1
|
Khan F, Elsori D, Verma M, Pandey S, Obaidur Rab S, Siddiqui S, Alabdallah NM, Saeed M, Pandey P. Unraveling the intricate relationship between lipid metabolism and oncogenic signaling pathways. Front Cell Dev Biol 2024; 12:1399065. [PMID: 38933330 PMCID: PMC11199418 DOI: 10.3389/fcell.2024.1399065] [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: 03/11/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Lipids, the primary constituents of the cell membrane, play essential roles in nearly all cellular functions, such as cell-cell recognition, signaling transduction, and energy provision. Lipid metabolism is necessary for the maintenance of life since it regulates the balance between the processes of synthesis and breakdown. Increasing evidence suggests that cancer cells exhibit abnormal lipid metabolism, significantly affecting their malignant characteristics, including self-renewal, differentiation, invasion, metastasis, and drug sensitivity and resistance. Prominent oncogenic signaling pathways that modulate metabolic gene expression and elevate metabolic enzyme activity include phosphoinositide 3-kinase (PI3K)/AKT, MAPK, NF-kB, Wnt, Notch, and Hippo pathway. Conversely, when metabolic processes are not regulated, they can lead to malfunctions in cellular signal transduction pathways. This, in turn, enables uncontrolled cancer cell growth by providing the necessary energy, building blocks, and redox potentials. Therefore, targeting lipid metabolism-associated oncogenic signaling pathways could be an effective therapeutic approach to decrease cancer incidence and promote survival. This review sheds light on the interactions between lipid reprogramming and signaling pathways in cancer. Exploring lipid metabolism as a target could provide a promising approach for creating anticancer treatments by identifying metabolic inhibitors. Additionally, we have also provided an overview of the drugs targeting lipid metabolism in cancer in this review.
Collapse
Affiliation(s)
- Fahad Khan
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Deena Elsori
- Faculty of Resilience, Rabdan Academy, Abu Dhabi, United Arab Emirates
| | - Meenakshi Verma
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India
| | - Shivam Pandey
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Samra Siddiqui
- Department of Health Service Management, College of Public Health and Health Informatics, University of Hail, Haʼil, Saudi Arabia
| | - Nadiyah M. Alabdallah
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- Basic and Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Science, University of Hail, Haʼil, Saudi Arabia
| | - Pratibha Pandey
- Chitkara Centre for Research and Development, Chitkara University, Himachal Pradesh, India
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India
| |
Collapse
|
2
|
Yayan J, Franke KJ, Berger M, Windisch W, Rasche K. Adhesion, metastasis, and inhibition of cancer cells: a comprehensive review. Mol Biol Rep 2024; 51:165. [PMID: 38252369 PMCID: PMC10803487 DOI: 10.1007/s11033-023-08920-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/23/2023] [Indexed: 01/23/2024]
Abstract
This comprehensive review delves into cancer's complexity, focusing on adhesion, metastasis, and inhibition. It explores the pivotal role of these factors in disease progression and therapeutic strategies. This review covers cancer cell migration, invasion, and colonization of distant organs, emphasizing the significance of cell adhesion and the intricate metastasis process. Inhibition approaches targeting adhesion molecules, such as integrins and cadherins, are discussed. Overall, this review contributes significantly to advancing cancer research and developing targeted therapies, holding promise for improving patient outcomes worldwide. Exploring different inhibition strategies revealed promising therapeutic targets to alleviate adhesion and metastasis of cancer cells. The effectiveness of integrin-blocking antibodies, small molecule inhibitors targeting Focal adhesion kinase (FAK) and the Transforming Growth Factor β (TGF-β) pathway, and combination therapies underscores their potential to disrupt focal adhesions and control epithelial-mesenchymal transition processes. The identification of as FAK, Src, β-catenin and SMAD4 offers valuable starting points for further research and the development of targeted therapies. The complex interrelationships between adhesion and metastatic signaling networks will be relevant to the development of new treatment approaches.
Collapse
Affiliation(s)
- Josef Yayan
- Department of Internal Medicine, Division of Pulmonary, Allergy, and Sleep Medicine, Witten/Herdecke University, HELIOS Clinic Wuppertal, Heusnerstr. 40, 42283, Wuppertal, Germany.
