1
|
Moghaddam SJ, Savai R, Salehi-Rad R, Sengupta S, Kammer MN, Massion P, Beane JE, Ostrin EJ, Priolo C, Tennis MA, Stabile LP, Bauer AK, Sears CR, Szabo E, Rivera MP, Powell CA, Kadara H, Jenkins BJ, Dubinett SM, Houghton AM, Kim CF, Keith RL. Premalignant Progression in the Lung: Knowledge Gaps and Novel Opportunities for Interception of Non-Small Cell Lung Cancer. An Official American Thoracic Society Research Statement. Am J Respir Crit Care Med 2024; 210:548-571. [PMID: 39115548 PMCID: PMC11389570 DOI: 10.1164/rccm.202406-1168st] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Indexed: 08/13/2024] Open
Abstract
Rationale: Despite significant advances in precision treatments and immunotherapy, lung cancer is the most common cause of cancer death worldwide. To reduce incidence and improve survival rates, a deeper understanding of lung premalignancy and the multistep process of tumorigenesis is essential, allowing timely and effective intervention before cancer development. Objectives: To summarize existing information, identify knowledge gaps, formulate research questions, prioritize potential research topics, and propose strategies for future investigations into the premalignant progression in the lung. Methods: An international multidisciplinary team of basic, translational, and clinical scientists reviewed available data to develop and refine research questions pertaining to the transformation of premalignant lung lesions to advanced lung cancer. Results: This research statement identifies significant gaps in knowledge and proposes potential research questions aimed at expanding our understanding of the mechanisms underlying the progression of premalignant lung lesions to lung cancer in an effort to explore potential innovative modalities to intercept lung cancer at its nascent stages. Conclusions: The identified gaps in knowledge about the biological mechanisms of premalignant progression in the lung, together with ongoing challenges in screening, detection, and early intervention, highlight the critical need to prioritize research in this domain. Such focused investigations are essential to devise effective preventive strategies that may ultimately decrease lung cancer incidence and improve patient outcomes.
Collapse
|
2
|
Sun W, Cai B, Zhao Z, Li S, He Y, Xie S. Redirecting Tumor Evolution with Nanocompiler Precision for Enhanced Therapeutic Outcomes. Adv Healthc Mater 2024:e2400366. [PMID: 39039965 DOI: 10.1002/adhm.202400366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/16/2024] [Indexed: 07/24/2024]
Abstract
Precisely programming the highly plastic tumor expression profile to render it devoid of drug resistance and metastatic potential presents immense challenges. Here, a transformative nanocompiler designed to reprogram and stabilize the mutable state of tumor cells is introduced. This nanocompiler features a trio of components: 2-deoxy-d-glucose-modified lipid nanoparticles to inhibit glucose uptake, iron oxide nanoparticles to induce oxidative stress, and a deubiquitinase inhibitor to block adaptive protein profile changes in tumor cells. By specifically targeting the hypermetabolic nature of tumors, this approach disrupted their energy production, ultimately fostering a state of vulnerability and impeding their ability to adapt and resist. The results of this study indicate a substantial reduction in tumor growth and metastasis, thus demonstrating the potential of this strategy to manipulate tumor protein expression and fate. This proactive nanocompiler approach promises to steer cancer therapy toward more effective and lasting outcomes.
Collapse
Affiliation(s)
- Wenshe Sun
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, 250117, China
| | - Biao Cai
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Zejun Zhao
- Department of Ultrasound, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Shilun Li
- Department of Vascular Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yutian He
- Department of Ultrasound, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Shaowei Xie
- Department of Ultrasound, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| |
Collapse
|
3
|
Cho IY, Chang Y, Sung E, Park B, Kang JH, Shin H, Wild SH, Byrne CD, Ryu S. Glycemic status, insulin resistance, and mortality from lung cancer among individuals with and without diabetes. Cancer Metab 2024; 12:17. [PMID: 38902745 PMCID: PMC11188269 DOI: 10.1186/s40170-024-00344-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/13/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND The effects of glycemic status and insulin resistance on lung cancer remain unclear. We investigated the associations between both glycemic status and insulin resistance, and lung cancer mortality, in a young and middle-aged population with and without diabetes. METHODS This cohort study involved individuals who participated in routine health examinations. Lung cancer mortality was identified using national death records. Cox proportional hazards models were used to calculate hazard ratios (HRs) with 95% CIs for lung cancer mortality risk. RESULTS Among 666,888 individuals (mean age 39.9 ± 10.9 years) followed for 8.3 years (interquartile range, 4.6-12.7), 602 lung cancer deaths occurred. Among individuals without diabetes, the multivariable-adjusted HRs (95% CI) for lung cancer mortality comparing hemoglobin A1c categories (5.7-5.9, 6.0-6.4, and ≥ 6.5% or 39-41, 42-46, and ≥ 48 mmol/mol, respectively) with the reference (< 5.7% or < 39 mmol/mol) were 1.39 (1.13-1.71), 1.72 (1.33-2.20), and 2.22 (1.56-3.17), respectively. Lung cancer mortality was associated with fasting blood glucose categories in a dose-response manner (P for trend = 0.001) and with previously diagnosed diabetes. Insulin resistance (HOMA-IR ≥ 2.5) in individuals without diabetes was also associated with lung cancer mortality (multivariable-adjusted HR, 1.41; 95% CI, 1.13-1.75). These associations remained after adjusting for changing status in glucose, hemoglobin A1c, insulin resistance, smoking status, and other confounders during follow-up as time-varying covariates. CONCLUSIONS Glycemic status within both diabetes and prediabetes ranges and insulin resistance were independently associated with an increased risk of lung cancer mortality.
Collapse
Affiliation(s)
- In Young Cho
- Department of Family Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-Ro, Jongno-Gu, Seoul, 03181, Republic of Korea
- Department of Family Medicine & Supportive Care Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea
| | - Yoosoo Chang
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 04514, Republic of Korea
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Samsung Main Building B2, 250 Taepyung-Ro 2Ga, Jung-Gu, Seoul, 04514, Republic of Korea
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea
| | - Eunju Sung
- Department of Family Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-Ro, Jongno-Gu, Seoul, 03181, Republic of Korea.
| | - Boyoung Park
- Department of Preventive Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Jae-Heon Kang
- Department of Family Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-Ro, Jongno-Gu, Seoul, 03181, Republic of Korea
| | - Hocheol Shin
- Department of Family Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-Ro, Jongno-Gu, Seoul, 03181, Republic of Korea
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 04514, Republic of Korea
| | - Sarah H Wild
- Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Christopher D Byrne
- Nutrition and Metabolism, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Seungho Ryu
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 04514, Republic of Korea.
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Samsung Main Building B2, 250 Taepyung-Ro 2Ga, Jung-Gu, Seoul, 04514, Republic of Korea.
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea.
| |
Collapse
|
4
|
Fuentes-Rodriguez A, Mitchell A, Guérin SL, Landreville S. Recent Advances in Molecular and Genetic Research on Uveal Melanoma. Cells 2024; 13:1023. [PMID: 38920653 PMCID: PMC11201764 DOI: 10.3390/cells13121023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/27/2024] Open
Abstract
Uveal melanoma (UM), a distinct subtype of melanoma, presents unique challenges in its clinical management due to its complex molecular landscape and tendency for liver metastasis. This review highlights recent advancements in understanding the molecular pathogenesis, genetic alterations, and immune microenvironment of UM, with a focus on pivotal genes, such as GNAQ/11, BAP1, and CYSLTR2, and delves into the distinctive genetic and chromosomal classifications of UM, emphasizing the role of mutations and chromosomal rearrangements in disease progression and metastatic risk. Novel diagnostic biomarkers, including circulating tumor cells, DNA and extracellular vesicles, are discussed, offering potential non-invasive approaches for early detection and monitoring. It also explores emerging prognostic markers and their implications for patient stratification and personalized treatment strategies. Therapeutic approaches, including histone deacetylase inhibitors, MAPK pathway inhibitors, and emerging trends and concepts like CAR T-cell therapy, are evaluated for their efficacy in UM treatment. This review identifies challenges in UM research, such as the limited treatment options for metastatic UM and the need for improved prognostic tools, and suggests future directions, including the discovery of novel therapeutic targets, immunotherapeutic strategies, and advanced drug delivery systems. The review concludes by emphasizing the importance of continued research and innovation in addressing the unique challenges of UM to improve patient outcomes and develop more effective treatment strategies.
Collapse
Affiliation(s)
- Aurélie Fuentes-Rodriguez
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| | - Andrew Mitchell
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| | - Sylvain L. Guérin
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
| | - Solange Landreville
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| |
Collapse
|
5
|
Bajinka O, Ouedraogo SY, Golubnitschaja O, Li N, Zhan X. Energy metabolism as the hub of advanced non-small cell lung cancer management: a comprehensive view in the framework of predictive, preventive, and personalized medicine. EPMA J 2024; 15:289-319. [PMID: 38841622 PMCID: PMC11147999 DOI: 10.1007/s13167-024-00357-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 03/20/2024] [Indexed: 06/07/2024]
Abstract
Energy metabolism is a hub of governing all processes at cellular and organismal levels such as, on one hand, reparable vs. irreparable cell damage, cell fate (proliferation, survival, apoptosis, malignant transformation etc.), and, on the other hand, carcinogenesis, tumor development, progression and metastazing versus anti-cancer protection and cure. The orchestrator is the mitochondria who produce, store and invest energy, conduct intracellular and systemically relevant signals decisive for internal and environmental stress adaptation, and coordinate corresponding processes at cellular and organismal levels. Consequently, the quality of mitochondrial health and homeostasis is a reliable target for health risk assessment at the stage of reversible damage to the health followed by cost-effective personalized protection against health-to-disease transition as well as for targeted protection against the disease progression (secondary care of cancer patients against growing primary tumors and metastatic disease). The energy reprogramming of non-small cell lung cancer (NSCLC) attracts particular attention as clinically relevant and instrumental for the paradigm change from reactive medical services to predictive, preventive and personalized medicine (3PM). This article provides a detailed overview towards mechanisms and biological pathways involving metabolic reprogramming (MR) with respect to inhibiting the synthesis of biomolecules and blocking common NSCLC metabolic pathways as anti-NSCLC therapeutic strategies. For instance, mitophagy recycles macromolecules to yield mitochondrial substrates for energy homeostasis and nucleotide synthesis. Histone modification and DNA methylation can predict the onset of diseases, and plasma C7 analysis is an efficient medical service potentially resulting in an optimized healthcare economy in corresponding areas. The MEMP scoring provides the guidance for immunotherapy, prognostic assessment, and anti-cancer drug development. Metabolite sensing mechanisms of nutrients and their derivatives are potential MR-related therapy in NSCLC. Moreover, miR-495-3p reprogramming of sphingolipid rheostat by targeting Sphk1, 22/FOXM1 axis regulation, and A2 receptor antagonist are highly promising therapy strategies. TFEB as a biomarker in predicting immune checkpoint blockade and redox-related lncRNA prognostic signature (redox-LPS) are considered reliable predictive approaches. Finally, exemplified in this article metabolic phenotyping is instrumental for innovative population screening, health risk assessment, predictive multi-level diagnostics, targeted prevention, and treatment algorithms tailored to personalized patient profiles-all are essential pillars in the paradigm change from reactive medical services to 3PM approach in overall management of lung cancers. This article highlights the 3PM relevant innovation focused on energy metabolism as the hub to advance NSCLC management benefiting vulnerable subpopulations, affected patients, and healthcare at large. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00357-5.
Collapse
Affiliation(s)
- Ousman Bajinka
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Serge Yannick Ouedraogo
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, University Hospital Bonn, Venusberg Campus 1, Rheinische Friedrich-Wilhelms-University of Bonn, 53127 Bonn, Germany
| | - Na Li
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
6
|
Li G, Chen L, Bai H, Zhang L, Wang J, Li W. Depletion of squalene epoxidase in synergy with glutathione peroxidase 4 inhibitor RSL3 overcomes oxidative stress resistance in lung squamous cell carcinoma. PRECISION CLINICAL MEDICINE 2024; 7:pbae011. [PMID: 38779359 PMCID: PMC11109822 DOI: 10.1093/pcmedi/pbae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Background Lung squamous cell carcinoma (LUSC) lacks effective targeted therapies and has a poor prognosis. Disruption of squalene epoxidase (SQLE) has been implicated in metabolic disorders and cancer. However, the role of SQLE as a monooxygenase involved in oxidative stress remains unclear. Methods We analyzed the expression and prognosis of lung adenocarcinoma (LUAD) and LUSC samples from GEO and TCGA databases. The proliferative activity of the tumors after intervention of SQLE was verified by cell and animal experiments. JC-1 assay, flow cytometry, and Western blot were used to show changes in apoptosis after intervention of SQLE. Flow cytometry and fluorescence assay of ROS levels were used to indicate oxidative stress status. Results We investigated the unique role of SQLE expression in the diagnosis and prognosis prediction of LUSC. Knockdown of SQLE or treatment with the SQLE inhibitor terbinafine can suppress the proliferation of LUSC cells by inducing apoptosis and reactive oxygen species accumulation. However, depletion of SQLE also results in the impairment of lipid peroxidation and ferroptosis resistance such as upregulation of glutathione peroxidase 4. Therefore, prevention of SQLE in synergy with glutathione peroxidase 4 inhibitor RSL3 effectively mitigates the proliferation and growth of LUSC. Conclusion Our study indicates that the low expression of SQLE employs adaptive survival through regulating the balance of apoptosis and ferroptosis resistance. In future, the combinational therapy of targeting SQLE and ferroptosis could be a promising approach in treating LUSC.
