DOI: 10.1371/journal.ppat.1008957] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/01/2020] [Accepted: 09/03/2020] [Indexed: 01/19/2023] Open
Abstract
Infection with the influenza virus triggers an innate immune response that initiates the adaptive response to halt viral replication and spread. However, the metabolic response fueling the molecular mechanisms underlying changes in innate immune cell homeostasis remain undefined. Although influenza increases parasitized cell metabolism, it does not productively replicate in dendritic cells. To dissect these mechanisms, we compared the metabolism of dendritic cells to that of those infected with active and inactive influenza A virus and those treated with toll-like receptor agonists. Using quantitative mass spectrometry, pulse chase substrate utilization assays and metabolic flux measurements, we found global metabolic changes in dendritic cells 17 hours post infection, including significant changes in carbon commitment via glycolysis and glutaminolysis, as well as mitochondrial respiration. Influenza infection of dendritic cells led to a metabolic phenotype distinct from that induced by TLR agonists, with significant resilience in terms of metabolic plasticity. We identified c-Myc as one transcription factor modulating this response. Restriction of c-Myc activity or mitochondrial substrates significantly changed the immune functions of dendritic cells, such as reducing motility and T cell activation. Transcriptome analysis of inflammatory dendritic cells isolated following influenza infection showed similar metabolic reprogramming occurs in vivo. Thus, early in the infection process, dendritic cells respond with global metabolic restructuring, that is present in inflammatory lung dendritic cells after infection, and this is important for effector function. These findings suggest metabolic switching in dendritic cells plays a vital role in initiating the immune response to influenza infection. Dendritic cells are critical in mounting an effective immune response to influenza infection by initiating the immune response to influenza and activating the adaptive response to mediate viral clearance and manifest immune memory for protection against subsequent infections. We found dendritic cells undergo a profound metabolic shift after infection. They alter the concentration and location of hundreds of proteins, including c-Myc, facilitating a shift to a highly glycolytic phenotype that is also flexible in terms of fueling respiration. Nonetheless, we found limiting access to specific metabolic pathways or substrates diminished key immune functions. We previously described an immediate, fixed hypermetabolic state in infected respiratory epithelial cells. Here we present data indicating the metabolic response of dendritic cells is increased yet flexible, distinct from what we previously showed for epithelial cells. Additionally, we demonstrate dendritic cells tailor their metabolic response to the pathogen or TLR stimulus. This metabolic reprogramming occurs rapidly in vitro and is sustained in inflammatory dendritic cells in vivo for at least 9 days following influenza infection. These studies introduce the possibility of modulating the immune response to viral infection using customized metabolic therapy to enhance or diminish the function of specific cells.
Collapse
Affiliation(s)
- Svetlana Rezinciuc
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Lavanya Bezavada
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Azadeh Bahadoran
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Susu Duan
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Disease, The Research Institute at Nationwide Children's Hospital, The Ohio State University School of Medicine, Columbus, Ohio, United States of America
| | - Daniel Lopez-Ferrer
- Chromatography and Mass Spectrometry Division, Thermo Fisher Scientific, CA, United States of America
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Maureen A. McGargill
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Douglas R. Green
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Ljiljana Pasa-Tolic
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington, United States of America
| | - Heather S. Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
210
|
de Jonge AV, Mutis T, Roemer MGM, Scheijen B, Chamuleau MED. Impact of MYC on Anti-Tumor Immune Responses in Aggressive B Cell Non-Hodgkin Lymphomas: Consequences for Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12103052. [PMID: 33092116 PMCID: PMC7589056 DOI: 10.3390/cancers12103052] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 02/08/2023] Open
Abstract
Simple Summary The human immune system has several mechanisms to attack and eliminate lymphomas. However, the MYC oncogene is thought to facilitate escape from this anti-tumor immune response. Since patients with MYC overexpressing lymphomas face a significant dismal prognosis after treatment with standard immunochemotherapy, understanding the role of MYC in regulating the anti-tumor immune response is highly relevant. In this review, we describe the mechanisms by which MYC attenuates the anti-tumor immune responses in B cell non-Hodgkin lymphomas. We aim to implement this knowledge in the deployment of novel immunotherapeutic approaches. Therefore, we also provide a comprehensive overview of current immunotherapeutic options and we discuss potential future treatment strategies for MYC overexpressing lymphomas. Abstract Patients with MYC overexpressing high grade B cell lymphoma (HGBL) face significant dismal prognosis after treatment with standard immunochemotherapy regimens. Recent preclinical studies indicate that MYC not only contributes to tumorigenesis by its effects on cell proliferation and differentiation, but also plays an important role in promoting escape from anti-tumor immune responses. This is of specific interest, since reversing tumor immune inhibition with immunotherapy has shown promising results in the treatment of both solid tumors and hematological malignancies. In this review, we outline the current understanding of impaired immune responses in B cell lymphoid malignancies with MYC overexpression, with a particular emphasis on diffuse large B cell lymphoma. We also discuss clinical consequences of MYC overexpression in the treatment of HGBL with novel immunotherapeutic agents and potential future treatment strategies.
Collapse
Affiliation(s)
- A. Vera de Jonge
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, The Netherlands; (T.M.); (M.E.D.C.)
- Correspondence:
| | - Tuna Mutis
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, The Netherlands; (T.M.); (M.E.D.C.)
| | - Margaretha G. M. Roemer
- Department of Pathology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, The Netherlands;
| | - Blanca Scheijen
- Department of Pathology, Radboud UMC, Radboud Institute for Molecular Life Sciences, 6525GA Nijmegen, The Netherlands;
| | - Martine E. D. Chamuleau
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, The Netherlands; (T.M.); (M.E.D.C.)
| |
Collapse
|
211
|
Digging deeper through glucose metabolism and its regulators in cancer and metastasis. Life Sci 2020; 264:118603. [PMID: 33091446 DOI: 10.1016/j.lfs.2020.118603] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/04/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022]
Abstract
Glucose metabolism enzymes and transporters play major role in cancer development and metastasis. In this study, we discuss glucose metabolism, transporters, receptors, hormones, oncogenes and tumor suppressors which interact with glucose metabolism and we try to discuss their major role in cancer development and cancer metabolism. We try to highlight the. Metabolic changes in cancer and metastasis upregulation of glycolysis is observed in many primary and metastatic cancers and aerobic glycolysis is the most favorable mechanism for glucose metabolism in cancer cells, and it is a kind of evolutionary change. The question that is posed at this juncture is: Can we use aerobic glycolysis phenotype and enzymes beyond this mechanism in estimating cancer prognosis and metastasis? Lactate is a metabolite of glucose metabolism and it is a key player in cancer and metastasis in both normoxic and hypoxic condition so lactate dehydrogenase can be a good prognostic biomarker. Furthermore, monocarboxylic transporter which is the main lactate transporter can be good target in therapeutic studies. Glycolysis enzymes are valuable enzymes in cancer and metastasis diagnosis and can be used as therapeutic targets in cancer treatment. Designing a diagnostic and prognostic profile for cancer metastasis seems to be possible base on glycolysis enzymes and glucose transporters. Also, glucose metabolism enzymes and agents can give us a clear vision in estimating cancer metastasis. We can promote a panel of genes that detect genetic changes in glucose metabolism agents to diagnose cancer metastasis.
Collapse
|
212
|
Li J, Eu JQ, Kong LR, Wang L, Lim YC, Goh BC, Wong ALA. Targeting Metabolism in Cancer Cells and the Tumour Microenvironment for Cancer Therapy. Molecules 2020; 25:molecules25204831. [PMID: 33092283 PMCID: PMC7588013 DOI: 10.3390/molecules25204831] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022] Open
Abstract
Targeting altered tumour metabolism is an emerging therapeutic strategy for cancer treatment. The metabolic reprogramming that accompanies the development of malignancy creates targetable differences between cancer cells and normal cells, which may be exploited for therapy. There is also emerging evidence regarding the role of stromal components, creating an intricate metabolic network consisting of cancer cells, cancer-associated fibroblasts, endothelial cells, immune cells, and cancer stem cells. This metabolic rewiring and crosstalk with the tumour microenvironment play a key role in cell proliferation, metastasis, and the development of treatment resistance. In this review, we will discuss therapeutic opportunities, which arise from dysregulated metabolism and metabolic crosstalk, highlighting strategies that may aid in the precision targeting of altered tumour metabolism with a focus on combinatorial therapeutic strategies.
Collapse
Affiliation(s)
- Jiaqi Li
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK;
| | - Jie Qing Eu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
| | - Li Ren Kong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Yaw Chyn Lim
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Department of Pathology, National University Health System, Singapore 119074, Singapore
| | - Boon Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Department of Haematology-Oncology, National University Health System, Singapore 119228, Singapore
| | - Andrea L. A. Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Department of Haematology-Oncology, National University Health System, Singapore 119228, Singapore
- Correspondence: ; Tel.: +65-6779-5555
| |
Collapse
|
213
|
Zhang D, Xu X, Ye Q. Metabolism and immunity in breast cancer. Front Med 2020; 15:178-207. [PMID: 33074528 DOI: 10.1007/s11684-020-0793-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 04/17/2020] [Indexed: 12/12/2022]
Abstract
Breast cancer is one of the most common malignancies that seriously threaten women's health. In the process of the malignant transformation of breast cancer, metabolic reprogramming and immune evasion represent the two main fascinating characteristics of cancer and facilitate cancer cell proliferation. Breast cancer cells generate energy through increased glucose metabolism. Lipid metabolism contributes to biological signal pathways and forms cell membranes except energy generation. Amino acids act as basic protein units and metabolic regulators in supporting cell growth. For tumor-associated immunity, poor immunogenicity and heightened immunosuppression cause breast cancer cells to evade the host's immune system. For the past few years, the complex mechanisms of metabolic reprogramming and immune evasion are deeply investigated, and the genes involved in these processes are used as clinical therapeutic targets for breast cancer. Here, we review the recent findings related to abnormal metabolism and immune characteristics, regulatory mechanisms, their links, and relevant therapeutic strategies.