| | - Karl-Josef Franke
- Department of Internal Medicine, Pulmonary Division, Internal Intensive Care Medicine, Infectiology, and Sleep Medicine, Märkische Clinics Health Holding Ltd, Clinic Lüdenscheid, Witten/Herdecke University, Lüdenscheid, Germany
| | - Melanie Berger
- Department of Pneumology, Cologne Merheim Hospital, Witten/Herdecke University, Cologne, Germany
| | - Wolfram Windisch
- Department of Pneumology, Cologne Merheim Hospital, Witten/Herdecke University, Cologne, Germany
| | - Kurt Rasche
- Department of Internal Medicine, Division of Pulmonary, Allergy, and Sleep Medicine, Witten/Herdecke University, HELIOS Clinic Wuppertal, Heusnerstr. 40, 42283, Wuppertal, Germany
| |
Collapse
|
3
|
Qin L, Wu J. Targeting anticancer immunity in oral cancer: Drugs, products, and nanoparticles. ENVIRONMENTAL RESEARCH 2023; 239:116751. [PMID: 37507044 DOI: 10.1016/j.envres.2023.116751] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 07/30/2023]
Abstract
Oral cavity carcinomas are the most frequent malignancies among head and neck malignancies. Oral tumors include not only oral cancer cells with different potency and stemness but also consist of diverse cells, containing anticancer immune cells, stromal and also immunosuppressive cells that influence the immune system reactions. The infiltrated T and natural killer (NK) cells are the substantial tumor-suppressive immune compartments in the tumor. The infiltration of these cells has substantial impacts on the response of tumors to immunotherapy, chemotherapy, and radiotherapy. Nevertheless, cancer cells, stromal cells, and some other compartments like regulatory T cells (Tregs), macrophages, and myeloid-derived suppressor cells (MDSCs) can repress the immune responses against malignant cells. Boosting anticancer immunity by inducing the immune system or repressing the tumor-promoting cells is one of the intriguing approaches for the eradication of malignant cells such as oral cancers. This review aims to concentrate on the secretions and interactions in the oral tumor immune microenvironment. We review targeting tumor stroma, immune system and immunosuppressive interactions in oral tumors. This review will also focus on therapeutic targets and therapeutic agents such as nanoparticles and products with anti-tumor potency that can boost anticancer immunity in oral tumors. We also explain possible future perspectives including delivery of various cells, natural products and drugs by nanoparticles for boosting anticancer immunity in oral tumors.
Collapse
Affiliation(s)
- Liling Qin
- Gezhouba Central Hospital of the Third Clinical Medical College of Three Gorges University, Yichang, Hubei, 443002, China
| | - Jianan Wu
- Experimental and Practical Teaching Center, Hubei College of Chinese Medicine, Jingzhou, Hubei, 434000, China.
| |
Collapse
|
4
|
Ctortecka C, Hartlmayr D, Seth A, Mendjan S, Tourniaire G, Udeshi ND, Carr SA, Mechtler K. An Automated Nanowell-Array Workflow for Quantitative Multiplexed Single-Cell Proteomics Sample Preparation at High Sensitivity. Mol Cell Proteomics 2023; 22:100665. [PMID: 37839701 PMCID: PMC10684380 DOI: 10.1016/j.mcpro.2023.100665] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/17/2023] Open
Abstract
Multiplexed and label-free mass spectrometry-based approaches with single-cell resolution have attributed surprising heterogeneity to presumed homogenous cell populations. Even though specialized experimental designs and instrumentation have demonstrated remarkable advances, the efficient sample preparation of single cells still lags. Here, we introduce the proteoCHIP, a universal option for single-cell proteomics sample preparation including multiplexed labeling up to 16-plex with high sensitivity and throughput. The automated processing using a commercial system combining single-cell isolation and picoliter dispensing, the cellenONE, reduces final sample volumes to low nanoliters submerged in a hexadecane layer simultaneously eliminating error-prone manual sample handling and overcoming evaporation. The specialized proteoCHIP design allows direct injection of single cells via a standard autosampler resulting in around 1500 protein groups per TMT10-plex with reduced or eliminated need for a carrier proteome. We evaluated the effect of wider precursor isolation windows at single-cell input levels and found that using 2 Da isolation windows increased overall sensitivity without significantly impacting interference. Using the dedicated mass spectrometry acquisition strategies detailed here, we identified on average close to 2000 proteins per TMT10-plex across 170 multiplexed single cells that readily distinguished human cell types. Overall, our workflow combines highly efficient sample preparation, chromatographic and ion mobility-based filtering, rapid wide-window data-dependent acquisition analysis, and intelligent data analysis for optimal multiplexed single-cell proteomics. This versatile and automated proteoCHIP-based sample preparation approach is sufficiently sensitive to drive biological applications of single-cell proteomics and can be readily adopted by proteomics laboratories.
Collapse
Affiliation(s)
- Claudia Ctortecka
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.
| | - David Hartlmayr
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria; Cellenion SASU, Lyon, France
| | | | - Sasha Mendjan
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | | | - Namrata D Udeshi
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.
| | - Karl Mechtler
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria; Cellenion SASU, Lyon, France; Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria; The Gregor Mendel Institute of Molecular Plant Biology of the Austrian Academy of Sciences (GMI), Vienna BioCenter (VBC), Vienna, Austria.