Collapse
Affiliation(s)
- Guo Li
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing 100021, China
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lu Chen
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hua Bai
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing 100021, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, China
| | - Jie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing 100021, China
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu 610041, China
| |
Collapse
|
7
|
Liu Y, Li X, Yang J, Chen S, Zhu C, Shi Y, Dang S, Zhang W, Li W. Pan-cancer analysis of SLC2A family genes as prognostic biomarkers and therapeutic targets. Heliyon 2024; 10:e29655. [PMID: 38655365 PMCID: PMC11036058 DOI: 10.1016/j.heliyon.2024.e29655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024] Open
Abstract
Background The major facilitator superfamily glucose transporters (GLUTs), encoded by solute carrier 2A (SLC2A) genes, mediate the transmembrane movement and uptake of glucose. To satisfy the improved energy demands, glycolysis flux is increased in cancers compared with healthy tissues. Multiple diseases, including cancer, have been associated with GLUTs. Nevertheless, not much research has been done on the functions of SLC2As in pan-cancer prognosis or their clinical treatment potential. Methods The SLC2A family genes' level of expression and prognostic values were analyzed in relation to pan-cancer. We then examined the association among SLC2As expression and TME, Stemness score, clinical characteristics, immune subtypes, and drug sensitivity. We merged bioinformatics analysis techniques with up-to-date public databases. Additionally, SLC2As from the KOBAS database were subjected to enrichment analysis. Results We discovered that SLC2As' gene expression differed significantly between normal tissues and many malignancies. A number of tumors from various databases demonstrate a relationship between prognosis and SLC2A family gene expression. For instance, SLC2A2 and SLC2A5 were associated with the overall survival (OS) of hepatocellular carcinoma. SLC2A1 was associated with the OS of lung adenocarcinoma and pancreatic adenocarcinoma. Moreover, the SLC2A family gene expression is significantly correlated with the pan-cancer stromal and immune scores, and the RNA and DNA stemness scores. Furthermore, we found that the majority of SLC2As had a strong correlation with the tumor stages in KIRC. The immunological subtypes and all members of the SLC2A gene family exhibited a substantial correlation. Moreover, pathways containing insulin resistance and adipocytokine signaling pathway may influence the progression of some cancers. Finally, there is a significant positive or negative connection between drug sensitivity and SLC2A1 expression. Conclusion Our research highlights the significant promise of SLC2As as prognostic indicators and offers insightful approaches for upcoming exploration of SLC2As as putative therapeutic targets in malignancies.
Collapse
Affiliation(s)
- Yating Liu
- Department of Cancer Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xinyu Li
- Department of Cancer Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jie Yang
- Department of Pediatric Dentistry, Peking University School of Stomatology, Beijing, China
| | - Shanshan Chen
- Department of Cancer Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Changyu Zhu
- Department of Cancer Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yijun Shi
- Department of Cancer Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Shoutao Dang
- Department of Cancer Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Weitao Zhang
- Department of Cancer Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Wei Li
- Department of Cancer Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Zhu L, Chen C, Cai Y, Li Y, Gong L, Zhu T, Kong L, Luo J. Identification of a ferritinophagy inducer via sinomenine modification for the treatment of colorectal cancer. Eur J Med Chem 2024; 268:116250. [PMID: 38417218 DOI: 10.1016/j.ejmech.2024.116250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/15/2024] [Accepted: 02/15/2024] [Indexed: 03/01/2024]
Abstract
Ferritinophagy is a cellular process to release redox-active iron. Excessive activation of ferritinophagy ultimately results in ferroptosis characterized by ROS accumulation which plays important roles in the development and progression of cancer. Sinomenine, a main bioactive alkaloid from the traditional Chinese medicine Sinomenum acutum, inhibits the proliferation of cancer cells by promoting ROS production. Herein, new compounds were designed and synthesized through the stepwise optimization of sinomenine. Among them, D3-3 induced the production of lipid ROS, and significantly promoted colorectal cancer cells to release the ferrous ion in an autophagy-dependent manner. Moreover, D3-3 enhanced the interaction of FTH1-NCOA4, indicating the activation of ferritinophagy. In vivo experiments showed that D3-3 restrained tumor growth and promoted lipid peroxidation in the HCT-116 xenograft model. These findings demonstrated that D3-3 is an inducer of ferritinophagy, eventually triggering ferroptosis. Compound D3-3, as the first molecule to be definitively demonstrated to induce ferritinophagy, is worth further evaluation as a promising drug candidate in the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Ling Zhu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Chen Chen
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuxing Cai
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yalin Li
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Lijie Gong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Tianyu Zhu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Jianguang Luo
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
9
|
Maman A, Çiğdem S, Kaya İ, Demirtaş R, Ceylan O, Özmen S. Diagnostic value of FDG PET-CT in differentiating lung adenocarcinoma from squamous cell carcinoma. EJNMMI REPORTS 2024; 8:1. [PMID: 38748067 PMCID: PMC10962626 DOI: 10.1186/s41824-024-00187-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/19/2023] [Indexed: 05/19/2024]
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related deaths worldwide. The combination of fluorine-18 fluorodeoxyglucose positron emission tomography (18F-FDG PET) and computed tomography (CT) has a major impact on the diagnosis, staging, treatment planning and follow-up of lung cancer patients. The maximum standardized uptake value (SUVmax) is an easily performed and most widely used semi-quantitative index for the analysis of FDG PET images and estimation of metabolic activity. This study aimed to investigate the role of PET/CT in differentiating adenocarcinoma (ADC), the most common lung cancer, from squamous cell carcinoma (SCC) by comparing FDG uptake measured as SUVmax. RESULTS Between 2019 and 2022, 76 patients diagnosed with non-small cell lung cancer (NSCLC) at the Department of Pathology, Atatürk University Faculty of Medicine, with histopathologic evidence of adenocarcinoma or squamous cell carcinoma, underwent retrospective analysis using PET/CT scanning to measure PET parameters of the lesions and compare them with histopathology. Among 76 NSCLC patients included in the study, 43 (57%) were histopathologically diagnosed as ADC and 33 (43%) as SCC. SUVmax, SUVmean, metabolic tumor volume (MTV) and total lesion glycolysis (TLG) values of lesions in patients with SCC were statistically significantly higher than those in patients with ADC (p values 0.007, 0.009, 0.003 and 0.04, respectively). CONCLUSIONS Lung SCC has higher metabolic uptake values than ADC, and PET/CT can be used to differentiate them.
Collapse
Affiliation(s)
- Adem Maman
- Department of Nuclear Medicine, Faculty of Medicine, Atatürk University, Erzurum, Turkey.
| | - Sadık Çiğdem
- Vocational School of Health Services, Istanbul Aydın University, Istanbul, Turkey
| | - İdris Kaya
- Department of Radiology, Private Buhara Hospital, Erzurum, Turkey
| | - Rabia Demirtaş
- Department of Medical Pathology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Onur Ceylan
- Department of Medical Pathology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Sevilay Özmen
- Department of Medical Pathology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| |
Collapse
|
10
|
Saggese P, Pandey A, Alcaraz M, Fung E, Hall A, Yanagawa J, Rodriguez EF, Grogan TR, Giurato G, Nassa G, Salvati A, Shirihai OS, Weisz A, Dubinett SM, Scafoglio C. Glucose Deprivation Promotes Pseudohypoxia and Dedifferentiation in Lung Adenocarcinoma. Cancer Res 2024; 84:305-327. [PMID: 37934116 PMCID: PMC10790128 DOI: 10.1158/0008-5472.can-23-1148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/12/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
Increased utilization of glucose is a hallmark of cancer. Sodium-glucose transporter 2 (SGLT2) is a critical player in glucose uptake in early-stage and well-differentiated lung adenocarcinoma (LUAD). SGLT2 inhibitors, which are FDA approved for diabetes, heart failure, and kidney disease, have been shown to significantly delay LUAD development and prolong survival in murine models and in retrospective studies in diabetic patients, suggesting that they may be repurposed for lung cancer. Despite the antitumor effects of SGLT2 inhibition, tumors eventually escape treatment. Here, we studied the mechanisms of resistance to glucose metabolism-targeting treatments. Glucose restriction in LUAD and other tumors induced cancer cell dedifferentiation, leading to a more aggressive phenotype. Glucose deprivation caused a reduction in alpha-ketoglutarate (αKG), leading to attenuated activity of αKG-dependent histone demethylases and histone hypermethylation. The dedifferentiated phenotype depended on unbalanced EZH2 activity that suppressed prolyl-hydroxylase PHD3 and increased expression of hypoxia-inducible factor 1α (HIF1α), triggering epithelial-to-mesenchymal transition. Finally, a HIF1α-dependent transcriptional signature of genes upregulated by low glucose correlated with prognosis in human LUAD. Overall, this study furthers current knowledge of the relationship between glucose metabolism and cell differentiation in cancer, characterizing the epigenetic adaptation of cancer cells to glucose deprivation and identifying targets to prevent the development of resistance to therapies targeting glucose metabolism. SIGNIFICANCE Epigenetic adaptation allows cancer cells to overcome the tumor-suppressive effects of glucose restriction by inducing dedifferentiation and an aggressive phenotype, which could help design better metabolic treatments.
Collapse
Affiliation(s)
- Pasquale Saggese
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Aparamita Pandey
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Martín Alcaraz
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Eileen Fung
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
- Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Abbie Hall
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Jane Yanagawa
- Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Erika F. Rodriguez
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Tristan R. Grogan
- Division of General Internal Medicine and Health Services Research, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana,’ University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health – CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana,’ University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health – CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
| | - Annamaria Salvati
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana,’ University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health – CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
- Medical Genomics Program and Division of Onco-Hematology, AOU “S. Giovanni di Dio e Ruggi d'Aragona,” University of Salerno, Salerno, Italy
| | - Orian S. Shirihai
- Department of Medicine (Endocrinology), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana,’ University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health – CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
- Medical Genomics Program and Division of Onco-Hematology, AOU “S. Giovanni di Dio e Ruggi d'Aragona,” University of Salerno, Salerno, Italy
| | - Steven M. Dubinett
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Claudio Scafoglio
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
11
|
Cheng H, Zheng Y. Advances in macrophage and T cell metabolic reprogramming and immunotherapy in the tumor microenvironment. PeerJ 2024; 12:e16825. [PMID: 38239299 PMCID: PMC10795528 DOI: 10.7717/peerj.16825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/02/2024] [Indexed: 01/22/2024] Open
Abstract
Macrophages and T cells in the tumor microenvironment (TME) play an important role in tumorigenesis and progression. However, TME is also characterized by metabolic reprogramming, which may affect macrophage and metabolic activity of T cells and promote tumor escape. Immunotherapy is an approach to fight tumors by stimulating the immune system in the host, but requires support and modulation of cellular metabolism. In this process, the metabolic roles of macrophages and T cells become increasingly important, and their metabolic status and interactions play a critical role in the success of immunotherapy. Therefore, understanding the metabolic state of T cells and macrophages in the TME and the impact of metabolic reprogramming on tumor therapy will help optimize subsequent immunotherapy strategies.
Collapse
Affiliation(s)
- Hua Cheng
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yongbin Zheng
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
12
|
Zheng L, Liu H, Chen L, You X, Lv F, Fan H, Hui Q, Liu B, Wang X. Expression and Purification of FGFR1-Fc Fusion Protein and Its Effects on Human Lung Squamous Carcinoma. Appl Biochem Biotechnol 2024; 196:573-587. [PMID: 37160564 DOI: 10.1007/s12010-023-04542-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/11/2023]
Abstract
Molecular-targeted therapies for lung squamous cell carcinoma (LSCC) are limited mainly because targetable oncogenic aberrations are absent in LSCC. Recent genomic analyses have revealed that the fibroblast growth factor (FGF) signaling pathway plays a fundamental role in LSCC progression via cancer cell proliferation and angiogenesis. In the present study, we designed, expressed, and purified a fibroblast growth factor receptor fragment (FGFR1-Fc) fusion protein using NS/0 cells. In FGF2-FGFR1 overexpressed NCI-H1703 cells, the FGFR1-Fc fusion protein effectively inhibited proliferation and invasion and arrested the cell cycle at the G0-G1 phase. In NCI-H1703 cells treated with the FGFR1-Fc fusion protein, the phosphorylation levels of FGFR1, FRS2, ERK, and AKT were significantly reduced. Using an siRNA assay, we demonstrated that FGF2-FGFR1 is the major anti-tumor target of FGFR1-Fc fusion the FGFR1-Fc fusion protein, which also significantly inhibited proliferation and invasion by NCI-H1703 cells via the FGF2-FGFR1 signaling pathway. In addition, the FGFR1-Fc fusion protein significantly inhibited angiogenesis in an embryonic chorioallantoic membrane model. The FGFR1-Fc fusion protein may be an effective therapeutic candidate for LSCC.
Collapse
Affiliation(s)
- Lulu Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Zhejiang, 310000, Hangzhou, China
| | - Huan Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Lingfeng Chen
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, 310012, Hangzhou, China
| | - Xinyi You
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Fangyi Lv
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Haibing Fan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Qi Hui
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Baohua Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Department of Neurological Rehabilitation, The Second Asffiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| | - Xiaojie Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
13
|
Zhang C, Yu JJ, Yang C, Yuan ZL, Zeng H, Wang JJ, Shang S, Lv XX, Liu XT, Liu J, Xue Q, Cui B, Tan FW, Hua F. Wild-type IDH1 maintains NSCLC stemness and chemoresistance through activation of the serine biosynthetic pathway. Sci Transl Med 2023; 15:eade4113. [PMID: 38091408 DOI: 10.1126/scitranslmed.ade4113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/08/2023] [Indexed: 12/18/2023]
Abstract
Tumor-initiating cells (TICs) reprogram their metabolic features to meet their bioenergetic, biosynthetic, and redox demands. Our previous study established a role for wild-type isocitrate dehydrogenase 1 (IDH1WT) as a potential diagnostic and prognostic biomarker for non-small cell lung cancer (NSCLC), but how IDH1WT modulates NSCLC progression remains elusive. Here, we report that IDH1WT activates serine biosynthesis by enhancing the expression of phosphoglycerate dehydrogenase (PHGDH) and phosphoserine aminotransferase 1 (PSAT1), the first and second enzymes of de novo serine synthetic pathway. Augmented serine synthesis leads to GSH/ROS imbalance and supports pyrimidine biosynthesis, maintaining tumor initiation capacity and enhancing gemcitabine chemoresistance. Mechanistically, we identify that IDH1WT interacts with and stabilizes PHGDH and fragile X-related protein-1 (FXR1) by impeding their association with the E3 ubiquitin ligase parkin by coimmunoprecipitation assay and proximity ligation assay. Subsequently, stabilized FXR1 supports PSAT1 mRNA stability and translation, as determined by actinomycin D chase experiment and in vitro translation assay. Disrupting IDH1WT-PHGDH and IDH1WT-FXR1 interactions synergistically reduces NSCLC stemness and sensitizes NSCLC cells to gemcitabine and serine/glycine-depleted diet therapy in lung cancer xenograft models. Collectively, our findings offer insights into the role of IDH1WT in serine metabolism, highlighting IDH1WT as a potential therapeutic target for eradicating TICs and overcoming gemcitabine chemoresistance in NSCLC.