Collapse
Affiliation(s)
- Deyu Zhang
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, 100850, China
| | - Xiaojie Xu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, 100850, China.
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, 100850, China.
| |
Collapse
|
214
|
Cancer Metabolism: Phenotype, Signaling and Therapeutic Targets. Cells 2020; 9:cells9102308. [PMID: 33081387 PMCID: PMC7602974 DOI: 10.3390/cells9102308] [Citation(s) in RCA: 288] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/10/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
Aberrant metabolism is a major hallmark of cancer. Abnormal cancer metabolism, such as aerobic glycolysis and increased anabolic pathways, has important roles in tumorigenesis, metastasis, drug resistance, and cancer stem cells. Well-known oncogenic signaling pathways, such as phosphoinositide 3-kinase (PI3K)/AKT, Myc, and Hippo pathway, mediate metabolic gene expression and increase metabolic enzyme activities. Vice versa, deregulated metabolic pathways contribute to defects in cellular signal transduction pathways, which in turn provide energy, building blocks, and redox potentials for unrestrained cancer cell proliferation. Studies and clinical trials are being performed that focus on the inhibition of metabolic enzymes by small molecules or dietary interventions (e.g., fasting, calorie restriction, and intermittent fasting). Similar to genetic heterogeneity, the metabolic phenotypes of cancers are highly heterogeneous. This heterogeneity results from diverse cues in the tumor microenvironment and genetic mutations. Hence, overcoming metabolic plasticity is an important goal of modern cancer therapeutics. This review highlights recent findings on the metabolic phenotypes of cancer and elucidates the interactions between signal transduction pathways and metabolic pathways. We also provide novel rationales for designing the next-generation cancer metabolism drugs.
Collapse
|
215
|
Kamada S, Takeiwa T, Ikeda K, Horie-Inoue K, Inoue S. Long Non-coding RNAs Involved in Metabolic Alterations in Breast and Prostate Cancers. Front Oncol 2020; 10:593200. [PMID: 33123488 PMCID: PMC7573247 DOI: 10.3389/fonc.2020.593200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
Breast and prostate cancers are the most prevalent cancers in females and males, respectively. These cancers exhibit sex hormone dependence and thus, hormonal therapies are used to treat these cancers. However, acquired resistance to hormone therapies is a major clinical problem. In addition, certain portions of these cancers initially exhibit hormone-independence due to the absence of sex hormone receptors. Therefore, precise and profound understanding of the cancer pathophysiology is required to develop novel clinical strategies against breast and prostate cancers. Metabolic reprogramming is currently recognized as one of the hallmarks of cancer, as exemplified by the alteration of glucose metabolism, oxidative phosphorylation, and lipid metabolism. Dysregulation of metabolic enzymes and their regulators such as kinases, transcription factors, and other signaling molecules contributes to metabolic alteration in cancer. Moreover, accumulating lines of evidence reveal that long non-coding RNAs (lncRNAs) regulate cancer development and progression by modulating metabolism. Understanding the mechanism and function of lncRNAs associated with cancer-specific metabolic alteration will therefore provide new knowledge for cancer diagnosis and treatment. This review provides an overview of recent studies regarding the role of lncRNAs in metabolism in breast and prostate cancers, with a focus on both sex hormone-dependent and -independent pathways.
Collapse
Affiliation(s)
- Shuhei Kamada
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan.,Department of Urology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshihiko Takeiwa
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan
| | - Kazuhiro Ikeda
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan
| | - Kuniko Horie-Inoue
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan
| | - Satoshi Inoue
- Division of Systems Medicine and Gene Therapy, Saitama Medical University, Saitama, Japan.,Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
216
|
Brito A, Pereira PMR, Reis RL, Ulijn RV, Lewis JS, Pires RA, Pashkuleva I. Aromatic carbohydrate amphiphile disrupts cancer spheroids and prevents relapse. NANOSCALE 2020; 12:19088-19092. [PMID: 32955076 PMCID: PMC7541621 DOI: 10.1039/d0nr05008c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Spheroids recapitulate the organization, heterogeneity and microenvironment of solid tumors. Herein, we targeted spatiotemporally the accelerated metabolism of proliferative cells located on the spheroid surface that ensure structure maintenance and/or growth. We demonstrate that phosphorylated carbohydrate amphiphile acts as a potent antimetabolite due to glycolysis inhibition and to in situ formation of supramolecular net around spheroid surface where alkaline phosphatase is overexpressed. The efficiency of the treatment is higher in spheroids as compared to the conventional 2D cultures because of the 2-fold higher expression of glucose transporter 1 (GLUT1). Moreover, treated spheroids do not undergo following relapse.
Collapse
Affiliation(s)
- Alexandra Brito
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal and Department of Radiology, Memorial Sloan Kettering Cancer Center, USA and Advanced Science Research Center (ASRC) at the Graduate Center, City University of New York (CUNY), 85 St Nicholas Terrace, New York, New York 10031, USA
| | | | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rein V Ulijn
- Advanced Science Research Center (ASRC) at the Graduate Center, City University of New York (CUNY), 85 St Nicholas Terrace, New York, New York 10031, USA and Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, New York 10065, USA and PhD programs in Biochemistry and Chemistry, The Graduate Center of the City University of New York, New York 10016, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, USA and Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA and Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA and Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ricardo A Pires
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Iva Pashkuleva
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
217
|
Protein Translation Inhibition is Involved in the Activity of the Pan-PIM Kinase Inhibitor PIM447 in Combination with Pomalidomide-Dexamethasone in Multiple Myeloma. Cancers (Basel) 2020; 12:cancers12102743. [PMID: 32987735 PMCID: PMC7598606 DOI: 10.3390/cancers12102743] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Proviral Insertion site for Moloney murine leukemia virus (PIM) kinases are overexpressed in hematologic malignancies, including multiple myeloma. Previous preclinical data from our group demonstrated the anti-myeloma effect of the pan-PIM kinase inhibitor PIM447. METHODS Based on those data, we evaluate here, by in vitro and in vivo studies, the activity of the triple combination of PIM447 + pomalidomide + dexamethasone (PIM-Pd) in multiple myeloma. RESULTS Our results show that the PIM-Pd combination exerts a potent anti-myeloma effect in vitro and in vivo, where it markedly delays tumor growth and prolongs survival of treated mice. Mechanism of action studies performed in vitro and on mice tumor samples suggest that the combination PIM-Pd inhibits protein translation processes through the convergent inhibition of c-Myc and mTORC1, which subsequently disrupts the function of eIF4E. Interestingly the MM pro-survival factor IRF4 is also downregulated after PIM-Pd treatment. As a whole, all these molecular changes would promote cell cycle arrest and deregulation of metabolic pathways, including glycolysis and lipid biosynthesis, leading to inhibition of myeloma cell proliferation. CONCLUSIONS Altogether, our data support the clinical evaluation of the triple combination PIM-Pd for the treatment of patients with multiple myeloma.
Collapse
|
218
|
Tan KN, Avery VM, Carrasco-Pozo C. Metabolic Roles of Androgen Receptor and Tip60 in Androgen-Dependent Prostate Cancer. Int J Mol Sci 2020; 21:ijms21186622. [PMID: 32927797 PMCID: PMC7555377 DOI: 10.3390/ijms21186622] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 01/10/2023] Open
Abstract
Androgen receptor (AR)-mediated signaling is essential for the growth and differentiation of the normal prostate and is the primary target for androgen deprivation therapy in prostate cancer. Tat interactive protein 60 kDa (Tip60) is a histone acetyltransferase that is critical for AR activation. It is well known that cancer cells rewire their metabolic pathways in order to sustain aberrant proliferation. Growing evidence demonstrates that the AR and Tip60 modulate key metabolic processes to promote the survival of prostate cancer cells, in addition to their classical roles. AR activation enhances glucose metabolism, including glycolysis, tricarboxylic acid cycle and oxidative phosphorylation, as well as lipid metabolism in prostate cancer. The AR also interacts with other metabolic regulators, including calcium/calmodulin-dependent kinase kinase 2 and mammalian target of rapamycin. Several studies have revealed the roles of Tip60 in determining cell fate indirectly by modulating metabolic regulators, such as c-Myc, hypoxia inducible factor 1α (HIF-1α) and p53 in various cancer types. Furthermore, Tip60 has been shown to regulate the activity of key enzymes in gluconeogenesis and glycolysis directly through acetylation. Overall, both the AR and Tip60 are master metabolic regulators that mediate cellular energy metabolism in prostate cancer, providing a framework for the development of novel therapeutic targets in androgen-dependent prostate cancer.
Collapse
Affiliation(s)
- Kah Ni Tan
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia; (K.N.T.); (V.M.A.)
- CRC for Cancer Therapeutics, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| | - Vicky M. Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia; (K.N.T.); (V.M.A.)
- CRC for Cancer Therapeutics, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| | - Catalina Carrasco-Pozo
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia; (K.N.T.); (V.M.A.)
- CRC for Cancer Therapeutics, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
- Correspondence: ; Tel.: +617-3735-6034
| |
Collapse
|
219
|
Bozzetti F, Stanga Z. Does nutrition for cancer patients feed the tumour? A clinical perspective. Crit Rev Oncol Hematol 2020; 153:103061. [DOI: 10.1016/j.critrevonc.2020.103061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
|
220
|
Abstract
Through the successes of checkpoint blockade and adoptive cellular therapy, immunotherapy has become an established treatment modality for cancer. Cellular metabolism has emerged as a critical determinant of the viability and function of both cancer cells and immune cells. In order to sustain prodigious anabolic needs, tumours employ a specialized metabolism that differs from untransformed somatic cells. This metabolism leads to a tumour microenvironment that is commonly acidic, hypoxic and/or depleted of critical nutrients required by immune cells. In this context, tumour metabolism itself is a checkpoint that can limit immune-mediated tumour destruction. Because our understanding of immune cell metabolism and cancer metabolism has grown significantly in the past decade, we are on the cusp of being able to unravel the interaction of cancer cell metabolism and immune metabolism in therapeutically meaningful ways. Although there are metabolic processes that are seemingly fundamental to both cancer and responding immune cells, metabolic heterogeneity and plasticity may serve to distinguish the two. As such, understanding the differential metabolic requirements of the diverse cells that comprise an immune response to cancer offers an opportunity to selectively regulate immune cell function. Such a nuanced evaluation of cancer and immune metabolism can uncover metabolic vulnerabilities and therapeutic windows upon which to intervene for enhanced immunotherapy.
Collapse
Affiliation(s)
- Robert D Leone
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan D Powell
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
221
|
Tong Y, Gao WQ, Liu Y. Metabolic heterogeneity in cancer: An overview and therapeutic implications. Biochim Biophys Acta Rev Cancer 2020; 1874:188421. [PMID: 32835766 DOI: 10.1016/j.bbcan.2020.188421] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/19/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022]
Abstract
Recent research on cancer metabolism has revealed that individual tumors have highly heterogeneous metabolic profiles that contribute to the connective metabolic networks within the tumor and its environment. Indeed, tumor-associated cells types, including tumor cells, cancer-associated fibroblasts (CAFs) and immune cells, reprogram their metabolism in many different ways due to diverse genetic backgrounds and complex environmental stimuli. This intratumoral metabolic heterogeneity and the derived metabolic interactions play an instrumental role in cancer progression. Understanding how this heterogeneity occurs may provide promising therapeutic strategies. Here, we review the diverse metabolic profiles of several important cell subpopulations in tumors and their impact on tumor progression and discuss the consequent metabolic interactions as well as the related therapeutic concerns.
Collapse
Affiliation(s)
- Yu Tong
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
| | - Yanfeng Liu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
222
|
Mendes C, Serpa J. Revisiting lactate dynamics in cancer—a metabolic expertise or an alternative attempt to survive? J Mol Med (Berl) 2020; 98:1397-1414. [DOI: 10.1007/s00109-020-01965-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/14/2020] [Accepted: 08/14/2020] [Indexed: 12/15/2022]
|
223
|
Liu X, Feng S, Zhang XD, Li J, Zhang K, Wu M, Thorne RF. Non-coding RNAs, metabolic stress and adaptive mechanisms in cancer. Cancer Lett 2020; 491:60-69. [PMID: 32726612 DOI: 10.1016/j.canlet.2020.06.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/12/2020] [Accepted: 06/28/2020] [Indexed: 12/18/2022]
Abstract
Metabolic reprogramming in cancer describes the multifaceted alterations in metabolism that contribute to tumorigenesis. Major determinants of metabolic phenotypes are the changes in signalling pathways associated with oncogenic activation together with cues from the tumor microenvironment. Therein, depleted oxygen and nutrient levels elicit metabolic stress, requiring cancer cells to engage adaptive mechanisms. Non-coding RNAs (ncRNAs) act as regulatory elements within metabolic pathways and their widespread dysregulation in cancer contributes to altered metabolic phenotypes. Indeed, ncRNAs are the regulatory accomplices of many prominent effectors of metabolic reprogramming including c-MYC and HIFs that are activated by metabolic stress. By example, this review illustrates the range of ncRNAs mechanisms impacting these effectors throughout their DNA-RNA-protein lifecycle along with presenting the mechanistic roles of ncRNAs in adaptive responses to glucose, glutamine and lipid deprivation. We also discuss the facultative activation of metabolic enzymes by ncRNAs, a phenomenon which may reflect a broad but currently invisible level of metabolic regulation. Finally, the translational challenges associated with ncRNA discoveries are discussed, emphasizing the gaps in knowledge together with importance of understanding the molecular basis of ncRNA regulatory mechanisms.
Collapse
Affiliation(s)
- Xiaoying Liu
- Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Molecular Pathology Centre, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China; School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Shanshan Feng
- Key Laboratory of Regenerative Medicine, Ministry of Education, Department of Developmental & Regenerative Biology, School of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xu Dong Zhang
- Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Molecular Pathology Centre, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China; School of Biomedical Sciences & Pharmacy, University of Newcastle, Newcastle, NSW, Australia
| | - Jinming Li
- Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Molecular Pathology Centre, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China
| | - Kaiguang Zhang
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230027, China.
| | - Mian Wu
- Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Molecular Pathology Centre, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China; The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230027, China; Key Laboratory of Stem Cell Differentiation & Modification, School of Clinical Medicine, Henan University, Zhengzhou, China.
| | - Rick F Thorne
- Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Molecular Pathology Centre, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China; School of Environmental & Life Sciences, University of Newcastle, NSW, Australia.
| |
Collapse
|
224
|
Cheung KCP, Fanti S, Mauro C, Wang G, Nair AS, Fu H, Angeletti S, Spoto S, Fogolari M, Romano F, Aksentijevic D, Liu W, Li B, Cheng L, Jiang L, Vuononvirta J, Poobalasingam TR, Smith DM, Ciccozzi M, Solito E, Marelli-Berg FM. Preservation of microvascular barrier function requires CD31 receptor-induced metabolic reprogramming. Nat Commun 2020; 11:3595. [PMID: 32681081 PMCID: PMC7367815 DOI: 10.1038/s41467-020-17329-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/09/2020] [Indexed: 12/19/2022] Open
Abstract
Endothelial barrier (EB) breaching is a frequent event during inflammation, and it is followed by the rapid recovery of microvascular integrity. The molecular mechanisms of EB recovery are poorly understood. Triggering of MHC molecules by migrating T-cells is a minimal signal capable of inducing endothelial contraction and transient microvascular leakage. Using this model, we show that EB recovery requires a CD31 receptor-induced, robust glycolytic response sustaining junction re-annealing. Mechanistically, this response involves src-homology phosphatase activation leading to Akt-mediated nuclear exclusion of FoxO1 and concomitant β-catenin translocation to the nucleus, collectively leading to cMyc transcription. CD31 signals also sustain mitochondrial respiration, however this pathway does not contribute to junction remodeling. We further show that pathologic microvascular leakage in CD31-deficient mice can be corrected by enhancing the glycolytic flux via pharmacological Akt or AMPK activation, thus providing a molecular platform for the therapeutic control of EB response.
Collapse
Affiliation(s)
- Kenneth C P Cheung
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- School of Life Sciences, Centre for Cell & Developmental Biology and Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Silvia Fanti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Claudio Mauro
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Mindelson Way, Birmingham, B152WB, UK
| | - Guosu Wang
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Anitha S Nair
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Hongmei Fu
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Silvia Angeletti
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, Rome, Italy
| | - Silvia Spoto
- Internal Medicine Department, University campus Bio-Medico of Rome, Rome, Italy
| | - Marta Fogolari
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, Rome, Italy
| | - Francesco Romano
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, Rome, Italy
| | - Dunja Aksentijevic
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Weiwei Liu
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, 510060, People's Republic of China
| | - Baiying Li
- School of Life Sciences, Centre for Cell & Developmental Biology and Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lixin Cheng
- School of Life Sciences, Centre for Cell & Developmental Biology and Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Liwen Jiang
- School of Life Sciences, Centre for Cell & Developmental Biology and Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Juho Vuononvirta
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Thanushiyan R Poobalasingam
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - David M Smith
- AstraZeneca R&D, Cambridge Science Park, Milton Road, Cambridge, CB4 0WG, UK
| | - Massimo Ciccozzi
- Unit of Medical Statistic and Molecular Epidemiology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Egle Solito
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universita degli studi di Napoli "Federico II", 80131, Naples, Italy
| | - Federica M Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
- Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, Charterhouse Square, London, UK.
| |
Collapse
|
225
|
Dong Y, Tu R, Liu H, Qing G. Regulation of cancer cell metabolism: oncogenic MYC in the driver's seat. Signal Transduct Target Ther 2020; 5:124. [PMID: 32651356 PMCID: PMC7351732 DOI: 10.1038/s41392-020-00235-2] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/31/2022] Open
Abstract
Cancer cells must rewire cellular metabolism to satisfy the demands of unbridled growth and proliferation. As such, most human cancers differ from normal counterpart tissues by a plethora of energetic and metabolic reprogramming. Transcription factors of the MYC family are deregulated in up to 70% of all human cancers through a variety of mechanisms. Oncogenic levels of MYC regulates almost every aspect of cellular metabolism, a recently revisited hallmark of cancer development. Meanwhile, unrestrained growth in response to oncogenic MYC expression creates dependency on MYC-driven metabolic pathways, which in principle provides novel targets for development of effective cancer therapeutics. In the current review, we summarize the significant progress made toward understanding how MYC deregulation fuels metabolic rewiring in malignant transformation.