Collapse
Affiliation(s)
- Cheng Zhang
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, P.R. China
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, P.R. China
| | - Jiao-Jiao Yu
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, P.R. China
| | - Chen Yang
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, P.R. China
| | - Zhen-Long Yuan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P.R. China
| | - Hui Zeng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P.R. China
| | - Jun-Jian Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, P.R. China
| | - Shuang Shang
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, P.R. China
| | - Xiao-Xi Lv
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, P.R. China
| | - Xiao-Tong Liu
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, P.R. China
| | - Jing Liu
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, P.R. China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P.R. China
| | - Bing Cui
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, P.R. China
| | - Feng-Wei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P.R. China
| | - Fang Hua
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, P.R. China
| |
Collapse
|
14
|
Wu X, Ma Y, Wang L, Qin X. A Route for Investigating Psoriasis: From the Perspective of the Pathological Mechanisms and Therapeutic Strategies of Cancer. Int J Mol Sci 2023; 24:14390. [PMID: 37762693 PMCID: PMC10532365 DOI: 10.3390/ijms241814390] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/20/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
Psoriasis is an incurable skin disease that develops in about two-thirds of patients before the age of 40 and requires lifelong treatment; its pathological mechanisms have not been fully elucidated. The core pathological process of psoriasis is epidermal thickening caused by the excessive proliferation of epidermal keratinocytes, which is similar to the key feature of cancer; the malignant proliferation of cancer cells causes tumor enlargement, suggesting that there is a certain degree of commonality between psoriasis and cancer. This article reviews the pathological mechanisms that are common to psoriasis and cancer, including the interaction between cell proliferation and an abnormal immune microenvironment, metabolic reprogramming, and epigenetic reprogramming. In addition, there are common therapeutic agents and drug targets between psoriasis and cancer. Thus, psoriasis and cancer share a common pathological mechanisms-drug targets-therapeutic agents framework. On this basis, it is proposed that investigating psoriasis from a cancer perspective is beneficial to enriching the research strategies related to psoriasis.
Collapse
Affiliation(s)
- Xingkang Wu
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, China; (Y.M.); (L.W.)
| | | | | | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, China; (Y.M.); (L.W.)
| |
Collapse
|
15
|
Leshem Y, Dolev Y, Siegelmann-Danieli N, Sharman Moser S, Apter L, Chodick G, Nikolaevski-Berlin A, Shamai S, Merimsky O, Wolf I. Association between diabetes mellitus and reduced efficacy of pembrolizumab in non-small cell lung cancer. Cancer 2023; 129:2789-2797. [PMID: 37354065 DOI: 10.1002/cncr.34918] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 02/06/2023] [Accepted: 02/17/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Diabetes mellitus (DM) is a highly prevalent chronic metabolic disorder. Although DM has been associated with immune dysfunction, the effect of DM on the efficacy of immunotherapy is unknown. This study aimed to evaluate the impact of DM on the efficacy of pembrolizumab in metastatic non-small cell lung cancer (NSCLC). METHODS The authors reviewed the medical records of consecutive metastatic NSCLC patients treated with first-line pembrolizumab either alone or in combination with chemotherapy at a single tertiary center. For validation, a computerized data from Maccabi Healthcare Services, a 2.5-million-member state health service was used. RESULTS Of the 203 eligible patients, 51 (25%) had DM. Patients with DM had a significantly shorter median progression-free survival (PFS) (5.9 vs. 7.1 months, p = .004) and overall survival (OS) (12 vs. 21 months, p = .006). The shorter OS in diabetic patients was more pronounced when pembrolizumab was given alone (12 vs. 27 months, p = .03) than when combined with chemotherapy (14.3 vs. 19.4 months, p = .06). Multivariate analysis confirmed DM as an independent risk factor for shorter PFS (hazard ratio [HR], 1.67; 95% confidence interval [CI], 1.11-2.50, p = .01) and OS (HR, 1.73; 95% CI, 1.09-2.76, p = .02). In a validation cohort of 452 metastatic NSCLC patients, the time on pembrolizumab treatment was shorter in diabetic patients (p = .025), with only 19.6% of patients remaining on treatment at 12 months compared to 31.7% of the nondiabetic patients. CONCLUSIONS This study suggests immunotherapy is less beneficial in diabetic NSCLC patients. More work is needed to verify our findings and explore similar effects in other cancer entities.
Collapse
Affiliation(s)
- Yasmin Leshem
- Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yardenna Dolev
- Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Nava Siegelmann-Danieli
- Maccabi Institute for Research and Innovation (Maccabitech), Maccabi Healthcare Services, Tel Aviv, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sarah Sharman Moser
- Maccabi Institute for Research and Innovation (Maccabitech), Maccabi Healthcare Services, Tel Aviv, Israel
| | - Lior Apter
- Maccabi Institute for Research and Innovation (Maccabitech), Maccabi Healthcare Services, Tel Aviv, Israel
- Department of Health Systems Management, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Gabriel Chodick
- Maccabi Institute for Research and Innovation (Maccabitech), Maccabi Healthcare Services, Tel Aviv, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Sivan Shamai
- Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ofer Merimsky
- Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ido Wolf
- Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
16
|
Liu Y, Zhang X, Cheng F, Cao W, Geng Y, Chen Z, Wei W, Zhang L. Xanthatin induce DDP-resistance lung cancer cells apoptosis through regulation of GLUT1 mediated ROS accumulation. Drug Dev Res 2023; 84:1266-1278. [PMID: 37260173 DOI: 10.1002/ddr.22084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/09/2023] [Accepted: 05/19/2023] [Indexed: 06/02/2023]
Abstract
Chemoresistance to cisplatin (DDP) therapy is a major obstacle that needs to be overcome in treating lung cancer patients. Xanthatin has been reported to exhibit an antitumor effect on various cancers, but the function of xanthatin in DDP-resistance lung cancer remains unclear. The study aimed to explore the effect and mechanisms of xanthatin on proliferation, apoptosis, and migration in DDP-resistance lung cancer cells. In the present study, xanthatin suppresses the expression of glucose transporter 1 (GLUT1), attenuates the pentose phosphate pathway (PPP), and causes ROS accumulation and apoptosis, thereby mitigating the antioxidative capacity in DDP-resistance cells. Previous studies have shown that GLUT1 is associated with resistance to platinum drugs. We found that GLUT1 was significantly increased in the DDP-resistant lung cancer cell line compared to the parental cell line, and xanthatin significantly downregulated GLUT1 expression in DDP-resistant lung cancer cells. Notably, overexpression of GLUT1 significantly reduced the production of ROS and increased cellular NADPH/NADP+ and GSH/GSSG ratios. Thus, these results suggest that xanthatin induces DDP-resistance lung cancer cells apoptosis through regulation of GLUT1-mediated ROS accumulation. These findings might provide a possible strategy for the clinical treatment of DDP-resistant lung cancer.
Collapse
Affiliation(s)
- Yunxiao Liu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Xinge Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Fenting Cheng
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Cao
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yadi Geng
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhaolin Chen
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Lei Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
17
|
Yue SW, Liu HL, Su HF, Luo C, Liang HF, Zhang BX, Zhang W. m6A-regulated tumor glycolysis: new advances in epigenetics and metabolism. Mol Cancer 2023; 22:137. [PMID: 37582735 PMCID: PMC10426175 DOI: 10.1186/s12943-023-01841-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/05/2023] [Indexed: 08/17/2023] Open
Abstract
Glycolytic reprogramming is one of the most important features of cancer and plays an integral role in the progression of cancer. In cancer cells, changes in glucose metabolism meet the needs of self-proliferation, angiogenesis and lymphangiogenesis, metastasis, and also affect the immune escape, prognosis evaluation and therapeutic effect of cancer. The n6-methyladenosine (m6A) modification of RNA is widespread in eukaryotic cells. Dynamic and reversible m6A modifications are widely involved in the regulation of cancer stem cell renewal and differentiation, tumor therapy resistance, tumor microenvironment, tumor immune escape, and tumor metabolism. Lately, more and more evidences show that m6A modification can affect the glycolysis process of tumors in a variety of ways to regulate the biological behavior of tumors. In this review, we discussed the role of glycolysis in tumor genesis and development, and elaborated in detail the profound impact of m6A modification on different tumor by regulating glycolysis. We believe that m6A modified glycolysis has great significance and potential for tumor treatment.
Collapse
Affiliation(s)
- Shi-Wei Yue
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato‑Pancreatic‑Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Hai-Ling Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato‑Pancreatic‑Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Hong-Fei Su
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato‑Pancreatic‑Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Chu Luo
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato‑Pancreatic‑Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Hui-Fang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Hepato‑Pancreatic‑Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
| | - Bi-Xiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Hepato‑Pancreatic‑Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
| | - Wei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Hepato‑Pancreatic‑Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
| |
Collapse
|
18
|
Kokeza J, Strikic A, Ogorevc M, Kelam N, Vukoja M, Dilber I, Zekic Tomas S. The Effect of GLUT1 and HIF-1α Expressions on Glucose Uptake and Patient Survival in Non-Small-Cell Lung Carcinoma. Int J Mol Sci 2023; 24:10575. [PMID: 37445752 DOI: 10.3390/ijms241310575] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Lung cancer is the second-most-common cancer while being the leading cause of cancer deaths worldwide. It has been found that glucose transporter 1 (GLUT1) and hypoxia-inducible factor 1α (HIF-1α) are overexpressed in various malignancies and that they correlate with the maximum standard uptake values (SUVmax) on 18F-fluorodeoxyglucose-positron emission tomography/computed tomography (18F-FDG PET/CT) and poor prognosis. In this study, we aim to evaluate the relationship between the SUVmax, GLUT1, and HIF-1α expression with primary tumor size, histological type, lymph node metastases, and patient survival. Of the 48 patients with non-small-cell lung cancer, those with squamous cell carcinomas (SCCs) had significantly higher GLUT1 and HIF-1α immunohistochemical expressions in comparison to adenocarcinomas (ACs), while there was no statistically significant difference in FDG accumulation between them. No significant correlation was noted between either GLUT1 or HIF-1α protein expression and FDG uptake and overall survival. However, an analysis of tumor transcriptomics showed a significant difference in overall survival depending on mRNA expression; patients with SCC and high HIF-1α levels survived longer compared to those with low HIF-1α levels, while patients with AC and low GLUT1 levels had a higher average survival time than those with high GLUT1 levels. Further studies are needed to determine the prognostic value of the expression of these factors depending on the histologic type.
Collapse
Affiliation(s)
- Josipa Kokeza
- Department of Pulmonology, University Hospital of Split, Spinčićeva 1, 21000 Split, Croatia
| | - Ante Strikic
- Department of Oncology and Radiotherapy, University Hospital of Split, Spinčićeva 1, 21000 Split, Croatia
| | - Marin Ogorevc
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia
| | - Martina Vukoja
- Laboratory of Morphology, Department of Histology and Embryology, School of Medicine, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
| | - Ivo Dilber
- Department of Oncology and Nuclear Medicine, General Hospital Zadar, Ul. Bože Peričića 5, 23000 Zadar, Croatia
| | - Sandra Zekic Tomas
- Department of Pathology, Forensic Medicine and Cytology, University Hospital of Split, Spinčićeva 1, 21000 Split, Croatia
- Department of Pathology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia
| |
Collapse
|
19
|
Liu X, Qin H, Zhang L, Jia C, Chao Z, Qin X, Zhang H, Chen C. Hyperoxia induces glucose metabolism reprogramming and intracellular acidification by suppressing MYC/MCT1 axis in lung cancer. Redox Biol 2023; 61:102647. [PMID: 36867943 PMCID: PMC10011425 DOI: 10.1016/j.redox.2023.102647] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
The perils and promises of inspiratory hyperoxia (IH) in oncology are still controversial, especially for patients with lung cancer. Increasing evidence shows that hyperoxia exposure is relevant to the tumor microenvironment. However, the detailed role of IH on the acid-base homeostasis of lung cancer cells remains unclear. In this study, the effects of 60% oxygen exposure on intra- and extracellular pH were systematically evaluated in H1299 and A549 cells. Our data indicate that hyperoxia exposure reduces intracellular pH, which might be expected to reduce the proliferation, invasion, and epithelial-to-mesenchymal transition of lung cancer cells. RNA sequencing, Western blot, and PCR analysis reveal that monocarboxylate transporter 1 (MCT1) mediates intracellular lactate accumulation and intracellular acidification of H1299 and A549 cells at 60% oxygen exposure. In vivo studies further demonstrate that MCT1 knockdown dramatically reduces lung cancer growth, invasion, and metastasis. The results of luciferase and ChIP-qPCR assays further confirm that MYC is a transcription factor of MCT1, and PCR and Western blot assays confirm that MYC is downregulated under hyperoxic conditions. Collectively, our data reveal that hyperoxia can suppress the MYC/MCT1 axis and cause the accumulation of lactate and intracellular acidification, thereby retarding tumor growth and metastasis.