Collapse
Affiliation(s)
- Yang Dong
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Frontier Science Center for Immunology & Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Rongfu Tu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Frontier Science Center for Immunology & Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Hudan Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Frontier Science Center for Immunology & Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Guoliang Qing
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China. .,Frontier Science Center for Immunology & Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
226
|
Lafita-Navarro MC, Perez-Castro L, Zacharias LG, Barnes S, DeBerardinis RJ, Conacci-Sorrell M. The transcription factors aryl hydrocarbon receptor and MYC cooperate in the regulation of cellular metabolism. J Biol Chem 2020; 295:12398-12407. [PMID: 32611766 DOI: 10.1074/jbc.ac120.014189] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/26/2020] [Indexed: 12/31/2022] Open
Abstract
The transcription factor AHR (aryl hydrocarbon receptor) drives the expression of genes involved in detoxification pathways in cells exposed to pollutants and other small molecules. Moreover, AHR supports transcriptional programs that promote ribosome biogenesis and protein synthesis in cells stimulated to proliferate by the oncoprotein MYC. Thus, AHR is necessary for the proliferation of MYC-overexpressing cells. To define metabolic pathways in which AHR cooperates with MYC in supporting cell growth, here we used LC-MS-based metabolomics to examine the metabolome of MYC-expressing cells upon AHR knockdown. We found that AHR knockdown reduced lactate, S-lactoylglutathione, N-acetyl-l-alanine, 2-hydroxyglutarate, and UMP levels. Using our previously obtained RNA sequencing data, we found that AHR mediates the expression of the UMP-generating enzymes dihydroorotate dehydrogenase (quinone) (DHODH) and uridine monophosphate synthetase (UMPS), as well as lactate dehydrogenase A (LDHA), establishing a mechanism by which AHR regulates lactate and UMP production in MYC-overexpressing cells. AHR knockdown in glioblastoma cells also reduced the expression of LDHA (and lactate), DHODH, and UMPS but did not affect UMP levels, likely because of compensatory mechanisms in these cells. Our results indicate that AHR contributes to the regulation of metabolic pathways necessary for the proliferation of transformed cells.
Collapse
Affiliation(s)
- M Carmen Lafita-Navarro
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lizbeth Perez-Castro
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lauren G Zacharias
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Spencer Barnes
- Bioinformatics Core Facility, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Howard Hughes Medical Institute, Dallas, Texas, USA
| | - Maralice Conacci-Sorrell
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA .,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
227
|
The progress and development of GLUT1 inhibitors targeting cancer energy metabolism. Future Med Chem 2020; 11:2333-2352. [PMID: 31581916 DOI: 10.4155/fmc-2019-0052] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A major difference between glucose metabolism in cancer cells and normal cells is that glucose in cancer cells is preferably converted to lactate in aerobic conditions rather than oxidized in mitochondria. This process is called aerobic glycolysis, known as the 'Warburg effect'. In this review, we focus on the energy-metabolism characteristics between tumor and normal cells, analyzing the regulation mechanism of energy metabolism based on glycolysis, and summarizing two targets on the upstream proteins of glycolysis, including glucose transporter (GLUT) and hexokinase. In addition, we proposed the risks and limitations of GLUT1-based drug research and summarized the current research progress of representative drugs, including natural and synthetic GLUT1 inhibitors. This will provide guidance for designing and synthesizing small molecule drugs targeting GLUT1 in glycolysis.
Collapse
|
228
|
Zhu X, Chen HH, Gao CY, Zhang XX, Jiang JX, Zhang Y, Fang J, Zhao F, Chen ZG. Energy metabolism in cancer stem cells. World J Stem Cells 2020; 12:448-461. [PMID: 32742562 PMCID: PMC7360992 DOI: 10.4252/wjsc.v12.i6.448] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/09/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023] Open
Abstract
Normal cells mainly rely on oxidative phosphorylation as an effective energy source in the presence of oxygen. In contrast, most cancer cells use less efficient glycolysis to produce ATP and essential biomolecules. Cancer cells gain the characteristics of metabolic adaptation by reprogramming their metabolic mechanisms to meet the needs of rapid tumor growth. A subset of cancer cells with stem characteristics and the ability to regenerate exist throughout the tumor and are therefore called cancer stem cells (CSCs). New evidence indicates that CSCs have different metabolic phenotypes compared with differentiated cancer cells. CSCs can dynamically transform their metabolic state to favor glycolysis or oxidative metabolism. The mechanism of the metabolic plasticity of CSCs has not been fully elucidated, and existing evidence indicates that the metabolic phenotype of cancer cells is closely related to the tumor microenvironment. Targeting CSC metabolism may provide new and effective methods for the treatment of tumors. In this review, we summarize the metabolic characteristics of cancer cells and CSCs and the mechanisms of the metabolic interplay between the tumor microenvironment and CSCs, and discuss the clinical implications of targeting CSC metabolism.
Collapse
Affiliation(s)
- Xuan Zhu
- Department of Radiation Oncology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Hui-Hui Chen
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou 310000, Zhejiang Province, China
- Department of Breast Surgery, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Chen-Yi Gao
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
- Department of Breast Surgery, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Xin-Xin Zhang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
- Department of Breast Surgery, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Jing-Xin Jiang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
- Department of Breast Surgery, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Yi Zhang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
- Department of Breast Surgery, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Jun Fang
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Feng Zhao
- Department of Radiation Oncology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Zhi-Gang Chen
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
- Department of Breast Surgery, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China.
| |
Collapse
|
229
|
ERK signalling: a master regulator of cell behaviour, life and fate. Nat Rev Mol Cell Biol 2020; 21:607-632. [PMID: 32576977 DOI: 10.1038/s41580-020-0255-7] [Citation(s) in RCA: 648] [Impact Index Per Article: 129.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2020] [Indexed: 12/13/2022]
Abstract
The proteins extracellular signal-regulated kinase 1 (ERK1) and ERK2 are the downstream components of a phosphorelay pathway that conveys growth and mitogenic signals largely channelled by the small RAS GTPases. By phosphorylating widely diverse substrates, ERK proteins govern a variety of evolutionarily conserved cellular processes in metazoans, the dysregulation of which contributes to the cause of distinct human diseases. The mechanisms underlying the regulation of ERK1 and ERK2, their mode of action and their impact on the development and homeostasis of various organisms have been the focus of much attention for nearly three decades. In this Review, we discuss the current understanding of this important class of kinases. We begin with a brief overview of the structure, regulation, substrate recognition and subcellular localization of ERK1 and ERK2. We then systematically discuss how ERK signalling regulates six fundamental cellular processes in response to extracellular cues. These processes are cell proliferation, cell survival, cell growth, cell metabolism, cell migration and cell differentiation.
Collapse
|
230
|
Xu Y, Bankhead A, Tian X, Tang J, Ljungman M, Neamati N. Deletion of Glutathione S-Transferase Omega 1 Activates Type I Interferon Genes and Downregulates Tissue Factor. Cancer Res 2020; 80:3692-3705. [PMID: 32571799 DOI: 10.1158/0008-5472.can-20-0530] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/16/2020] [Accepted: 06/17/2020] [Indexed: 12/18/2022]
Abstract
GST omega 1 (GSTO1) is an atypical GST isoform that is overexpressed in several cancers and has been implicated in drug resistance. Currently, no small-molecule drug targeting GSTO1 is under clinical development. Here we have validated GSTO1 as an impactful target in oncology. Transcriptional profiling coupled with proteomics uncovered novel pharmacodynamic markers and cellular pathways regulated by GSTO1. CRISPR/Cas9 GSTO1 knockout (KO) cell lines failed to form tumors or displayed growth delay in vivo; they also formed smaller 3D spheroids in vitro. Multiomics analysis in GSTO1 KO cells found a strong positive correlation with cell adhesion molecules and IFN response pathways and a strong negative correlation with Myc transcriptional signature. In addition, several clinically used drugs showed significant synthetic lethality with loss or inhibition of GSTO1. Transcription and protein expression of tissue factor (gene name, F3) were downregulated in response to GSTO1 KO. F3 is associated with poor patient survival and promotion of tumor progression in multiple cancers and is a known risk factor for metastasis. Transcription of F3 was regulated by IL1β, whose secretion decreased upon inhibition of GSTO1, suggesting that IL1β links GSTO1 expression and F3 transcription. In summary, our results implicate GSTO1 as a potential therapeutic target in cancer and offer new mechanistic insights into its significant role in cancer progression. SIGNIFICANCE: These findings validate GSTO1 as a therapeutic target in cancer and implicate GSTO1 in the modulation of tumor growth, immune responses, and expression of F3.