Collapse
Affiliation(s)
- Xiucheng Liu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China; Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| | - Hao Qin
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China; Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, China
| | - Li Zhang
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| | - Caili Jia
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| | - Zhixiang Chao
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| | - Xichun Qin
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| | - Hao Zhang
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China; Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China.
| |
Collapse
|
20
|
Chang YC, Chan MH, Yang YF, Li CH, Hsiao M. Glucose transporter 4: Insulin response mastermind, glycolysis catalyst and treatment direction for cancer progression. Cancer Lett 2023; 563:216179. [PMID: 37061122 DOI: 10.1016/j.canlet.2023.216179] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 04/17/2023]
Abstract
The glucose transporter family (GLUT) consists of fourteen members. It is responsible for glucose homeostasis and glucose transport from the extracellular space to the cell cytoplasm to further cascade catalysis. GLUT proteins are encoded by the solute carrier family 2 (SLC2) genes and are members of the major facilitator superfamily of membrane transporters. Moreover, different GLUTs also have their transporter kinetics and distribution, so each GLUT member has its uniqueness and importance to play essential roles in human physiology. Evidence from many studies in the field of diabetes showed that GLUT4 travels between the plasma membrane and intracellular vesicles (GLUT4-storage vesicles, GSVs) and that the PI3K/Akt pathway regulates this activity in an insulin-dependent manner or by the AMPK pathway in response to muscle contraction. Moreover, some published results also pointed out that GLUT4 mediates insulin-dependent glucose uptake. Thus, dysfunction of GLUT4 can induce insulin resistance, metabolic reprogramming in diverse chronic diseases, inflammation, and cancer. In addition to the relationship between GLUT4 and insulin response, recent studies also referred to the potential upstream transcription factors that can bind to the promoter region of GLUT4 to regulating downstream signals. Combined all of the evidence, we conclude that GLUT4 has shown valuable unknown functions and is of clinical significance in cancers, which deserves our in-depth discussion and design compounds by structure basis to achieve therapeutic effects. Thus, we intend to write up a most updated review manuscript to include the most recent and critical research findings elucidating how and why GLUT4 plays an essential role in carcinogenesis, which may have broad interests and impacts on this field.
Collapse
Affiliation(s)
- Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Hsien Chan
- Department of Biomedical Imaging and Radiological Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Fang Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chien-Hsiu Li
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan; Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
21
|
Paik PK, Fan PD, Qeriqi B, Namakydoust A, Daly B, Ahn L, Kim R, Plodkowski A, Ni A, Chang J, Fanaroff R, Ladanyi M, de Stanchina E, Rudin CM. Targeting NFE2L2/KEAP1 Mutations in Advanced NSCLC With the TORC1/2 Inhibitor TAK-228. J Thorac Oncol 2023; 18:516-526. [PMID: 36240971 PMCID: PMC10500888 DOI: 10.1016/j.jtho.2022.09.225] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/21/2022] [Accepted: 09/25/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION Increased insight into the mutational landscape of squamous cell lung cancers (LUSCs) in the past decade has not translated into effective targeted therapies for patients with this disease. NRF2, encoded by NFE2L2, and its upstream regulator, KEAP1, control key aspects of redox balance and are frequently mutated in NSCLCs. METHODS Here, we describe the specific potent activity of TAK-228, a TORC1/2 inhibitor, in NSCLC models harboring NRF2-activating alterations and results of a phase 2 clinical trial of TAK-228 in patients with advanced NSCLC harboring NRF2-activating alterations including three cohorts (NFE2L2-mutated LUSC, KEAP1-mutated LUSC, KRAS/NFE2L2- or KEAP1-mutated NSCLC). RESULTS TAK-228 was most efficacious in a LUSC cohort with NFE2L2 alterations; the overall response rate was 25% and median progression-free survival was 8.9 months. Additional data suggest that concurrent inhibition of glutaminase with the glutaminase inhibitor CB-839 might overcome metabolic resistance to therapy in these patients. CONCLUSIONS TAK-228 has single-agent activity in patients with NRF2-activated LUSC. This study reframes oncogenic alterations as biologically relevant based on their downstream effects on metabolism. This trial represents, to the best of our knowledge, the first successful attempt at metabolically targeting NSCLC and identifies a promising targeted therapy for patients with LUSC, who are bereft of genotype-directed therapies.
Collapse
Affiliation(s)
- Paul K Paik
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York.
| | - Pang-Dian Fan
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Current Affiliation, Daiichi Sankyo, Inc., Basking Ridge, New Jersey
| | | | - Azadeh Namakydoust
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bobby Daly
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Linda Ahn
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rachel Kim
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew Plodkowski
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ai Ni
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rachel Fanaroff
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Charles M Rudin
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| |
Collapse
|
22
|
Park SY, Cho DG, Shim BY, Cho U. Relationship between Systemic Inflammatory Markers, GLUT1 Expression, and Maximum 18F-Fluorodeoxyglucose Uptake in Non-Small Cell Lung Carcinoma and Their Prognostic Significance. Diagnostics (Basel) 2023; 13:diagnostics13061013. [PMID: 36980320 PMCID: PMC10047418 DOI: 10.3390/diagnostics13061013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Factors involved in inflammation and cancer interact in various ways with each other, and biomarkers of systemic inflammation may have a prognostic value in cancer. Glucose transporter 1 (GLUT1) plays a pivotal role in glucose transport and metabolism and it is aberrantly expressed in various cancer types. We evaluated the differential expression of GLUT1, along with 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) in non-small-cell lung cancer (NSCLC), and then analyzed their prognostic significance. METHODS A total of 163 patients with resectable NSCLC were included in this study. Tumor sections were immunohistochemically stained for GLUT1 and GLUT3. Maximum standardized uptake value (SUVmax) was measured by preoperative FDG-PET, and neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), and lymphocyte-monocyte ratio (LMR) were derived from pretreatment blood count. RESULTS GLUT1 and GLUT3 was positively expressed in 74.8% and 6.1% of the NSCLC tissues, respectively. GLUT1 expression was significantly correlated with squamous cell carcinoma histology, poor differentiation, high pathologic stage, old age, male, smoking, and high SUVmax (>7) (all p < 0.05). The squamous cell carcinoma and smoker group also showed significantly higher SUVmax (both p < 0.001). Systemic inflammation markers, including NLR, PLR, and LMR, were positively correlated with high SUVmax (all p < 0.05). High GLUT1 expression, high SUVmax, high NLR, and low LMR, were significantly associated with poor overall survival in patients with NSCLC. However, in the multivariate survival analysis, LMR was an independent prognostic factor overall (HR 1.86, 95% CI 1.05-3.3) and for the stage I/II cohort (HR 2.3, 95% CI 1.24-4.3) (all p < 0.05). CONCLUSIONS Systemic inflammatory markers-NLR, PLR, and LMR are strongly correlated with the SUVmax and are indicators of aggressive tumor behavior. Specifically, LMR is a promising prognostic biomarker in NSCLC patients.
Collapse
Affiliation(s)
- Sonya Youngju Park
- Department of Nuclear Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Deog-Gon Cho
- Department of Thoracic Surgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Byoung-Yong Shim
- Division of Medical Oncology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Uiju Cho
- Department of Pathology, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
23
|
Han M, Bushong EA, Segawa M, Tiard A, Wong A, Brady MR, Momcilovic M, Wolf DM, Zhang R, Petcherski A, Madany M, Xu S, Lee JT, Poyurovsky MV, Olszewski K, Holloway T, Gomez A, John MS, Dubinett SM, Koehler CM, Shirihai OS, Stiles L, Lisberg A, Soatto S, Sadeghi S, Ellisman MH, Shackelford DB. Spatial mapping of mitochondrial networks and bioenergetics in lung cancer. Nature 2023; 615:712-719. [PMID: 36922590 PMCID: PMC10033418 DOI: 10.1038/s41586-023-05793-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 02/03/2023] [Indexed: 03/17/2023]
Abstract
Mitochondria are critical to the governance of metabolism and bioenergetics in cancer cells1. The mitochondria form highly organized networks, in which their outer and inner membrane structures define their bioenergetic capacity2,3. However, in vivo studies delineating the relationship between the structural organization of mitochondrial networks and their bioenergetic activity have been limited. Here we present an in vivo structural and functional analysis of mitochondrial networks and bioenergetic phenotypes in non-small cell lung cancer (NSCLC) using an integrated platform consisting of positron emission tomography imaging, respirometry and three-dimensional scanning block-face electron microscopy. The diverse bioenergetic phenotypes and metabolic dependencies we identified in NSCLC tumours align with distinct structural organization of mitochondrial networks present. Further, we discovered that mitochondrial networks are organized into distinct compartments within tumour cells. In tumours with high rates of oxidative phosphorylation (OXPHOSHI) and fatty acid oxidation, we identified peri-droplet mitochondrial networks wherein mitochondria contact and surround lipid droplets. By contrast, we discovered that in tumours with low rates of OXPHOS (OXPHOSLO), high glucose flux regulated perinuclear localization of mitochondria, structural remodelling of cristae and mitochondrial respiratory capacity. Our findings suggest that in NSCLC, mitochondrial networks are compartmentalized into distinct subpopulations that govern the bioenergetic capacity of tumours.
Collapse
Affiliation(s)
- Mingqi Han
- Pulmonary and Critical Care Medicine, David Geffen School of Medicine (DGSOM), University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Eric A Bushong
- Department of Neurosciences, University of California San Diego (UCSD), San Diego, CA, USA
- National Center for Microscopy and Imaging Research, UCSD, San Diego, CA, USA
| | | | | | - Alex Wong
- Department of Computer Science, Yale University, New Haven, CT, USA
| | - Morgan R Brady
- Pulmonary and Critical Care Medicine, David Geffen School of Medicine (DGSOM), University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Milica Momcilovic
- Pulmonary and Critical Care Medicine, David Geffen School of Medicine (DGSOM), University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Dane M Wolf
- University of Cambridge, Cambridge, UK
- Imperial College, London, UK
| | - Ralph Zhang
- Pulmonary and Critical Care Medicine, David Geffen School of Medicine (DGSOM), University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | | | - Matthew Madany
- Department of Neurosciences, University of California San Diego (UCSD), San Diego, CA, USA
- National Center for Microscopy and Imaging Research, UCSD, San Diego, CA, USA
| | - Shili Xu
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Jason T Lee
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
- Molecular Imaging Program, Department of Radiology, Stanford University, Stanford, CA, USA
| | | | | | - Travis Holloway
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
| | - Adrian Gomez
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - Maie St John
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
- Department of Head and Neck Surgery, DGSOM UCLA, Los Angeles, CA, USA
| | - Steven M Dubinett
- Pulmonary and Critical Care Medicine, David Geffen School of Medicine (DGSOM), University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, DGSOM UCLA, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Carla M Koehler
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA, USA
| | - Orian S Shirihai
- Department of Endocrinology, DGSOM UCLA, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Linsey Stiles
- Department of Endocrinology, DGSOM UCLA, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
| | - Aaron Lisberg
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
- Department Hematology and Oncology, DGSOM UCLA, Los Angeles, CA, USA
| | - Stefano Soatto
- Department of Computer Science, UCLA, Los Angeles, CA, USA
| | - Saman Sadeghi
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Mark H Ellisman
- Department of Neurosciences, University of California San Diego (UCSD), San Diego, CA, USA
- National Center for Microscopy and Imaging Research, UCSD, San Diego, CA, USA
| | - David B Shackelford
- Pulmonary and Critical Care Medicine, David Geffen School of Medicine (DGSOM), University of California Los Angeles (UCLA), Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Kathote G, Ma Q, Angulo G, Chen H, Jakkamsetti V, Dobariya A, Good LB, Posner B, Park JY, Pascual JM. Identification of Glucose Transport Modulators In Vitro and Method for Their Deep Learning Neural Network Behavioral Evaluation in Glucose Transporter 1-Deficient Mice. J Pharmacol Exp Ther 2023; 384:393-405. [PMID: 36635085 DOI: 10.1124/jpet.122.001428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/07/2022] [Accepted: 12/27/2022] [Indexed: 01/14/2023] Open
Abstract
Metabolic flux augmentation via glucose transport activation may be desirable in glucose transporter 1 (Glut1) deficiency syndrome (G1D) and dementia, whereas suppression might prove useful in cancer. Using lung adenocarcinoma cells that predominantly express Glut1 relative to other glucose transporters, we screened 9,646 compounds for effects on the accumulation of an extracellularly applied fluorescent glucose analog. Five drugs currently prescribed for unrelated indications or preclinically characterized robustly enhanced intracellular fluorescence. Additionally identified were 37 novel activating and nine inhibitory compounds lacking previous biologic characterization. Because few glucose-related mechanistic or pharmacological studies were available for these compounds, we developed a method to quantify G1D mouse behavior to infer potential therapeutic value. To this end, we designed a five-track apparatus to record and evaluate spontaneous locomotion videos. We applied this to a G1D mouse model that replicates the ataxia and other manifestations cardinal to the human disorder. Because the first two drugs that we examined in this manner (baclofen and acetazolamide) exerted various impacts on several gait aspects, we used deep learning neural networks to more comprehensively assess drug effects. Using this method, 49 locomotor parameters differentiated G1D from control mice. Thus, we used parameter modifiability to quantify efficacy on gait. We tested this by measuring the effects of saline as control and glucose as G1D therapy. The results indicate that this in vivo approach can estimate preclinical suitability from the perspective of G1D locomotion. This justifies the use of this method to evaluate our drugs or other interventions and sort candidates for further investigation. SIGNIFICANCE STATEMENT: There are few or no activators and few clinical inhibitors of glucose transport. Using Glut1-rich cells exposed to a glucose analog, we identified, in highthroughput fashion, a series of novel modulators. Some were drugs used to modify unrelated processes and some represented large but little studied chemical compound families. To facilitate their preclinical efficacy characterization regardless of potential mechanism of action, we developed a gait testing platform for deep learning neural network analysis of drug impact on Glut1-deficient mouse locomotion.
Collapse
Affiliation(s)
- Gauri Kathote
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Qian Ma
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Gustavo Angulo
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hong Chen
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Vikram Jakkamsetti
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Aksharkumar Dobariya
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Levi B Good
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bruce Posner
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jason Y Park
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas.
| | - Juan M Pascual
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
25
|
Phosphohistidine signaling promotes FAK-RB1 interaction and growth factor-independent proliferation of esophageal squamous cell carcinoma. Oncogene 2023; 42:449-460. [PMID: 36513743 DOI: 10.1038/s41388-022-02568-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 11/08/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Current clinical therapies targeting receptor tyrosine kinases including focal adhesion kinase (FAK) have had limited or no effect on esophageal squamous cell carcinoma (ESCC). Unlike esophageal adenocarcinomas, ESCC acquire glucose in excess of their anabolic need. We recently reported that glucose-induced growth factor-independent proliferation requires the phosphorylation of FAKHis58. Here, we confirm His58 phosphorylation in FAK immunoprecipitates of glucose-stimulated, serum-starved ESCC cells using antibodies specific for 3-phosphohistidine and mass spectrometry. We also confirm a role for the histidine kinase, NME1, in glucose-induced FAKpoHis58 and ESCC cell proliferation, correlating with increased levels of NME1 in ESCC tumors versus normal esophageal tissues. Unbiased screening identified glucose-induced retinoblastoma transcriptional corepressor 1 (RB1) binding to FAK, mediated through a "LxCxE" RB1-binding motif in FAK's FERM domain. Importantly, in the absence of growth factors, glucose increased FAK scaffolding of RB1 in the cytoplasm, correlating with increased ESCC G1→S phase transition. Our data strongly suggest that this glucose-mediated mitogenic pathway is novel and represents a unique targetable opportunity in ESCC.
Collapse
|
26
|
Saggese P, Pandey A, Fung E, Hall A, Yanagawa J, Rodriguez EF, Grogan TR, Giurato G, Nassa G, Salvati A, Weisz A, Dubinett SM, Scafoglio C. Glucose deprivation promotes pseudo-hypoxia and de-differentiation in lung adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526207. [PMID: 36778362 PMCID: PMC9915520 DOI: 10.1101/2023.01.30.526207] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Increased utilization of glucose is a hallmark of cancer. Several studies are investigating the efficacy of glucose restriction by glucose transporter blockade or glycolysis inhibition. However, the adaptations of cancer cells to glucose restriction are unknown. Here, we report the discovery that glucose restriction in lung adenocarcinoma (LUAD) induces cancer cell de-differentiation, leading to a more aggressive phenotype. Glucose deprivation causes a reduction in alpha-ketoglutarate (αKG), leading to attenuated activity of αKG-dependent histone demethylases and histone hypermethylation. We further show that this de-differentiated phenotype depends on unbalanced EZH2 activity, causing inhibition of prolyl-hydroxylase PHD3 and increased expression of hypoxia inducible factor 1α (HIF1α), triggering epithelial to mesenchymal transition. Finally, we identified an HIF1α-dependent transcriptional signature with prognostic significance in human LUAD. Our studies further current knowledge of the relationship between glucose metabolism and cell differentiation in cancer, characterizing the epigenetic adaptation of cancer cells to glucose deprivation and identifying novel targets to prevent the development of resistance to therapies targeting glucose metabolism.
Collapse
Affiliation(s)
- Pasquale Saggese
- Division of Pulmonary Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Aparamita Pandey
- Division of Pulmonary Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Eileen Fung
- Division of Pulmonary Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Division of Thoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Abbie Hall
- Division of Pulmonary Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jane Yanagawa
- Division of Thoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Erika F. Rodriguez
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Tristan R. Grogan
- Division of General Internal Medicine and Health Services Research, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health - CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health - CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
| | - Annamaria Salvati
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health - CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
- Medical Genomics Program and Division of Onco-Hematology, AOU “S. Giovanni di Dio e Ruggi d’Aragona”, University of Salerno, Salerno, Italy
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health - CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
- Medical Genomics Program and Division of Onco-Hematology, AOU “S. Giovanni di Dio e Ruggi d’Aragona”, University of Salerno, Salerno, Italy
| | - Steven M. Dubinett
- Division of Pulmonary Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Claudio Scafoglio
- Division of Pulmonary Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
27
|
Ghezzi C, Chen BY, Damoiseaux R, Clark PM. Pacritinib inhibits glucose consumption in squamous cell lung cancer cells by targeting FLT3. Sci Rep 2023; 13:1442. [PMID: 36697489 PMCID: PMC9876922 DOI: 10.1038/s41598-023-28576-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Squamous cell lung cancer maintains its growth through elevated glucose consumption, but selective glucose consumption inhibitors are lacking. Here, we discovered using a high-throughput screen new compounds that block glucose consumption in three squamous cell lung cancer cell lines and identified 79 compounds that block glucose consumption in one or more of these cell lines. Based on its ability to block glucose consumption in all three cell lines, pacritinib, an inhibitor of FMS Related Receptor Tyrosine Kinase 3 (FLT3) and Janus Kinase 2 (JAK2), was further studied. Pacritinib decreased glucose consumption in squamous cell lung cancer cells in cell culture and in vivo without affecting glucose consumption in healthy tissues. Pacritinib blocked hexokinase activity, and Hexokinase 1 and 2 mRNA and protein expression. Overexpression of Hexokinase 1 blocked the ability of pacritinib to inhibit glucose consumption in squamous cell lung cancer cells. Overexpression of FLT3 but not JAK2 significantly increased glucose consumption and blocked the ability of pacritinib to inhibit glucose consumption in squamous cell lung cancer cells. Additional FLT3 inhibitors blocked glucose consumption in squamous cell lung cancer cells. Our study identifies FLT3 inhibitors as a new class of inhibitors that can block glucose consumption in squamous cell lung cancer.
Collapse
Affiliation(s)
- Chiara Ghezzi
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Box 951770, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Bao Ying Chen
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Box 951770, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Robert Damoiseaux
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Box 951770, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Peter M Clark
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Box 951770, Los Angeles, CA, 90095, USA.
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
28
|
Pieri V, Gallotti AL, Drago D, Cominelli M, Pagano I, Conti V, Valtorta S, Coliva A, Lago S, Michelatti D, Massimino L, Ungaro F, Perani L, Spinelli A, Castellano A, Falini A, Zippo A, Poliani PL, Moresco RM, Andolfo A, Galli R. Aberrant L-Fucose Accumulation and Increased Core Fucosylation Are Metabolic Liabilities in Mesenchymal Glioblastoma. Cancer Res 2023; 83:195-218. [PMID: 36409826 DOI: 10.1158/0008-5472.can-22-0677] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/28/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022]
Abstract
Glioblastoma (GBM) is a common and deadly form of brain tumor in adults. Dysregulated metabolism in GBM offers an opportunity to deploy metabolic interventions as precise therapeutic strategies. To identify the molecular drivers and the modalities by which different molecular subgroups of GBM exploit metabolic rewiring to sustain tumor progression, we interrogated the transcriptome, the metabolome, and the glycoproteome of human subgroup-specific GBM sphere-forming cells (GSC). L-fucose abundance and core fucosylation activation were elevated in mesenchymal (MES) compared with proneural GSCs; this pattern was retained in subgroup-specific xenografts and in subgroup-affiliated human patient samples. Genetic and pharmacological inhibition of core fucosylation significantly reduced tumor growth in MES GBM preclinical models. Liquid chromatography-mass spectrometry (LC-MS)-based glycoproteomic screening indicated that most MES-restricted core-fucosylated proteins are involved in therapeutically relevant GBM pathological processes, such as extracellular matrix interaction, cell adhesion, and integrin-mediated signaling. Selective L-fucose accumulation in MES GBMs was observed using preclinical minimally invasive PET, implicating this metabolite as a potential subgroup-restricted biomarker.Overall, these findings indicate that L-fucose pathway activation in MES GBM is a subgroup-specific dependency that could provide diagnostic markers and actionable therapeutic targets. SIGNIFICANCE Metabolic characterization of subgroup-specific glioblastoma (GBM) sphere-forming cells identifies the L-fucose pathway as a vulnerability restricted to mesenchymal GBM, disclosing a potential precision medicine strategy for targeting cancer metabolism.
Collapse
Affiliation(s)
- Valentina Pieri
- Division of Neuroscience, Neural Stem Cell Biology Unit, IRCCS San Raffaele Hospital, Milan, Italy.,Neuroradiology Unit and CERMAC, Vita-Salute San Raffaele University and IRCCS San Raffaele Hospital, Milan, Italy
| | - Alberto L Gallotti
- Division of Neuroscience, Neural Stem Cell Biology Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Denise Drago
- ProMeFa, Center for Omics Sciences, IRCCS San Raffaele Hospital, Milan, Italy
| | - Manuela Cominelli
- Molecular and Translational Medicine Department, Pathology Unit, University of Brescia, Brescia, Italy
| | - Ilaria Pagano
- Division of Neuroscience, Neural Stem Cell Biology Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Valentina Conti
- Division of Neuroscience, Neural Stem Cell Biology Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Silvia Valtorta
- Nuclear Medicine and PET Cyclotron Center, IRCCS San Raffaele Hospital, Milan, Italy
| | - Angela Coliva
- Nuclear Medicine and PET Cyclotron Center, IRCCS San Raffaele Hospital, Milan, Italy
| | - Sara Lago
- Department of Cellular, Computational and Integrative Biology (CIBIO), Laboratory of Chromatin Biology & Epigenetics, University of Trento, Trento, Italy
| | - Daniela Michelatti
- Department of Cellular, Computational and Integrative Biology (CIBIO), Laboratory of Chromatin Biology & Epigenetics, University of Trento, Trento, Italy
| | - Luca Massimino
- Gastroenterology and Endoscopy Department, Experimental Gastroenterology Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Federica Ungaro
- Gastroenterology and Endoscopy Department, Experimental Gastroenterology Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Laura Perani
- Experimental Imaging Center, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Antonella Castellano
- Neuroradiology Unit and CERMAC, Vita-Salute San Raffaele University and IRCCS San Raffaele Hospital, Milan, Italy
| | - Andrea Falini
- Neuroradiology Unit and CERMAC, Vita-Salute San Raffaele University and IRCCS San Raffaele Hospital, Milan, Italy
| | - Alessio Zippo
- Department of Cellular, Computational and Integrative Biology (CIBIO), Laboratory of Chromatin Biology & Epigenetics, University of Trento, Trento, Italy
| | - Pietro L Poliani
- Molecular and Translational Medicine Department, Pathology Unit, University of Brescia, Brescia, Italy
| | - Rosa Maria Moresco
- Nuclear Medicine and PET Cyclotron Center, IRCCS San Raffaele Hospital, Milan, Italy.,Institute of Bioimaging and Molecular Physiology (IBFM), CNR, Segrate, Italy.,Department of Medicine and Surgery and Tecnomed Foundation, University of Milano-Bicocca, Monza, Italy
| | - Annapaola Andolfo
- ProMeFa, Center for Omics Sciences, IRCCS San Raffaele Hospital, Milan, Italy
| | - Rossella Galli
- Division of Neuroscience, Neural Stem Cell Biology Unit, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
29
|
Huang X, Zhang F, Lin J, Lin S, Shen G, Chen X, Chen W. Systematically analyzed molecular characteristics of lung adenocarcinoma using metabolism-related genes classification. Genet Mol Biol 2023; 45:e20220121. [PMID: 36622242 PMCID: PMC9830935 DOI: 10.1590/1678-4685-gmb-2022-0121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 11/06/2022] [Indexed: 01/10/2023] Open
Abstract
High heterogeneity of lung adenocarcinoma (LUAD) is a major clinical challenge. This study aims to characterize the molecular features of LUAD through classification based on metabolism-related genes. A total of 500 LUAD samples from The Cancer Genome Atlas (TCGA) and 612 from Gene Expression Omnibus (GEO) were integrated with 2,753 metabolism-related genes to determine the molecular classification. Systematic bioinformatics analysis was used to conduct correlation analysis between metabolism-related classification and molecular characteristics of LUAD. LUAD patients were divided into three molecular clusters (C1-C3). Survival analysis revealed that C1 and C2 showed good and poor prognoses, respectively. Associational analysis of classification and molecular characteristics revealed that C1 was associated with low pathological stage, metabolic pathways, high metabolic process, active immune process and checkpoint, sensitive drug response, as well as a low genetic mutation. Nevertheless, C2 was associated with high pathological stage, carcinogenic pathways, low metabolic process, inactive immune signatures, resistant drug response, and frequent genetic mutation. Eventually, a classifier with 60 metabolic genes was constructed, confirming the robustness of molecular classification on LUAD. Our findings promote the understanding of LUAD molecular characteristics, and the research data may be used for providing information be helpful for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaoming Huang
- The Affiliated Hospital of Southern Medical University, People’s Hospital of Longhua, Department of respiratory medicine, Shenzhen, China
| | - Feng Zhang
- The First Affiliated Hospital of Jinan University, Department of Intensive Care Unit, Guangzhou, China
| | - Junqi Lin
- The Affiliated Hospital of Southern Medical University, People’s Hospital of Longhua, Department of respiratory medicine, Shenzhen, China
| | - Shaoming Lin
- The Affiliated Hospital of Southern Medical University, People’s Hospital of Longhua, Department of respiratory medicine, Shenzhen, China
| | - Guanle Shen
- The Affiliated Hospital of Southern Medical University, People’s Hospital of Longhua, Department of respiratory medicine, Shenzhen, China
| | - Xiaozhu Chen
- The Affiliated Hospital of Southern Medical University, People’s Hospital of Longhua, Department of Medical Ultrasound Department, Shenzhen, China
| | - Wenbiao Chen
- The Affiliated Hospital of Southern Medical University, People’s Hospital of Longhua, Department of respiratory medicine, Shenzhen, China
| |
Collapse
|
30
|
Li B, Sun S, Li JJ, Yuan JP, Sun SR, Wu Q. Adipose tissue macrophages: implications for obesity-associated cancer. Mil Med Res 2023; 10:1. [PMID: 36593475 PMCID: PMC9809128 DOI: 10.1186/s40779-022-00437-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 12/12/2022] [Indexed: 01/04/2023] Open
Abstract
Obesity is one of the most serious global health problems, with an incidence that increases yearly and coincides with the development of cancer. Adipose tissue macrophages (ATMs) are particularly important in this context and contribute to linking obesity-related inflammation and tumor progression. However, the functions of ATMs on the progression of obesity-associated cancer remain unclear. In this review, we describe the origins, phenotypes, and functions of ATMs. Subsequently, we summarize the potential mechanisms on the reprogramming of ATMs in the obesity-associated microenvironment, including the direct exchange of dysfunctional metabolites, inordinate cytokines and other signaling mediators, transfer of extracellular vesicle cargo, and variations in the gut microbiota and its metabolites. A better understanding of the properties and functions of ATMs under conditions of obesity will lead to the development of new therapeutic interventions for obesity-related cancer.