Collapse
Affiliation(s)
- Yibin Xu
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan.,Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Armand Bankhead
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.,Department of Biostatistics and Department of Computational Medicine and Bioinformatics, Ann Arbor, Michigan
| | - Xiaoli Tian
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan.,Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Jianming Tang
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan.,Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Mats Ljungman
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.,Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.,Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan. .,Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
231
|
Ma L, Liu W, Xu A, Ji Q, Ma Y, Tai Y, Wang Y, Shen C, Liu Y, Wang T, Han J, Zhao C. Activator of thyroid and retinoid receptor increases sorafenib resistance in hepatocellular carcinoma by facilitating the Warburg effect. Cancer Sci 2020; 111:2028-2040. [PMID: 32279388 PMCID: PMC7293092 DOI: 10.1111/cas.14412] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/27/2020] [Accepted: 04/01/2020] [Indexed: 01/02/2023] Open
Abstract
Sorafenib resistance is a major challenge in the therapy for advanced hepatocellular carcinoma (HCC). However, the underlying molecular mechanisms of HCC resistance to sorafenib remain unclear. Activator of thyroid and retinoid receptor (ACTR, also known as SRC‐3), overexpressed in HCC patients, plays an important oncogenic role in HCC; however, the link between ACTR and sorafenib resistance in HCC is unknown. Our study demonstrated that ACTR was one of the most upregulated genes in sorafenib‐resistant HCC xenografts. ACTR increases sorafenib resistance through regulation of the Warburg effect. ACTR promotes glycolysis through upregulation of glucose uptake, ATP and lactate production, and reduction of the extracellular acidification and the oxygen consumption rates. Glycolysis regulated by ACTR is vital for the susceptibility of HCC to sorafenib in vitro and in vivo. Mechanistically, ACTR knockout or knockdown decreases the expression of glycolytic enzymes. In HCC patients, ACTR expression is positively correlated with glycolytic gene expression and is associated with poorer outcome. Furthermore, ACTR interacts with the central regulator of the Warburg effect, c‐Myc, and promotes its recruitment to glycolytic gene promoters. Our findings provide new clues regarding the role of ACTR as a prospective sensitizing target for sorafenib therapy in HCC.
Collapse
Affiliation(s)
- Luyuan Ma
- Department of Infectious Diseases, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wenpeng Liu
- Department of Infectious Diseases, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - An Xu
- Department of Oncology, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Quanbo Ji
- Department of Thoracic Surgery, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Yongfu Ma
- Department of Thoracic Surgery, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Yanhong Tai
- Department of Oncology, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Yadong Wang
- Department of Infectious Diseases, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chuan Shen
- Department of Infectious Diseases, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ying Liu
- Department of Ophthalmology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Tao Wang
- Department of Oncology, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Juqiang Han
- Department of Liver Disease, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Caiyan Zhao
- Department of Infectious Diseases, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
232
|
Heterogeneity of Glucose Transport in Lung Cancer. Biomolecules 2020; 10:biom10060868. [PMID: 32517099 PMCID: PMC7356687 DOI: 10.3390/biom10060868] [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: 05/11/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
Increased glucose uptake is a known hallmark of cancer. Cancer cells need glucose for energy production via glycolysis and the tricarboxylic acid cycle, and also to fuel the pentose phosphate pathway, the serine biosynthetic pathway, lipogenesis, and the hexosamine pathway. For this reason, glucose transport inhibition is an emerging new treatment for different malignancies, including lung cancer. However, studies both in animal models and in humans have shown high levels of heterogeneity in the utilization of glucose and other metabolites in cancer, unveiling a complexity that is difficult to target therapeutically. Here, we present an overview of different levels of heterogeneity in glucose uptake and utilization in lung cancer, with diagnostic and therapeutic implications.
Collapse
|
233
|
Heydarzadeh S, Moshtaghie AA, Daneshpoor M, Hedayati M. Regulators of glucose uptake in thyroid cancer cell lines. Cell Commun Signal 2020; 18:83. [PMID: 32493394 PMCID: PMC7268348 DOI: 10.1186/s12964-020-00586-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/27/2020] [Indexed: 01/03/2023] Open
Abstract
Abstract Thyroid cancer is the most common sort of endocrine-related cancer with more prevalent in women and elderly individuals which has quickly widespread expansion in worldwide over the recent decades. Common features of malignant thyroid cells are to have accelerated metabolism and increased glucose uptake to optimize their energy supply which provides a fundamental advantage for growth. In tumor cells the retaining of required energy charge for cell survival is imperative, indeed glucose transporters are enable of promoting of this task. According to this relation it has been reported the upregulation of glucose transporters in various types of cancers. Human studies indicated that poor survival can be occurred following the high levels of GLUT1 expression in tumors. GLUT-1 and GLUT3 are the glucose transporters which seems to be mainly engaged with the oncogenesis of thyroid cancer and their expression in malignant tissues is much more than in the normal one. They are promising targets for the advancement of anticancer strategies. The lack of oncosuppressors have dominant effect on the membrane expression of GLUT1 and glucose uptake. Overexpression of hypoxia inducible factors have been additionally connected with distant metastasis in thyroid cancers which mediates transcriptional regulation of glycolytic genes including GLUT1 and GLUT3. Though the physiological role of the thyroid gland is well illustrated, but the metabolic regulations in thyroid cancer remain evasive. In this study we discuss proliferation pathways of the key regulators and signaling molecules such as PI3K-Akt, HIF-1, MicroRNA, PTEN, AMPK, BRAF, c-Myc, TSH, Iodide and p53 which includes in the regulation of GLUTs in thyroid cancer cells. Incidence of deregulations in cellular energetics and metabolism are the most serious signs of cancers. In conclusion, understanding the mechanisms of glucose transportation in normal and pathologic thyroid tissues is critically important and could provide significant insights in science of diagnosis and treatment of thyroid disease. Video Abstract
Graphical abstract ![]()
Collapse
Affiliation(s)
- Shabnam Heydarzadeh
- Department of Biochemistry, School of Biological Sciences, Falavarjan Branch Islamic Azad University, Isfahan, Iran
| | - Ali Asghar Moshtaghie
- Department of Biochemistry, School of Biological Sciences, Falavarjan Branch Islamic Azad University, Isfahan, Iran
| | - Maryam Daneshpoor
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
234
|
Rodríguez-Hernández MA, de la Cruz-Ojeda P, López-Grueso MJ, Navarro-Villarán E, Requejo-Aguilar R, Castejón-Vega B, Negrete M, Gallego P, Vega-Ochoa Á, Victor VM, Cordero MD, Del Campo JA, Bárcena JA, Padilla CA, Muntané J. Integrated molecular signaling involving mitochondrial dysfunction and alteration of cell metabolism induced by tyrosine kinase inhibitors in cancer. Redox Biol 2020; 36:101510. [PMID: 32593127 PMCID: PMC7322178 DOI: 10.1016/j.redox.2020.101510] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/16/2020] [Indexed: 12/21/2022] Open
Abstract
Cancer cells have unlimited replicative potential, insensitivity to growth-inhibitory signals, evasion of apoptosis, cellular stress, and sustained angiogenesis, invasiveness and metastatic potential. Cancer cells adequately adapt cell metabolism and integrate several intracellular and redox signaling to promote cell survival in an inflammatory and hypoxic microenvironment in order to maintain/expand tumor phenotype. The administration of tyrosine kinase inhibitor (TKI) constitutes the recommended therapeutic strategy in different malignancies at advanced stages. There are important interrelationships between cell stress, redox status, mitochondrial function, metabolism and cellular signaling pathways leading to cell survival/death. The induction of apoptosis and cell cycle arrest widely related to the antitumoral properties of TKIs result from tightly controlled events involving different cellular compartments and signaling pathways. The aim of the present review is to update the most relevant studies dealing with the impact of TKI treatment on cell function. The induction of endoplasmic reticulum (ER) stress and Ca2+ disturbances, leading to alteration of mitochondrial function, redox status and phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt)-mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) signaling pathways that involve cell metabolism reprogramming in cancer cells will be covered. Emphasis will be given to studies that identify key components of the integrated molecular pattern including receptor tyrosine kinase (RTK) downstream signaling, cell death and mitochondria-related events that appear to be involved in the resistance of cancer cells to TKI treatments.
Collapse
Affiliation(s)
- María A Rodríguez-Hernández
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - P de la Cruz-Ojeda
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Mª José López-Grueso
- Department of Biochemistry and Molecular Biology, University of Cordoba, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Elena Navarro-Villarán
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Raquel Requejo-Aguilar
- Department of Biochemistry and Molecular Biology, University of Cordoba, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Beatriz Castejón-Vega
- Research Laboratory, Oral Medicine Department, University of Seville, Seville, Spain
| | - María Negrete
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Paloma Gallego
- Unit for the Clinical Management of Digestive Diseases, Hospital University "Nuestra Señora de Valme", Sevilla, Spain
| | - Álvaro Vega-Ochoa
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Victor M Victor
- Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Service of Endocrinology and Nutrition, Hospital University "Doctor Peset", Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; Department of Physiology, University of Valencia, Valencia, Spain
| | - Mario D Cordero
- Research Laboratory, Oral Medicine Department, University of Seville, Seville, Spain; Department of Physiology, Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center (CIBM), University of Granada, Armilla, Spain
| | - José A Del Campo
- Unit for the Clinical Management of Digestive Diseases, Hospital University "Nuestra Señora de Valme", Sevilla, Spain
| | - J Antonio Bárcena
- Department of Biochemistry and Molecular Biology, University of Cordoba, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - C Alicia Padilla
- Department of Biochemistry and Molecular Biology, University of Cordoba, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Department of General Surgery, Hospital University "Virgen del Rocío"/IBiS/CSIC/University of Seville, Seville, Spain.