Collapse
Affiliation(s)
- Bei Li
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Juan-Juan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jing-Ping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Sheng-Rong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China. .,Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
31
|
Baines DL, Vasiljevs S, Kalsi KK. Getting sweeter: new evidence for glucose transporters in specific cell types of the airway? Am J Physiol Cell Physiol 2023; 324:C153-C166. [PMID: 36409177 PMCID: PMC9829484 DOI: 10.1152/ajpcell.00140.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
New technologies such as single-cell RNA sequencing (scRNAseq) has enabled identification of the mRNA transcripts expressed by individual cells. This review provides insight from recent scRNAseq studies on the expression of glucose transporters in the epithelial cells of the airway epithelium from trachea to alveolus. The number of studies analyzed was limited, not all reported the full range of glucose transporters and there were differences between cells freshly isolated from the airways and those grown in vitro. Furthermore, glucose transporter mRNA transcripts were expressed at lower levels than other epithelial marker genes. Nevertheless, these studies highlighted that there were differences in cellular expression of glucose transporters. GLUT1 was the most abundant of the broadly expressed transporters that included GLUT8, 10, and 13. GLUT9 transcripts were more common in basal cells and GLUT12 in ionocytes/ciliated cells. In addition to alveolar cells, SGLT1 transcripts were present in secretory cells. GLUT3 mRNA transcripts were expressed in a cell cluster that expressed monocarboxylate (MCT2) transporters. Such distributions likely underlie cell-specific metabolic requirements to support proliferation, ion transport, mucous secretion, environment sensing, and airway glucose homeostasis. These studies have also highlighted the role of glucose transporters in the movement of dehydroascorbic acid/vitamin C/myoinositol/urate, which are factors important to the innate immune properties of the airways. Discrepancies remain between detection of mRNAs, protein, and function of glucose transporters in the lungs. However, collation of the data from further scRNAseq studies may provide a better consensus and understanding, supported by qPCR, immunohistochemistry, and functional experiments.
Collapse
Affiliation(s)
- Deborah L. Baines
- Institute for Infection and Immunity, St George’s, University of London, London, United Kingdom
| | - Stanislavs Vasiljevs
- Institute for Infection and Immunity, St George’s, University of London, London, United Kingdom
| | - Kameljit K. Kalsi
- Institute for Infection and Immunity, St George’s, University of London, London, United Kingdom
| |
Collapse
|
32
|
Xu JQ, Fu YL, Zhang J, Zhang KY, Ma J, Tang JY, Zhang ZW, Zhou ZY. Targeting glycolysis in non-small cell lung cancer: Promises and challenges. Front Pharmacol 2022; 13:1037341. [PMID: 36532721 PMCID: PMC9748442 DOI: 10.3389/fphar.2022.1037341] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/04/2022] [Indexed: 08/17/2023] Open
Abstract
Metabolic disturbance, particularly of glucose metabolism, is a hallmark of tumors such as non-small cell lung cancer (NSCLC). Cancer cells tend to reprogram a majority of glucose metabolism reactions into glycolysis, even in oxygen-rich environments. Although glycolysis is not an efficient means of ATP production compared to oxidative phosphorylation, the inhibition of tumor glycolysis directly impedes cell survival and growth. This review focuses on research advances in glycolysis in NSCLC and systematically provides an overview of the key enzymes, biomarkers, non-coding RNAs, and signaling pathways that modulate the glycolysis process and, consequently, tumor growth and metastasis in NSCLC. Current medications, therapeutic approaches, and natural products that affect glycolysis in NSCLC are also summarized. We found that the identification of appropriate targets and biomarkers in glycolysis, specifically for NSCLC treatment, is still a challenge at present. However, LDHB, PDK1, MCT2, GLUT1, and PFKM might be promising targets in the treatment of NSCLC or its specific subtypes, and DPPA4, NQO1, GAPDH/MT-CO1, PGC-1α, OTUB2, ISLR, Barx2, OTUB2, and RFP180 might be prognostic predictors of NSCLC. In addition, natural products may serve as promising therapeutic approaches targeting multiple steps in glycolysis metabolism, since natural products always present multi-target properties. The development of metabolic intervention that targets glycolysis, alone or in combination with current therapy, is a potential therapeutic approach in NSCLC treatment. The aim of this review is to describe research patterns and interests concerning the metabolic treatment of NSCLC.
Collapse
Affiliation(s)
- Jia-Qi Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan-Li Fu
- Department of Oncology, Shenzhen (Fu Tian) Hospital, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Jing Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kai-Yu Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Ma
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing-Yi Tang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhi-Wei Zhang
- Department of Oncology, Shenzhen (Fu Tian) Hospital, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Zhong-Yan Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
33
|
Lau SCM, Pan Y, Velcheti V, Wong KK. Squamous cell lung cancer: Current landscape and future therapeutic options. Cancer Cell 2022; 40:1279-1293. [PMID: 36270277 DOI: 10.1016/j.ccell.2022.09.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/19/2022] [Accepted: 09/27/2022] [Indexed: 01/09/2023]
Abstract
Squamous cell lung cancers (lung squamous cell carcinomas [LUSCs]) are associated with high mortality and a lack of therapies specific to this disease. Although recurrent molecular aberrations are present in LUSCs, efforts to develop targeted therapies against receptor tyrosine kinases, signaling transduction, and cell cycle checkpoints in LUSCs were met with significant challenges. The present therapeutic landscape focuses on epigenetic therapies to modulate the expression of lineage-dependent survival pathways and undruggable oncogenes. Another important therapeutic approach is to exploit metabolic vulnerabilities unique to LUSCs. These novel therapies may synergize with immune checkpoint inhibitors in the right therapeutic context. For example, the recognition that alterations in KEAP1-NFE2L2 in LUSCs affected antitumor immune responses created unique opportunities for targeted, metabolic, and immune combinations. This article provides a perspective on how lessons learned from the past influence the current therapeutic landscape and opportunities for future drug development for LUSCs.
Collapse
Affiliation(s)
- Sally C M Lau
- Department of Medical Oncology, Laura & Issac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, Smilow Building 10th Floor, Suite 1001, New York, NY 10016, USA
| | - Yuanwang Pan
- Department of Medical Oncology, Laura & Issac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, Smilow Building 10th Floor, Suite 1001, New York, NY 10016, USA
| | - Vamsidhar Velcheti
- Department of Medical Oncology, Laura & Issac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, Smilow Building 10th Floor, Suite 1001, New York, NY 10016, USA
| | - Kwok Kin Wong
- Department of Medical Oncology, Laura & Issac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, Smilow Building 10th Floor, Suite 1001, New York, NY 10016, USA.
| |
Collapse
|
34
|
Tang M, Xu H, Huang H, Kuang H, Wang C, Li Q, Zhang X, Ge Y, Song M, Zhang X, Wang Z, Ma C, Kang J, Zhang W, Wang Y, Zhang B, Zhang X, Chen Y, Cong M, Melino G, Wang X, Zhou F, Sun Q, Shi H. Metabolism-Based Molecular Subtyping Endows Effective Ketogenic Therapy in p53-Mutant Colon Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201992. [PMID: 36031388 PMCID: PMC9561794 DOI: 10.1002/advs.202201992] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/25/2022] [Indexed: 06/15/2023]
Abstract
Although targeting cancer metabolism is a promising therapeutic strategy, clinical success depends on accurate molecular and metabolic subtyping. Here, this study reports two metabolism-based molecular subtypes associated with the ketogenic treatment of colon cancer: glycolytic (glycolysis+ /ketolysis- ) and ketolytic (glycolysis+ /ketolysis+ ), which are manifested by distinct profiles of metabolic enzymes and mitochondrial dysfunction, and by different responses to ketone-containing interventions in vitro and in vivo. Notably, the glycolytic subtype is able to be transformed into the ketolytic subtype in p53-mutated tumors upon glucose limitation, rendering resistance to ketogenic therapy associated with upregulation of ketolytic enzymes, such as OXCT1 by mutant p53. The allosteric activator of mutant p53 effectively blocks the rewired molecular expression and the reprogrammed metabolism, leading to the suppression of tumor growth. The findings highlight the utility of metabolic subtyping to guide ketogenic therapy in colon cancer and identify mutant p53 as a synthetic lethality target for ketogenic treatment.
Collapse
Affiliation(s)
- Meng Tang
- Department of Radiation and Medical OncologyHubei Key Laboratory of Tumor Biological BehaviorsHubei Clinical Cancer Study CenterZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Gastrointestinal Surgery/ Department of Clinical NutritionBeijing Shijitan HospitalCapital Medical UniversityBeijing10038China
- Key Laboratory of Cancer FSMP for State Market RegulationBeijing100038China
- Laboratory of Cell Engineering, Institute of BiotechnologyResearch Unit of Cell Death Mechanism, 2021RU008Chinese Academy of Medical Science20 Dongda StreetBeijing100071China
- Comprehensive Oncology DepartmentNational Cancer Center/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100021China
| | - Hui Xu
- Department of Radiation and Medical OncologyHubei Key Laboratory of Tumor Biological BehaviorsHubei Clinical Cancer Study CenterZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Hongyan Huang
- Department of OncologyBeijing Shijitan HospitalCapital Medical UniversityBeijing10038China
| | - Hao Kuang
- Department of Radiation and Medical OncologyHubei Key Laboratory of Tumor Biological BehaviorsHubei Clinical Cancer Study CenterZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Radiation OncologySichuan Cancer HospitalChengdu610041China
| | - Chenxi Wang
- Laboratory of Cell Engineering, Institute of BiotechnologyResearch Unit of Cell Death Mechanism, 2021RU008Chinese Academy of Medical Science20 Dongda StreetBeijing100071China
| | - Qinqin Li
- Department of Gastrointestinal Surgery/ Department of Clinical NutritionBeijing Shijitan HospitalCapital Medical UniversityBeijing10038China
| | - Xin Zhang
- Department of Pediatric Hematology and OncologyXinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghai200092China
| | - Yizhong Ge
- Department of Gastrointestinal Surgery/ Department of Clinical NutritionBeijing Shijitan HospitalCapital Medical UniversityBeijing10038China
- Key Laboratory of Cancer FSMP for State Market RegulationBeijing100038China
| | - Mengmeng Song
- Department of Gastrointestinal Surgery/ Department of Clinical NutritionBeijing Shijitan HospitalCapital Medical UniversityBeijing10038China
- Key Laboratory of Cancer FSMP for State Market RegulationBeijing100038China
| | - Xi Zhang
- Department of Gastrointestinal Surgery/ Department of Clinical NutritionBeijing Shijitan HospitalCapital Medical UniversityBeijing10038China
- Key Laboratory of Cancer FSMP for State Market RegulationBeijing100038China
| | - Ziwen Wang
- Department of Gastrointestinal Surgery/ Department of Clinical NutritionBeijing Shijitan HospitalCapital Medical UniversityBeijing10038China
- Key Laboratory of Cancer FSMP for State Market RegulationBeijing100038China
| | - Chaobing Ma
- Laboratory of Cell Engineering, Institute of BiotechnologyResearch Unit of Cell Death Mechanism, 2021RU008Chinese Academy of Medical Science20 Dongda StreetBeijing100071China
| | - Jinlin Kang
- Department of Radiation and Medical OncologyHubei Key Laboratory of Tumor Biological BehaviorsHubei Clinical Cancer Study CenterZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Wanfang Zhang
- Department of Radiation and Medical OncologyHubei Key Laboratory of Tumor Biological BehaviorsHubei Clinical Cancer Study CenterZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - You Wang
- Department of Radiation and Medical OncologyHubei Key Laboratory of Tumor Biological BehaviorsHubei Clinical Cancer Study CenterZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Bo Zhang
- Department of OncologyBeijing Shijitan HospitalCapital Medical UniversityBeijing10038China
| | - Xiaowei Zhang
- Department of Gastrointestinal Surgery/ Department of Clinical NutritionBeijing Shijitan HospitalCapital Medical UniversityBeijing10038China
- Key Laboratory of Cancer FSMP for State Market RegulationBeijing100038China
| | - Yongbing Chen
- Department of Gastrointestinal Surgery/ Department of Clinical NutritionBeijing Shijitan HospitalCapital Medical UniversityBeijing10038China
- Key Laboratory of Cancer FSMP for State Market RegulationBeijing100038China
| | - Minghua Cong
- Comprehensive Oncology DepartmentNational Cancer Center/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100021China
| | - Gerry Melino
- Department of Experimental MedicineTORUniversity of Rome“Tor Vergata”Rome50‐00133Italy
| | - Xiaobin Wang
- Department of PopulationFamilyand Reproductive HealthJohns Hopkins University Bloomberg School of Public Health; and Department of PediatricsJohns Hopkins University School of MedicineBaltimoreMaryland21287USA
| | - Fuxiang Zhou
- Department of Radiation and Medical OncologyHubei Key Laboratory of Tumor Biological BehaviorsHubei Clinical Cancer Study CenterZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Qiang Sun
- Laboratory of Cell Engineering, Institute of BiotechnologyResearch Unit of Cell Death Mechanism, 2021RU008Chinese Academy of Medical Science20 Dongda StreetBeijing100071China
| | - Hanping Shi
- Department of Gastrointestinal Surgery/ Department of Clinical NutritionBeijing Shijitan HospitalCapital Medical UniversityBeijing10038China
- Key Laboratory of Cancer FSMP for State Market RegulationBeijing100038China
| |
Collapse
|
35
|
Ismail A, Tanasova M. Importance of GLUT Transporters in Disease Diagnosis and Treatment. Int J Mol Sci 2022; 23:8698. [PMID: 35955833 PMCID: PMC9368955 DOI: 10.3390/ijms23158698] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 12/04/2022] Open
Abstract
Facilitative sugar transporters (GLUTs) are the primary method of sugar uptake in all mammalian cells. There are 14 different types of those transmembrane proteins, but they transport only a handful of substrates, mainly glucose and fructose. This overlap and redundancy contradict the natural tendency of cells to conserve energy and resources, and has led researchers to hypothesize that different GLUTs partake in more metabolic roles than just sugar transport into cells. Understanding those roles will lead to better therapeutics for a wide variety of diseases and disorders. In this review we highlight recent discoveries of the role GLUTs play in different diseases and disease treatments.