| |
Collapse
|
235
|
BCG Vaccinations Upregulate Myc, a Central Switch for Improved Glucose Metabolism in Diabetes. iScience 2020; 23:101085. [PMID: 32380424 PMCID: PMC7205768 DOI: 10.1016/j.isci.2020.101085] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/03/2020] [Accepted: 04/15/2020] [Indexed: 12/11/2022] Open
Abstract
Myc has emerged as a pivotal transcription factor for four metabolic pathways: aerobic glycolysis, glutaminolysis, polyamine synthesis, and HIF-1α/mTOR. Each of these pathways accelerates the utilization of sugar. The BCG vaccine, a derivative of Mycobacteria-bovis, has been shown to trigger a long-term correction of blood sugar levels to near normal in type 1 diabetics (T1D). Here we reveal the underlying mechanisms behind this beneficial microbe-host interaction. We show that baseline glucose transport is deficient in T1D monocytes but is improved by BCG in vitro and in vivo. We then show, using RNAseq in monocytes and CD4 T cells, that BCG treatment over 56 weeks in humans is associated with upregulation of Myc and activation of nearly two dozen Myc-target genes underlying the four metabolic pathways. This is the first documentation of BCG induction of Myc and its association with systemic blood sugar control in a chronic disease like diabetes. T1D has insufficient aerobic glycolysis; this causes insufficient sugar utilization BCG vaccine lowers blood sugar levels in T1D by augmenting aerobic glycolysis BCG-induced shift to aerobic glycolysis is associated with Myc activation Host-microbe BCG interactions through Myc activate sugar-regulating genes in T1D
Collapse
|
236
|
Hamuro J, Numa K, Fujita T, Toda M, Ueda K, Tokuda Y, Mukai A, Nakano M, Ueno M, Kinoshita S, Sotozono C. Metabolites Interrogation in Cell Fate Decision of Cultured Human Corneal Endothelial Cells. Invest Ophthalmol Vis Sci 2020; 61:10. [PMID: 32049346 PMCID: PMC7324440 DOI: 10.1167/iovs.61.2.10] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Aiming to clarify the metabolic interrogation in cell fate decision of cultured human corneal endothelial cells (cHCECs). Methods To analyze the metabolites in the culture supernatants (CS), 34 metabolome measurements were carried out for mature differentiated and a variety of cHCECs with cell state transition through a facility service. Integrated proteomics research for cell lysates by liquid chromatography−tandem mass spectrometry (LC-MS/MS) was performed for 3 aliquots of each high-quality or low-quality cHCEC subpopulations (SP). The investigations for the focused genes involved in cHCEC metabolism were performed by using DAVID and its options “KEGG_PATHWAY.” Results The clusters of metabolites coincided well with the distinct content of CD44−/+ SPs. Both secreted pyruvic acid and lactic acid in the CS were negatively correlated with the content of high-quality SPs. Lactic acid and pyruvic acid in the CS exhibited the positive correlation with that of Ile, Leu, and Ser, whereas the negative correlation was with glutamine. Platelet-derived growth factor-ββ in the CS negatively correlated with lactic acid in CS, indicating indirectly the positive correlation with the content of CD44−/+ SPs. Upregulated glycolytic enzymes and influx of glutamine to the tricarboxylic acid cycle may be linked with a metabolic rewiring converting oxidative metabolism in mature differentiated CD44−/+SPs into a glycolytic flux-dependent state in immature SPs with cell state transition. Conclusions The findings suggest that the cell fate decision of cHCECs may be dictated at least partly through metabolic rewiring.
Collapse
|
237
|
Pant K, Richard S, Peixoto E, Gradilone SA. Role of Glucose Metabolism Reprogramming in the Pathogenesis of Cholangiocarcinoma. Front Med (Lausanne) 2020; 7:113. [PMID: 32318579 PMCID: PMC7146077 DOI: 10.3389/fmed.2020.00113] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/12/2020] [Indexed: 12/21/2022] Open
Abstract
Cholangiocarcinoma (CCA) is one of the most lethal cancers, and its rate of occurrence is increasing annually. The diagnoses of CCA patients remain elusive due to the lack of early symptoms and is misdiagnosed as HCC in a considerable percentage of patients. It is crucial to explore the underlying mechanisms of CCA carcinogenesis and development to find out specific biomarkers for early diagnosis of CCA and new promising therapeutic targets. In recent times, the reprogramming of tumor cells metabolism has been recognized as a hallmark of cancer. The modification from the oxidative phosphorylation metabolic pathway to the glycolysis pathway in CCA meets the demands of cancer cell proliferation and provides a favorable environment for tumor development. The alteration of metabolic programming in cancer cells is complex and may occur via mutations and epigenetic modifications within oncogenes, tumor suppressor genes, signaling pathways, and glycolytic enzymes. Herein we review the altered metabolism in cancer and the signaling pathways involved in this phenomena as they may affect CCA development. Understanding the regulatory pathways of glucose metabolism such as Akt/mTOR, HIF1α, and cMyc in CCA may further develop our knowledge of this devastating disease and may offer relevant information in the exploration of new diagnostic biomarkers and targeted therapeutic approaches for CCA.
Collapse
Affiliation(s)
- Kishor Pant
- The Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Seth Richard
- The Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Estanislao Peixoto
- The Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Sergio A Gradilone
- The Hormel Institute, University of Minnesota, Austin, MN, United States.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
238
|
Almahmoud S, Jin W, Geng L, Wang J, Wang X, Vennerstrom JL, Zhong HA. Ligand-based design of GLUT inhibitors as potential antitumor agents. Bioorg Med Chem 2020; 28:115395. [PMID: 32113844 PMCID: PMC11491113 DOI: 10.1016/j.bmc.2020.115395] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/09/2020] [Accepted: 02/14/2020] [Indexed: 01/01/2023]
Abstract
Glucose transporters (GLUTs) regulate glucose uptake and are often overexpressed in several human tumors. To identify new chemotypes targeting GLUT1, we built a pharmacophore model and searched against a NCI compound database. Sixteen hit molecules with good docking scores were screened for GLUT1 inhibition and antiproliferative activities. From these, we identified that compounds 2, 5, 6 and 13 inhibited the cell viability in a dose-dependent manner and that the IC50s of 2 and 6 are<10 µM concentration in the HCT116 colon cancer cell line. Lead compound 13 (NSC295720) was a GLUT1 inhibitor. Docking studies show that GLUT1 residues Phe291, Phe379, Glu380, Trp388, and Trp412 were important for inhibitor binding.
Collapse
Affiliation(s)
- Suliman Almahmoud
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Wei Jin
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, 460 W 12(th) Ave., Columbus, OH 43210, United States
| | - Liying Geng
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, 460 W 12(th) Ave., Columbus, OH 43210, United States
| | - Jing Wang
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, 460 W 12(th) Ave., Columbus, OH 43210, United States
| | - Xiaofang Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Jonathan L Vennerstrom
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Haizhen A Zhong
- Department of Chemistry, University of Nebraska at Omaha, 6001 Dodge Street, Omaha, Nebraska 68182, United States.
| |
Collapse
|
239
|
Commander R, Wei C, Sharma A, Mouw JK, Burton LJ, Summerbell E, Mahboubi D, Peterson RJ, Konen J, Zhou W, Du Y, Fu H, Shanmugam M, Marcus AI. Subpopulation targeting of pyruvate dehydrogenase and GLUT1 decouples metabolic heterogeneity during collective cancer cell invasion. Nat Commun 2020; 11:1533. [PMID: 32210228 PMCID: PMC7093428 DOI: 10.1038/s41467-020-15219-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 02/24/2020] [Indexed: 12/21/2022] Open
Abstract
Phenotypic heterogeneity exists within collectively invading packs of tumor cells, suggesting that cellular subtypes cooperate to drive invasion and metastasis. Here, we take a chemical biology approach to probe cell:cell cooperation within the collective invasion pack. These data reveal metabolic heterogeneity within invasive chains, in which leader cells preferentially utilize mitochondrial respiration and trailing follower cells rely on elevated glucose uptake. We define a pyruvate dehydrogenase (PDH) dependency in leader cells that can be therapeutically exploited with the mitochondria-targeting compound alexidine dihydrochloride. In contrast, follower cells highly express glucose transporter 1 (GLUT1), which sustains an elevated level of glucose uptake required to maintain proliferation. Co-targeting of both leader and follower cells with PDH and GLUT1 inhibitors, respectively, inhibits cell growth and collective invasion. Taken together, our work reveals metabolic heterogeneity within the lung cancer collective invasion pack and provides rationale for co-targeting PDH and GLUT1 to inhibit collective invasion. The presence of phenotypic heterogeneity in collectively invading cells suggests cooperation amongst distinct subtypes of cells to promote invasion and metastasis. Here, the authors use chemical biology tools and report metabolic heterogeneity within the lung cancer collective invasion pack.
Collapse
Affiliation(s)
- R Commander
- Graduate Program in Cancer Biology, Emory University, Atlanta, GA, USA
| | - C Wei
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - A Sharma
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - J K Mouw
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - L J Burton
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - E Summerbell
- Graduate Program in Cancer Biology, Emory University, Atlanta, GA, USA
| | - D Mahboubi
- Graduate Program in Molecular Systems Pharmacology, Emory University, Atlanta, GA, USA
| | - R J Peterson
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA, USA
| | - J Konen
- Department of Thoracic/Head & Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - W Zhou
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Y Du
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA.,Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA, USA
| | - H Fu
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.,Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA.,Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA, USA
| | - M Shanmugam
- Winship Cancer Institute, Emory University, Atlanta, GA, USA. .,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.
| | - A I Marcus
- Winship Cancer Institute, Emory University, Atlanta, GA, USA. .,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
240
|
Xia M, Feng S, Chen Z, Wen G, Zu X, Zhong J. Non-coding RNAs: Key regulators of aerobic glycolysis in breast cancer. Life Sci 2020; 250:117579. [PMID: 32209425 DOI: 10.1016/j.lfs.2020.117579] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/04/2020] [Accepted: 03/19/2020] [Indexed: 12/21/2022]
Abstract
Although extensive research progress has been made in breast cancer in recent years, yet the morbidity and mortality rates of breast cancer are rising, making it the major disease that endangers women's health. Energy metabolism reprogramming is featured by a state termed "aerobic glycolysis" or the Warburg effect that glycolysis is preferred even under aerobic conditions in neoplastic diseases. Widely acknowledged as an emerging hallmark in cancers, this metabolic switch shows a sophisticated role in the pathogenesis of breast cancer. The regulating effect of non-coding RNAs (ncRNAs) composed of microRNAs, long non-coding RNAs and circular RNAs is closely related to the glycolysis in breast cancer. Therefore, understand the mechanisms of ncRNAs of aerobic glycolysis in breast cancer may provide new strategy for the disease.
Collapse
Affiliation(s)
- Min Xia
- Institute of Clinical Medicine, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China; Department of Metabolism and Endocrinology, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China
| | - Shujun Feng
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, University of South China
| | - Zuyao Chen
- Institute of Clinical Medicine, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China
| | - Gebo Wen
- Institute of Clinical Medicine, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China; Department of Metabolism and Endocrinology, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China
| | - Xuyu Zu
- Institute of Clinical Medicine, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China; Cancer Research Institute, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China.
| | - Jing Zhong
- Institute of Clinical Medicine, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China; Cancer Research Institute, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
241
|
Zuo S, Wu L, Wang Y, Yuan X. Long Non-coding RNA MEG3 Activated by Vitamin D Suppresses Glycolysis in Colorectal Cancer via Promoting c-Myc Degradation. Front Oncol 2020; 10:274. [PMID: 32219064 PMCID: PMC7078156 DOI: 10.3389/fonc.2020.00274] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/17/2020] [Indexed: 01/04/2023] Open
Abstract
Colorectal cancer (CRC), a common tumor, is characterized by a high mortality rate. Long non-coding RNA maternally expressed gene 3 (MEG3) serves a regulatory role in the carcinogenesis and progression of several types of cancer; however, its role in CRC remains largely unknown. The aim of this study was to explore the regulatory role and mechanism(s) of MEG3 in CRC. The Warburg effect or aerobic glycolysis is characteristic of the metabolism of tumor cells. To determine the effect of MEG3 on glycolysis of CRC cells, we used an XF analyzer to perform glycolysis stress test assays and found that overexpression of MEG3 significantly inhibited glycolysis, glycolytic capacity, as well as lactate production in CRC cells, whereas knockdown of MEG3 produced the opposite effect. Mechanistically, overexpression of MEG3 induced ubiquitin-dependent degradation of c-Myc and inhibited c-Myc target genes involved in the glycolysis pathway such as lactate dehydrogenase A, pyruvate kinase muscle 2, and hexokinase 2. Moreover, we found that MEG3 can be activated by vitamin D and vitamin D receptor (VDR). Clinical data demonstrated that MEG3 was positively associated with serum vitamin D concentrations in patients with CRC. We found that 1,25(OH)2D3 treatment increased MEG3 expression, and knockdown of VDR abolished the effect of MEG3 on glycolysis. These results indicate that vitamin D-activated MEG3 suppresses aerobic glycolysis in CRC cells via degradation of c-Myc. Thus, vitamin D may have therapeutic value in the treatment of CRC.
Collapse
Affiliation(s)
- Siyu Zuo
- Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing, China
| | - Lei Wu
- Department of Clinical Medicine, First Clinical Medicine College, Nanjing Medical University, Nanjing, China
| | - Yi Wang
- Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing, China
| | - Xiaoqin Yuan
- Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing, China.,Key Laboratory for Aging and Disease, Nanjing Medical University, Nanjing, China
| |
Collapse
|
242
|
Kreuzaler P, Panina Y, Segal J, Yuneva M. Adapt and conquer: Metabolic flexibility in cancer growth, invasion and evasion. Mol Metab 2020; 33:83-101. [PMID: 31668988 PMCID: PMC7056924 DOI: 10.1016/j.molmet.2019.08.021] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/05/2019] [Accepted: 08/14/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND It has been known for close to a century that, on average, tumors have a metabolism that is different from those found in healthy tissues. Typically, tumors show a biosynthetic metabolism that distinguishes itself by engaging in large scale aerobic glycolysis, heightened flux through the pentose phosphate pathway, and increased glutaminolysis among other means. However, it is becoming equally clear that non tumorous tissues at times can engage in similar metabolism, while tumors show a high degree of metabolic flexibility reacting to cues, and stresses in their local environment. SCOPE OF THE REVIEW In this review, we want to scrutinize historic and recent research on metabolism, comparing and contrasting oncogenic and physiological metabolic states. This will allow us to better define states of bona fide tumor metabolism. We will further contextualize the stress response and the metabolic evolutionary trajectory seen in tumors, and how these contribute to tumor progression. Lastly, we will analyze the implications of these characteristics with respect to therapy response. MAJOR CONCLUSIONS In our review, we argue that there is not one single oncogenic state, but rather a diverse set of oncogenic states. These are grounded on a physiological proliferative/wound healing program but distinguish themselves due to their large scale of proliferation, mutations, and transcriptional changes in key metabolic pathways, and the adaptations to widespread stress signals within tumors. We find evidence for the necessity of metabolic flexibility and stress responses in tumor progression and how these responses in turn shape oncogenic progression. Lastly, we find evidence for the notion that the metabolic adaptability of tumors frequently frustrates therapeutic interventions.
Collapse
|
243
|
Tiemin P, Peng X, Qingfu L, Yan W, Junlin X, Zhefeng H, Ming Z, Desen L, Qinghui M. Dysregulation of the miR-148a-GLUT1 axis promotes the progression and chemoresistance of human intrahepatic cholangiocarcinoma. Oncogenesis 2020; 9:19. [PMID: 32054829 PMCID: PMC7018977 DOI: 10.1038/s41389-020-0207-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/30/2020] [Accepted: 02/04/2020] [Indexed: 11/09/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is a highly fatal malignant cancer worldwide. Elucidating the underlying molecular mechanism of iCCA progression is critical for the identification of new therapeutic targets. The present study explored the role of the miR-148a-GLUT1 axis in the progression of iCCA. The expression of GLUT1 was detected by using immunohistochemistry, western blot assays, and real-time polymerase chain reaction. The effects of GLUT1 on cell proliferation, invasion, and chemoresistance were investigated both in vitro and in vivo. A luciferase reporter assay was used to explore the effect of miR-148a on GLUT1 expression. GLUT1 was overexpressed in iCCA tissues. GLUT1 overexpression was associated with shorter overall and disease-free survival. Knockdown of GLUT1 reduced, while overexpression of GLUT1 promoted, the proliferation, motility, and invasiveness of iCCA cells in vitro and in vivo. Silencing GLUT1 significantly sensitized iCCA cells to gemcitabine in vitro and in vivo. GLUT1 was directly regulated by miR-148a, whose downregulation was associated with the proliferation, migration, and invasion of iCCA cells. WZB117, a GLUT1 inhibitor, inhibited tumor growth in an iCCA patient-derived xenograft model. These results indicate that downregulation of miR-148a levels results in GLUT1 overexpression in iCCA, leading to iCCA progression and gemcitabine resistance.
Collapse
Affiliation(s)
- Pei Tiemin
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao Peng
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lang Qingfu
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wang Yan
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xue Junlin
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - He Zhefeng
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhao Ming
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Liang Desen
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meng Qinghui
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
244
|
Marchingo JM, Sinclair LV, Howden AJ, Cantrell DA. Quantitative analysis of how Myc controls T cell proteomes and metabolic pathways during T cell activation. eLife 2020; 9:53725. [PMID: 32022686 PMCID: PMC7056270 DOI: 10.7554/elife.53725] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
T cell expansion and differentiation are critically dependent on the transcription factor c-Myc (Myc). Herein we use quantitative mass-spectrometry to reveal how Myc controls antigen receptor driven cell growth and proteome restructuring in murine T cells. Analysis of copy numbers per cell of >7000 proteins provides new understanding of the selective role of Myc in controlling the protein machinery that govern T cell fate. The data identify both Myc dependent and independent metabolic processes in immune activated T cells. We uncover that a primary function of Myc is to control expression of multiple amino acid transporters and that loss of a single Myc-controlled amino acid transporter effectively phenocopies the impact of Myc deletion. This study provides a comprehensive map of how Myc selectively shapes T cell phenotypes, revealing that Myc induction of amino acid transport is pivotal for subsequent bioenergetic and biosynthetic programs and licences T cell receptor driven proteome reprogramming.
Collapse
Affiliation(s)
- Julia M Marchingo
- Cell Signalling and Immunology Division, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Linda V Sinclair
- Cell Signalling and Immunology Division, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Andrew Jm Howden
- Cell Signalling and Immunology Division, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Doreen A Cantrell
- Cell Signalling and Immunology Division, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
245
|
Bi J, Chowdhry S, Wu S, Zhang W, Masui K, Mischel PS. Altered cellular metabolism in gliomas - an emerging landscape of actionable co-dependency targets. Nat Rev Cancer 2020; 20:57-70. [PMID: 31806884 DOI: 10.1038/s41568-019-0226-5] [Citation(s) in RCA: 223] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/31/2019] [Indexed: 12/18/2022]
Abstract
Altered cellular metabolism is a hallmark of gliomas. Propelled by a set of recent technological advances, new insights into the molecular mechanisms underlying glioma metabolism are rapidly emerging. In this Review, we focus on the dynamic nature of glioma metabolism and how it is shaped by the interaction between tumour genotype and brain microenvironment. Recent advances integrating metabolomics with genomics are discussed, yielding new insight into the mechanisms that drive glioma pathogenesis. Studies that shed light on interactions between the tumour microenvironment and tumour genotype are highlighted, providing important clues as to how gliomas respond to and adapt to their changing tissue and biochemical contexts. Finally, a road map for the discovery of potential new glioma drug targets is suggested, with the goal of translating these new insights about glioma metabolism into clinical benefits for patients.