Collapse
Affiliation(s)
- Abdelrahman Ismail
- Department of Chemistry, Michigan Technological University, 1400 Townsend Drive, Houghton, MI 49931, USA
| | - Marina Tanasova
- Department of Chemistry, Michigan Technological University, 1400 Townsend Drive, Houghton, MI 49931, USA
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA
| |
Collapse
|
36
|
Zhou Y, Wang Y. Prognostic implication of an energy metabolism‐related 11‐gene signature in lung cancer. J Biochem Mol Toxicol 2022; 36:e23171. [PMID: 35851973 DOI: 10.1002/jbt.23171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 05/03/2022] [Accepted: 07/01/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Yang Zhou
- Medical Oncology Department of Gastrointestinal Cancer Cancer Hospital of China Medical University Shenyang China
| | - Yuanhe Wang
- Medical Oncology Department of Gastrointestinal Cancer Cancer Hospital of China Medical University Shenyang China
| |
Collapse
|
37
|
Di Magno L, Coluccia A, Bufano M, Ripa S, La Regina G, Nalli M, Di Pastena F, Canettieri G, Silvestri R, Frati L. Discovery of novel human lactate dehydrogenase inhibitors: Structure-based virtual screening studies and biological assessment. Eur J Med Chem 2022; 240:114605. [PMID: 35868126 DOI: 10.1016/j.ejmech.2022.114605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/05/2022] [Accepted: 07/09/2022] [Indexed: 11/04/2022]
Abstract
Most cancer cells switch their metabolism from mitochondrial oxidative phosphorylation to aerobic glycolysis to generate ATP and precursors for the biosynthesis of key macromolecules. The aerobic conversion of pyruvate to lactate, coupled to oxidation of the nicotinamide cofactor, is a primary hallmark of cancer and is catalyzed by lactate dehydrogenase (LDH), a central effector of this pathological reprogrammed metabolism. Hence, inhibition of LDH is a potential new promising therapeutic approach for cancer. In the search for new LDH inhibitors, we carried out a structure-based virtual screening campaign. Here, we report the identification of a novel specific LDH inhibitor, the pyridazine derivative 18 (RS6212), that exhibits potent anticancer activity within the micromolar range in multiple cancer cell lines and synergizes with complex I inhibition in the suppression of tumor growth. Altogether, our data support the conclusion that compound 18 deserves to be further investigated as a starting point for the development of LDH inhibitors and for novel anticancer strategies based on the targeting of key metabolic steps.
Collapse
Affiliation(s)
- Laura Di Magno
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, I-00161, Rome, Italy.
| | - Antonio Coluccia
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Rome, Italy.
| | - Marianna Bufano
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Rome, Italy
| | - Silvia Ripa
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, I-00161, Rome, Italy
| | - Giuseppe La Regina
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Rome, Italy
| | - Marianna Nalli
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Rome, Italy
| | - Fiorella Di Pastena
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, I-00161, Rome, Italy
| | - Gianluca Canettieri
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, I-00161, Rome, Italy.
| | - Romano Silvestri
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Rome, Italy.
| | - Luigi Frati
- Institute Pasteur Italy - Cenci Bolognetti Foundation, Via Regina Elena 291, I-00161, Rome, Italy; IRCCS Neuromed S.p.A., Via Atinense 18, Pozzilli, Isernia, Italy.
| |
Collapse
|
38
|
Yu T, Liu Y, Xue J, Sun X, Zhu D, Ma L, Guo Y, Jin T, Cao H, Chen Y, Zhu T, Li X, Liang H, Du Z, Shan H. Gankyrin modulated non-small cell lung cancer progression via glycolysis metabolism in a YAP1-dependent manner. Cell Death Dis 2022; 8:312. [PMID: 35810157 PMCID: PMC9271063 DOI: 10.1038/s41420-022-01104-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/10/2022] [Accepted: 06/27/2022] [Indexed: 11/09/2022]
Abstract
Non-small cell lung cancer (NSCLC) is highly malignant and heterogeneous form of lung cancer and involves various oncogene alterations. Glycolysis, an important step in tumor metabolism, is closely related to cancer progression. In this study, we investigated the biological function and mechanism of action of Gankyrin in glycolysis and its association with NSCLC. Analyzed of data from The Cancer Genome Atlas as well as NSCLC specimens and adjacent tissues demonstrated that Gankyrin expression was upregulated in NSCLC tissues compared to adjacent normal tissues. Gankyrin was found to significantly aggravate cancer-related phenotypes, including cell viability, migration, invasion, and epithelial mesenchymal transition (EMT), whereas Gankyrin silencing alleviated the malignant phenotype of NSCLC cells. Our results reveal that Gankyrin exerted its function by regulating YAP1 expression and increasing its nuclear translocation. Importantly, YAP1 actuates glycolysis, which involves glucose uptake, lactic acid production, and ATP generation and thus might contribute to the tumorigenic effect of Gankyrin. Furthermore, the Gankyrin-accelerated glycolysis in NSCLC cells was reversed by YAP1 deficiency. Gankyrin knockdown reduced A549 cell tumorigenesis and EMT and decreased YAP1 expression in a subcutaneous xenograft nude mouse model. In conclusion, both Gankyrin and YAP1 play important roles in tumor metabolism, and Gankyrin-targeted inhibition may be a potential anti-cancer therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Tong Yu
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, P. R. China.,Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China.,Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, Heilongjiang, 150081, P. R. China
| | - Yanyan Liu
- Zhuhai People's Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, P. R. China
| | - Junwen Xue
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Xiang Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Di Zhu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Lu Ma
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Yingying Guo
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Tongzhu Jin
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Huiying Cao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Yingzhun Chen
- Department of Pathology, the Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Tong Zhu
- Department of General Surgery, the Fourth Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Xuelian Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Haihai Liang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China.,Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, Heilongjiang, 150081, P. R. China
| | - Zhimin Du
- Zhuhai People's Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, P. R. China. .,Institute of Clinical Pharmacy, the Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China.
| | - Hongli Shan
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, P. R. China. .,Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China. .,Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, Heilongjiang, 150081, P. R. China.
| |
Collapse
|
39
|
Tumour Cell Budding and Spread Through Air Spaces in Squamous Cell Carcinoma of the Lung – Determination and Validation of optimal prognostic cut-offs. Lung Cancer 2022; 169:1-12. [DOI: 10.1016/j.lungcan.2022.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/22/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022]
|
40
|
TRIM46 activates AKT/HK2 signaling by modifying PHLPP2 ubiquitylation to promote glycolysis and chemoresistance of lung cancer cells. Cell Death Dis 2022; 13:285. [PMID: 35354796 PMCID: PMC8967906 DOI: 10.1038/s41419-022-04727-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 02/23/2022] [Accepted: 03/11/2022] [Indexed: 12/13/2022]
Abstract
The incidence of lung cancer is increasing worldwide. Although great progress in lung cancer treatment has been made, the clinical outcome is still unsatisfactory. Tripartite motif (TRIM)-containing proteins has been shown to be closely related to tumor progression. However, the function of TRIM46 in lung cancer is largely unknown. Here, TRIM46 amplification was found in lung adenocarcinoma (LUAD) tissues and TRIM46 amplification was significantly associated with a poor survival rate. Overexpression of wild type TRIM46 increased the proliferation of LUAD cells and glycolysis, promoted xenografts growth, and enhanced cisplatin (DDP) resistance of LUAD cells via increased ubiquitination of pleckstrin homology domain leucine-rich repeat protein phosphatase 2 (PHLPP2) and upregulation of p-AKT. In contrast, overexpression of RING-mutant TRIM46 did not show any effects, suggesting the function of TRIM46 was dependent on the E3 ligase activity. Furthermore, we found that TRIM46 promoted LUAD cell proliferation and DDP resistance by enhancing glycolysis. PHLPP2 overexpression reversed the effects of TRIM46 overexpression. Amplification of TRIM46 also promoted LUAD growth and enhanced its DDP resistance in a patient-derived xenograft (PDX) model. In conclusion, our data highlight the importance of TRIM46/PHLPP2/AKT signaling in lung cancer and provide new insights into therapeutic strategies for lung cancer.
Collapse
|
41
|
Molecular Mechanisms of Cutaneous Squamous Cell Carcinoma. Int J Mol Sci 2022; 23:ijms23073478. [PMID: 35408839 PMCID: PMC8998533 DOI: 10.3390/ijms23073478] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/18/2022] [Accepted: 03/18/2022] [Indexed: 12/25/2022] Open
Abstract
Non-melanoma skin cancers are cutaneous malignancies representing the most common form of cancer in the United States. They are comprised predominantly of basal cell carcinomas and squamous cell carcinomas (cSCC). The incidence of cSCC is increasing, resulting in substantial morbidity and ever higher treatment costs; currently in excess of one billion dollars, per annum. Here, we review research defining the molecular basis and development of cSCC that aims to provide new insights into pathogenesis and drive the development of novel, cost and morbidity saving therapies.
Collapse
|
42
|
Qian Y, Ding P, Xu J, Nie X, Lu B. CCL2 activates AKT signaling to promote glycolysis and chemoresistance in glioma cells. Cell Biol Int 2022; 46:819-828. [PMID: 35178826 DOI: 10.1002/cbin.11778] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 12/26/2021] [Accepted: 01/22/2022] [Indexed: 11/06/2022]
Abstract
The incidence of gliomas is increasing. Although great progress in glioma treatment has been made, the clinical outcome remains unsatisfactory. Chemokine (C-C motif) ligand 2 (CCL2) plays a key role in different types of cancers, including glioma. However, the function of CCL2 in glioma chemoresistance is not fully understood. In the current study, CCL2 was significantly upregulated in glioma. More importantly, CCL2 and CCR2 were significantly upregulated in temozolomide (TMZ)-resistant glioma. TMZ-resistant malignant glioblastoma cells (U251/TMZ) had higher expressions of CCL2 and CCR2 and a higher level of glycolysis as compared to its parental cell line U251. Silencing of CCL2 in U251/TMZ cells inhibited glycolysis. Overexpression of CCL2 reduced TMZ-induced apoptosis through activation of the AKT pathway and promotion of glycolysis. Moreover, overexpression of CCL2 significantly reduced the antitumor effect of TMZ in vivo. In conclusion, CCL2 overexpression reduced the antitumor effect of TMZ by enhancing glycolysis through activation of AKT signaling. The findings highlighted the importance of CCL2/CCR2/glycolysis and its potential value i developing new treatment for glioma. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yafang Qian
- Huzhou Cent Hospital, Affiliated Cent Hospital HuZhou University
| | - Peng Ding
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University
| | - Jie Xu
- Huzhou Cent Hospital, Affiliated Cent Hospital HuZhou University
| | - Xiaohu Nie
- Huzhou Cent Hospital, Affiliated Cent Hospital HuZhou University
| | - Bin Lu
- Huzhou Cent Hospital, Affiliated Cent Hospital HuZhou University
| |
Collapse
|
43
|
Cífková E, Brumarová R, Ovčačíková M, Dobešová D, Mičová K, Kvasnička A, Vaňková Z, Šiller J, Sákra L, Friedecký D, Holčapek M. Lipidomic and metabolomic analysis reveals changes in biochemical pathways for non-small cell lung cancer tissues. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159082. [PMID: 34793969 DOI: 10.1016/j.bbalip.2021.159082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/23/2021] [Accepted: 11/09/2021] [Indexed: 02/04/2023]
Abstract
Lung cancer represents one of the leading worldwide causes of cancer death, but the pathobiochemistry of this disease is still not fully understood. Here we characterize the lipidomic and metabolomic profiles of the tumor and surrounding normal tissues for 23 patients with non-small cell lung cancer. In total, 500 molecular species were identified and quantified by a combination of the lipidomic shotgun tandem mass spectrometry (MS/MS) analysis and the targeted metabolomic approach using liquid chromatography (LC) - MS/MS. The statistical evaluation includes multivariate and univariate methods with the emphasis on paired statistical approaches. Our research revealed significant changes in several biochemical pathways related to the central carbon metabolism, acylcarnitines, dipeptides as well as the disruption in the lipid metabolism observed mainly for glycerophospholipids, sphingolipids, and cholesteryl esters.
Collapse
Affiliation(s)
- Eva Cífková
- University of Pardubice, Faculty of Chemical Technology, Department of Analytical Chemistry, Studentská 573, 53210 Pardubice, Czech Republic
| | - Radana Brumarová
- Palacký University Olomouc, Faculty of Medicine and Dentistry, Hněvotínská 5, 77900 Olomouc, Czech Republic
| | - Magdaléna Ovčačíková
- University of Pardubice, Faculty of Chemical Technology, Department of Analytical Chemistry, Studentská 573, 53210 Pardubice, Czech Republic
| | - Dana Dobešová
- Palacký University Olomouc, Faculty of Medicine and Dentistry, Hněvotínská 5, 77900 Olomouc, Czech Republic
| | - Kateřina Mičová
- Palacký University Olomouc, Faculty of Medicine and Dentistry, Hněvotínská 5, 77900 Olomouc, Czech Republic
| | - Aleš Kvasnička
- Palacký University Olomouc, Faculty of Medicine and Dentistry, Hněvotínská 5, 77900 Olomouc, Czech Republic
| | - Zuzana Vaňková
- University of Pardubice, Faculty of Chemical Technology, Department of Analytical Chemistry, Studentská 573, 53210 Pardubice, Czech Republic
| | - Jiří Šiller
- Regional Hospital Pardubice, Surgery Clinic, Kyjevská 44, 53203 Pardubice, Czech Republic
| | - Lukáš Sákra
- Regional Hospital Pardubice, Surgery Clinic, Kyjevská 44, 53203 Pardubice, Czech Republic
| | - David Friedecký
- Palacký University Olomouc, Faculty of Medicine and Dentistry, Hněvotínská 5, 77900 Olomouc, Czech Republic.
| | - Michal Holčapek
- University of Pardubice, Faculty of Chemical Technology, Department of Analytical Chemistry, Studentská 573, 53210 Pardubice, Czech Republic.
| |
Collapse
|
44
|
Zhao Q, Yuan X, Zheng L, Xue M. miR-30d-5p: A Non-Coding RNA With Potential Diagnostic, Prognostic and Therapeutic Applications. Front Cell Dev Biol 2022; 10:829435. [PMID: 35155437 PMCID: PMC8829117 DOI: 10.3389/fcell.2022.829435] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/14/2022] [Indexed: 12/26/2022] Open
Abstract
Cancer is a great challenge facing global public health. Scholars have made plentiful efforts in the research of cancer therapy, but the results are still not satisfactory. In relevant literature, the role of miRNA in cancer has been widely concerned. MicroRNAs (miRNAs) are a non-coding, endogenous, single-stranded RNAs that regulate a variety of biological functions. The abnormal level of miR-30d-5p, a type of miRNAs, has been associated with various human tumor types, including lung cancer, colorectal cancer, esophageal cancer, prostate cancer, liver cancer, cervical cancer, breast cancer and other types of human tumors. This reflects the vital function of miR-30d-5p in tumor prognosis. miR-30d-5p can be identified either as an inhibitor hindering the development of, or a promoter accelerating the occurrence of tumors. In addition, the role of miR-30d-5p in cell proliferation, motility, apoptosis, autophagy, tumorigenesis, and chemoresistance are also noteworthy. The multiple roles of miR-30d-5p in human cancer suggest that it has broad feasibility as a biomarker and therapeutic target. This review describes the connection between miR-30d-5p and the clinical indications of tumors, and summarizes the mechanisms by which miR-30d-5p mediates cancer progression.