Collapse
Affiliation(s)
- Junfeng Bi
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Sudhir Chowdhry
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Sihan Wu
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Wenjing Zhang
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Kenta Masui
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Paul S Mischel
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA.
- Department of Pathology, UCSD School of Medicine, La Jolla, CA, USA.
- Moores Cancer Center, UCSD School of Medicine, La Jolla, CA, USA.
| |
Collapse
|
246
|
Marengo B, Pulliero A, Izzotti A, Domenicotti C. miRNA Regulation of Glutathione Homeostasis in Cancer Initiation, Progression and Therapy Resistance. Microrna 2020; 9:187-197. [PMID: 31849293 PMCID: PMC7366003 DOI: 10.2174/2211536609666191218103220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/04/2019] [Accepted: 11/13/2019] [Indexed: 12/16/2022]
Abstract
Glutathione (GSH) is the most abundant antioxidant that contributes to regulating the cellular production of Reactive Oxygen Species (ROS) which, maintained at physiological levels, can exert a function of second messengers in living organisms. In fact, it has been demonstrated that moderate amounts of ROS can activate the signaling pathways involved in cell growth and proliferation, while high levels of ROS induce DNA damage leading to cancer development. Therefore, GSH is a crucial player in the maintenance of redox homeostasis and its metabolism has a role in tumor initiation, progression, and therapy resistance. Our recent studies demonstrated that neuroblastoma cells resistant to etoposide, a common chemotherapeutic drug, show a partial monoallelic deletion of the locus coding for miRNA 15a and 16-1 leading to a loss of these miRNAs and the activation of GSH-dependent responses. Therefore, the aim of this review is to highlight the role of specific miRNAs in the modulation of intracellular GSH levels in order to take into consideration the use of modulators of miRNA expression as a useful strategy to better sensitize tumors to current therapies.
Collapse
Affiliation(s)
- Barbara Marengo
- Address correspondence to this author at the Department of Experimental Medicine, University of Genoa, Genoa, Italy; Tel: +39 010 3538831; Fax: +39 010 3538836; E-mail:
| | | | | | | |
Collapse
|
247
|
López-Sánchez LM, Aranda E, Rodríguez-Ariza A. Nitric oxide and tumor metabolic reprogramming. Biochem Pharmacol 2019; 176:113769. [PMID: 31862448 DOI: 10.1016/j.bcp.2019.113769] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/13/2019] [Indexed: 12/20/2022]
Abstract
Nitric oxide (NO) has been highlighted as an important agent in tumor processes. However, a complete understanding of the mechanisms by which this simple diatomic molecule contributes in tumorigenesis is lacking. Evidence is rapidly accumulating that metabolic reprogramming is a major new aspect of NO biology and this review is aimed to summarize recent research progress on this novel feature that expands the complex and multifaceted role of NO in cancer. Therefore, we discuss how NO may influence glucose and glutamine utilization by tumor cells, and its participation in the regulation of mitochondrial function and dynamics, that is an important mechanism through which cancer cells reprogram their metabolism to meet the biosynthetic needs of rapid proliferation. Finally, we also discuss the NO-related metabolic rewiring involved in the modification of the tumor microenvironment to support cancer invasion and the escape from immune system-mediated recognition. Protein S-nitrosylation appears as a common mechanism by which NO signaling reprograms metabolism. Hence, future research is needed on dysregulated S-nitrosylation/denitrosylation in cancer to comprehend the NO-induced metabolic changes in tumor cells and the role of NO in the metabolic crosstalk within tumor microenvironment.
Collapse
Affiliation(s)
- Laura M López-Sánchez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Av. Menéndez Pidal s/n, E14004 Córdoba, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Av. Monforte de Lemos, 3-5, E 28029 Madrid, Spain
| | - Enrique Aranda
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Av. Menéndez Pidal s/n, E14004 Córdoba, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Av. Monforte de Lemos, 3-5, E 28029 Madrid, Spain; Unidad de Gestión Clínica de Oncología Médica, Hospital Reina Sofía, Universidad de Córdoba, Av. Menéndez Pidal s/n, E14004 Córdoba, Spain
| | - Antonio Rodríguez-Ariza
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Av. Menéndez Pidal s/n, E14004 Córdoba, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Av. Monforte de Lemos, 3-5, E 28029 Madrid, Spain; Unidad de Gestión Clínica de Oncología Médica, Hospital Reina Sofía, Universidad de Córdoba, Av. Menéndez Pidal s/n, E14004 Córdoba, Spain.
| |
Collapse
|
248
|
Jiang W, Liu P, Zhang J, Yang W. Identification of Key Candidate Genes and Pathways of Candida albicans-Infected Human Umbilical Vein Endothelial Cells and Drug Screening. Indian J Microbiol 2019; 60:62-69. [PMID: 32089575 PMCID: PMC7000633 DOI: 10.1007/s12088-019-00847-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 12/04/2019] [Indexed: 12/19/2022] Open
Abstract
Candida albicans is a common opportunistic pathogen that can cause serious infection by blood transmission. C. albicans enters the blood circulation and adheres to the endothelial cells of the vascular wall. However, the detailed mechanism of the effect of C. albicans on the endothelial cells remains unclear. In this study, the microarray expression profile of human umbilical vein endothelial cells exposed to C. albicans was analyzed. The 191 up-regulated genes were enriched in TNF, T cell receptor, and NF-kappa B signaling pathways. The 71 down-regulated genes were enriched in pyruvate metabolic, purine nucleotide metabolic, purine nucleotide biosynthetic, and humoral immune response processes. Gene set enrichment analysis showed that apoptosis, oxidative phosphorylation, IL6/JAK/STAT3 signaling pathways were enriched. Moreover, two hub genes with a high degree of connectivity, namely, MYC and IL6, were selected. Molecular screening of traditional Chinese medicine libraries was performed on the basis of the structure of MYC protein. The okanin had the highest docking score. MYC might be used as molecular targets for treatment. In addition, okanin may inhibit the infection of C. albicans. Thus, MYC can be subjected to further research.
Collapse
Affiliation(s)
- Wei Jiang
- 1Department of Infectious Diseases, Tianjin First Center Hospital, No. 24 Fukang Road, Nankai District, Tianjin, 300192 China
| | - Ping Liu
- 2Laboratory of Microbiology of Tianjin First Center Hospital, Tianjin, China
| | - Jianlei Zhang
- 2Laboratory of Microbiology of Tianjin First Center Hospital, Tianjin, China
| | - Wenjie Yang
- 1Department of Infectious Diseases, Tianjin First Center Hospital, No. 24 Fukang Road, Nankai District, Tianjin, 300192 China
| |
Collapse
|
249
|
Abstract
In this Review, Rashkovan et al. discuss the role of cancer metabolic circuitries feeding anabolism and redox potential in leukemia development and recent progress in translating these important findings to the clinic. Leukemia cell proliferation requires up-regulation and rewiring of metabolic pathways to feed anabolic cell growth. Oncogenic drivers directly and indirectly regulate metabolic pathways, and aberrant metabolism is central not only for leukemia proliferation and survival, but also mediates oncogene addiction with significant implications for the development of targeted therapies. This review explores leukemia metabolic circuitries feeding anabolism, redox potential, and energy required for tumor propagation with an emphasis on emerging therapeutic opportunities.
Collapse
Affiliation(s)
- Marissa Rashkovan
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Adolfo Ferrando
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA.,Department of Pediatrics, Columbia University, New York, NY 10032, USA.,Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
250
|
Satriano L, Lewinska M, Rodrigues PM, Banales JM, Andersen JB. Metabolic rearrangements in primary liver cancers: cause and consequences. Nat Rev Gastroenterol Hepatol 2019; 16:748-766. [PMID: 31666728 DOI: 10.1038/s41575-019-0217-8] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
Primary liver cancer (PLC) is the fourth most frequent cause of cancer-related death. The high mortality rates arise from late diagnosis and the limited accuracy of diagnostic and prognostic biomarkers. The liver is a major regulator, orchestrating the clearance of toxins, balancing glucose, lipid and amino acid uptake, managing whole-body metabolism and maintaining metabolic homeostasis. Tumour onset and progression is frequently accompanied by rearrangements of metabolic pathways, leading to dysregulation of metabolism. The limitation of current therapies targeting PLCs, such as hepatocellular carcinoma and cholangiocarcinoma, points towards the importance of deciphering this metabolic complexity. In this Review, we discuss the role of metabolic liver disruptions and the implications of these processes in PLCs, emphasizing their clinical relevance and value in early diagnosis and prognosis and as putative therapeutic targets. We also describe system biology approaches able to reconstruct the metabolic complexity of liver diseases. We also discuss whether metabolic rearrangements are a cause or consequence of PLCs, emphasizing the opportunity to clinically exploit the rewired metabolism. In line with this idea, we discuss circulating metabolites as promising biomarkers for PLCs.
Collapse
Affiliation(s)
- Letizia Satriano
- Biotech Research and Innovation Centre (BRIC) Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Monika Lewinska
- Biotech Research and Innovation Centre (BRIC) Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pedro M Rodrigues
- Biodonostia Health Research Institute, Donostia University Hospital, San Sebastian, Spain
| | - Jesus M Banales
- Biodonostia Health Research Institute, Donostia University Hospital, San Sebastian, Spain.,National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC) Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|