Collapse
Affiliation(s)
- Qinlu Zhao
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Yuan
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lian Zheng
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Lian Zheng, ; Miaomiao Xue,
| | - Miaomiao Xue
- Department of General Dentistry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Lian Zheng, ; Miaomiao Xue,
| |
Collapse
|
45
|
van Genugten EAJ, Weijers JAM, Heskamp S, Kneilling M, van den Heuvel MM, Piet B, Bussink J, Hendriks LEL, Aarntzen EHJG. Imaging the Rewired Metabolism in Lung Cancer in Relation to Immune Therapy. Front Oncol 2022; 11:786089. [PMID: 35070990 PMCID: PMC8779734 DOI: 10.3389/fonc.2021.786089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic reprogramming is recognized as one of the hallmarks of cancer. Alterations in the micro-environmental metabolic characteristics are recognized as important tools for cancer cells to interact with the resident and infiltrating T-cells within this tumor microenvironment. Cancer-induced metabolic changes in the micro-environment also affect treatment outcomes. In particular, immune therapy efficacy might be blunted because of somatic mutation-driven metabolic determinants of lung cancer such as acidity and oxygenation status. Based on these observations, new onco-immunological treatment strategies increasingly include drugs that interfere with metabolic pathways that consequently affect the composition of the lung cancer tumor microenvironment (TME). Positron emission tomography (PET) imaging has developed a wide array of tracers targeting metabolic pathways, originally intended to improve cancer detection and staging. Paralleling the developments in understanding metabolic reprogramming in cancer cells, as well as its effects on stromal, immune, and endothelial cells, a wave of studies with additional imaging tracers has been published. These tracers are yet underexploited in the perspective of immune therapy. In this review, we provide an overview of currently available PET tracers for clinical studies and discuss their potential roles in the development of effective immune therapeutic strategies, with a focus on lung cancer. We report on ongoing efforts that include PET/CT to understand the outcomes of interactions between cancer cells and T-cells in the lung cancer microenvironment, and we identify areas of research which are yet unchartered. Thereby, we aim to provide a starting point for molecular imaging driven studies to understand and exploit metabolic features of lung cancer to optimize immune therapy.
Collapse
Affiliation(s)
- Evelien A J van Genugten
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Jetty A M Weijers
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, Tuebingen, Germany.,Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | | | - Berber Piet
- Department of Respiratory Diseases, Radboudumc, Nijmegen, Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Netherlands
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre (UMC), Maastricht, Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| |
Collapse
|
46
|
DeNicola GM, Shackelford DB. Metabolic Phenotypes, Dependencies, and Adaptation in Lung Cancer. Cold Spring Harb Perspect Med 2021; 11:a037838. [PMID: 34127512 PMCID: PMC8559540 DOI: 10.1101/cshperspect.a037838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Lung cancer is a heterogeneous disease that is subdivided into histopathological subtypes with distinct behaviors. Each subtype is characterized by distinct features and molecular alterations that influence tumor metabolism. Alterations in tumor metabolism can be exploited by imaging modalities that use metabolite tracers for the detection and characterization of tumors. Microenvironmental factors, including nutrient and oxygen availability and the presence of stromal cells, are a critical influence on tumor metabolism. Recent technological advances facilitate the direct evaluation of metabolic alterations in patient tumors in this complex microenvironment. In addition, molecular alterations directly influence tumor cell metabolism and metabolic dependencies that influence response to therapy. Current therapeutic approaches to target tumor metabolism are currently being developed and translated into the clinic for patient therapy.
Collapse
Affiliation(s)
- Gina M DeNicola
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - David B Shackelford
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at the University of California, Los Angeles, California 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at the University of California, Los Angeles, California 90095, USA
| |
Collapse
|
47
|
Leiter A, Charokopos A, Bailey S, Gallagher EJ, Hirsch FR, LeRoith D, Wisnivesky JP. Assessing the association of diabetes with lung cancer risk. Transl Lung Cancer Res 2021; 10:4200-4208. [PMID: 35004250 PMCID: PMC8674590 DOI: 10.21037/tlcr-21-601] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/27/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND Diabetes is a well-established risk factor for many cancers, but its relationship with lung cancer incidence remains unclear. In this study, we aimed to assess if diabetes is independently associated with lung cancer risk and histology subtype among participants in a screening study. METHODS In a retrospective cohort study using data from the Prostate, Lung, Colorectal, and Ovarian (PLCO) study, we assessed the association of self-reported diabetes with lung cancer incidence using Poisson regression while adjusting for other established risk factors in the PLCOM2012, a validated lung cancer prediction model. The adjusted association of diabetes and lung cancer cell type was evaluated using nominal regression. Stratified analyses were also conducted according to sex, smoking history, and body mass index categories. RESULTS Overall, 140,395 participants were included in our analysis. Diabetes was not significantly associated with lung cancer incidence [incidence rate ratio (IRR): 1.03, 95% confidence interval (CI): 0.91-1.17]. Similarly, stratified analyses also did not show significant associations between diabetes and lung cancer risk (all P values >0.05). We found no significant difference in the distribution of lung cancer histology in participants with vs. without diabetes (P=0.30). CONCLUSIONS Diabetes was not an independent risk factor for lung cancer in a large cohort of PLCO participants. We did not observe differences in histology according to diabetes status. These results suggest that patients with diabetes do not need more aggressive lung cancer screening. Future research including more detailed metabolic parameters may further elucidate the relationship between metabolic disease and lung cancer risk.
Collapse
Affiliation(s)
- Amanda Leiter
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Antonios Charokopos
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Stacyann Bailey
- Division of General Internal Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily J. Gallagher
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA;,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fred R. Hirsch
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Derek LeRoith
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Juan P. Wisnivesky
- Division of General Internal Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
48
|
Dey S, Ashrafi A, Vidal C, Jain N, Kalainayakan SP, Ghosh P, Alemi PS, Salamat N, Konduri PC, Kim JW, Zhang L. Heme Sequestration Effectively Suppresses the Development and Progression of Both Lung Adenocarcinoma and Squamous Cell Carcinoma. Mol Cancer Res 2021; 20:139-149. [PMID: 34635508 DOI: 10.1158/1541-7786.mcr-21-0385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/21/2021] [Accepted: 10/04/2021] [Indexed: 11/16/2022]
Abstract
Lung adenocarcinoma (ADC) and squamous cell carcinoma (SCC) are two most common subtypes of lung cancer. Here, to identify new, targetable molecular properties of both subtypes, we monitored changes in the levels of heme- and oxidative phosphorylation (OXPHOS)-related proteins during lung tumorigenesis. Heme is a central molecule for oxidative metabolism and ATP generation via OXPHOS. Notably, both lung ADC and SCC tumors can be induced in the genetically engineered KLLuc mouse model harboring the G12D Kras mutation and a conditional Lkb1 knockout. We found that the levels of the rate-limiting heme synthesis enzyme ALAS1 and uptake protein SLC48A1, along with OXPHOS complex subunits, progressively increased as lung tumorigenesis advanced. Our data demonstrated that elevated levels of heme- and OXPHOS-related proteins were associated with both ADC and SCC. Importantly, treatment of KLLuc mice with a heme-sequestering protein, HeSP2, that inhibits heme uptake in tumor cells effectively arrested lung tumor progression, and both ADC and SCC tumors were strongly suppressed. Additionally, HeSP2 effectively suppressed the growth of both SCC and ADC tumor xenografts in NOD/SCID mice. Further analyses indicated that HeSP2 effectively diminished OXPHOS in both ADC and SCC, reduced angiogenesis, alleviated tumor hypoxia, and suppressed cell proliferation. These results show that the advancing of lung tumorigenesis requires progressive increase in cellular heme synthesis and uptake, leading to intensified OXPHOS activity and ATP generation and promoting aggressive tumorigenic functions. IMPLICATIONS: Heme sequestration is an effective strategy for the suppression of both ADC and SCC tumor initiation and development.
Collapse
Affiliation(s)
- Sanchareeka Dey
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Adnin Ashrafi
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Chantal Vidal
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Nivesh Jain
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | | | - Poorva Ghosh
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Parinaz Sadat Alemi
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Narges Salamat
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | | | - Jung-Whan Kim
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
- ReCerise Therapeutics Inc, Korea
| | - Li Zhang
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas.
| |
Collapse
|
49
|
King RJ, Qiu F, Yu F, Singh PK. Metabolic and Immunological Subtypes of Esophageal Cancer Reveal Potential Therapeutic Opportunities. Front Cell Dev Biol 2021; 9:667852. [PMID: 34307352 PMCID: PMC8295652 DOI: 10.3389/fcell.2021.667852] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/08/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Esophageal cancer has the sixth highest rate of cancer-associated deaths worldwide, with many patients displaying metastases and chemotherapy resistance. We sought to find subtypes to see if precision medicine could play a role in finding new potential targets and predicting responses to therapy. Since metabolism not only drives cancers but also serves as a readout, metabolism was examined as a key reporter for differences. METHODS Unsupervised and supervised classification methods, including hierarchical clustering, partial least squares discriminant analysis, k-nearest neighbors, and machine learning techniques, were used to discover and display two major subgroups. Genes, pathways, gene ontologies, survival, and immune differences between the groups were further examined, along with biomarkers between the groups and against normal tissue. RESULTS Esophageal cancer had two major unique metabolic profiles observed between the histological subtypes esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). The metabolic differences suggest that ESCC depends on glycolysis, whereas EAC relies more on oxidative metabolism, catabolism of glycolipids, the tricarboxylic acid (TCA) cycle, and the electron transport chain. We also noted a robust prognostic risk associated with COQ3 expression. In addition to the metabolic alterations, we noted significant alterations in key pathways regulating immunity, including alterations in cytokines and predicted immune infiltration. ESCC appears to have increased signature associated with dendritic cells, Th17, and CD8 T cells, the latter of which correlate with survival in ESCC. We bioinformatically observed that ESCC may be more responsive to checkpoint inhibitor therapy than EAC and postulate targets to enhance therapy further. Lastly, we highlight correlations between differentially expressed enzymes and the potential immune status. CONCLUSION Overall, these results highlight the extreme differences observed between the histological subtypes and may lead to novel biomarkers, therapeutic strategies, and differences in therapeutic response for targeting each esophageal cancer subtype.
Collapse
Affiliation(s)
- Ryan J. King
- The Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Fang Qiu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, United States
| | - Fang Yu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, United States
| | - Pankaj K. Singh
- The Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
50
|
Zhang G, Dong R, Kong D, Liu B, Zha Y, Luo M. The Effect of GLUT1 on Survival Rate and the Immune Cell Infiltration of Lung Adenocarcinoma and Squamous Cell Carcinoma: A Meta and Bioinformatics Analysis. Anticancer Agents Med Chem 2021; 22:223-238. [PMID: 34238200 DOI: 10.2174/1871520621666210708115406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC) are two major subtypes of non-small cell lung cancer (NSCLC). Studies have shown that abnormal expression of glucose transport type 1 (GLUT1) in NSCLC patients has been associated with progression, aggressiveness, and poor clinical outcome. However, the clinical effect of GLUT1 expression on LUAD and LUSC is unclear. OBJECTIVE This study aims to learn more about the character of GLUT1 in LUAD and LUSC. METHODS A meta-analysis was performed to evaluate the GLUT1 protein level, and bioinformatics analysis was used to detect the GLUT1 mRNA expression level, survival differences, and the infiltration abundance of immune cells in samples from TCGA. Meanwhile, functional and network analysis was conducted to detect important signaling pathways and key genes with the Gene Expression Omnibus (GEO) dataset. RESULTS Our results showed that GLUT1 was over-expressed both in LUAD and LUSC. LUAD patients with high GLUT1 expression had a poor prognosis. Additionally, GLUT1 was related to B cell and neutrophil infiltration of LUAD. In LUSC, GLUT1 was correlated with tumor purity, B cell, CD8+ T cell, CD4+ T cell, macrophage, neutrophil, and dendritic cell infiltration. The GEO dataset analysis results suggested GLUT1 potentially participated in the p53 signaling pathway and metabolism of xenobiotics by cytochrome P450 and was associated with KDR, TOX3, AGR2, FOXA1, ERBB3, ANGPT1, and COL4A3 gene in LUAD and LUSC. CONCLUSION GLUT1 might be a potential biomarker for aggressive progression and poor prognosis in LUAD, and a therapeutic biomarker in LUSC.
Collapse
Affiliation(s)
- Guihua Zhang
- Guizhou University School of Medicine, Guizhou University, Gui Yang, China
| | - Rong Dong
- Guizhou University School of Medicine, Guizhou University, Gui Yang, China
| | - Demiao Kong
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Gui Yang, China
| | - Bo Liu
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Gui Yang, China
| | - Yan Zha
- Guizhou University School of Medicine, Guizhou University, Gui Yang, China
| | - Meng Luo
- Guizhou University School of Medicine, Guizhou University, Gui Yang, China
| |
Collapse
|