| |
Collapse
|
5
|
Esposito C, Janneh M, Spaziani S, Calcagno V, Bernardi ML, Iammarino M, Verdone C, Tagliamonte M, Buonaguro L, Pisco M, Aversano L, Cusano A. Assessment of Primary Human Liver Cancer Cells by Artificial Intelligence-Assisted Raman Spectroscopy. Cells 2023; 12:2645. [PMID: 37998378 PMCID: PMC10670489 DOI: 10.3390/cells12222645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
We investigated the possibility of using Raman spectroscopy assisted by artificial intelligence methods to identify liver cancer cells and distinguish them from their Non-Tumor counterpart. To this aim, primary liver cells (40 Tumor and 40 Non-Tumor cells) obtained from resected hepatocellular carcinoma (HCC) tumor tissue and the adjacent non-tumor area (negative control) were analyzed by Raman micro-spectroscopy. Preliminarily, the cells were analyzed morphologically and spectrally. Then, three machine learning approaches, including multivariate models and neural networks, were simultaneously investigated and successfully used to analyze the cells' Raman data. The results clearly demonstrate the effectiveness of artificial intelligence (AI)-assisted Raman spectroscopy for Tumor cell classification and prediction with an accuracy of nearly 90% of correct predictions on a single spectrum.
Collapse
Affiliation(s)
- Concetta Esposito
- Optoelectronic Division-Engineering Department, University of Sannio, 82100 Benevento, Italy
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy; (M.L.B.); (L.B.)
| | - Mohammed Janneh
- Optoelectronic Division-Engineering Department, University of Sannio, 82100 Benevento, Italy
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy; (M.L.B.); (L.B.)
| | - Sara Spaziani
- Optoelectronic Division-Engineering Department, University of Sannio, 82100 Benevento, Italy
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy; (M.L.B.); (L.B.)
| | - Vincenzo Calcagno
- Optoelectronic Division-Engineering Department, University of Sannio, 82100 Benevento, Italy
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy; (M.L.B.); (L.B.)
| | - Mario Luca Bernardi
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy; (M.L.B.); (L.B.)
- Informatics Group, Engineering Department, University of Sannio, 82100 Benevento, Italy
| | - Martina Iammarino
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy; (M.L.B.); (L.B.)
- Informatics Group, Engineering Department, University of Sannio, 82100 Benevento, Italy
| | - Chiara Verdone
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy; (M.L.B.); (L.B.)
- Informatics Group, Engineering Department, University of Sannio, 82100 Benevento, Italy
| | - Maria Tagliamonte
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy; (M.L.B.); (L.B.)
- National Cancer Institute-IRCCS “Pascale”, Via Mariano Semmola, 52, 80131 Napoli, Italy
| | - Luigi Buonaguro
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy; (M.L.B.); (L.B.)
- National Cancer Institute-IRCCS “Pascale”, Via Mariano Semmola, 52, 80131 Napoli, Italy
| | - Marco Pisco
- Optoelectronic Division-Engineering Department, University of Sannio, 82100 Benevento, Italy
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy; (M.L.B.); (L.B.)
| | - Lerina Aversano
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy; (M.L.B.); (L.B.)
- Informatics Group, Engineering Department, University of Sannio, 82100 Benevento, Italy
| | - Andrea Cusano
- Optoelectronic Division-Engineering Department, University of Sannio, 82100 Benevento, Italy
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy; (M.L.B.); (L.B.)
| |
Collapse
|
6
|
Hirose Y, Taniguchi K. Intratumoral metabolic heterogeneity of colorectal cancer. Am J Physiol Cell Physiol 2023; 325:C1073-C1084. [PMID: 37661922 DOI: 10.1152/ajpcell.00139.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/31/2023] [Accepted: 08/28/2023] [Indexed: 09/05/2023]
Abstract
Although the metabolic phenotype within tumors is known to differ significantly from that of the surrounding normal tissue, the importance of this heterogeneity is just becoming widely recognized. Colorectal cancer (CRC) is often classified as the Warburg phenotype, a metabolic type in which the glycolytic system is predominant over oxidative phosphorylation (OXPHOS) in mitochondria for energy production. However, this dichotomy (glycolysis vs. OXPHOS) may be too simplistic and not accurately represent the metabolic characteristics of CRC. Therefore, in this review, we decompose metabolic phenomena into factors based on their source/origin and reclassify them into two categories: extrinsic and intrinsic. In the CRC context, extrinsic factors include those based on the environment, such as hypoxia, nutrient deprivation, and the tumor microenvironment, whereas intrinsic factors include those based on subpopulations, such as pathological subtypes and cancer stem cells. These factors form multiple layers inside and outside the tumor, affecting them additively, dominantly, or mutually exclusively. Consequently, the metabolic phenotype is a heterogeneous and fluid phenomenon reflecting the spatial distribution and temporal continuity of these factors. This allowed us to redefine the characteristics of specific metabolism-related factors in CRC and summarize and update our accumulated knowledge of their heterogeneity. Furthermore, we positioned tumor budding in CRC as an intrinsic factor and a novel form of metabolic heterogeneity, and predicted its metabolic dynamics, noting its similarity to circulating tumor cells and epithelial-mesenchymal transition. Finally, the possibilities and limitations of using human tumor tissue as research material to investigate and assess metabolic heterogeneity are discussed.
Collapse
Affiliation(s)
- Yoshinobu Hirose
- Department of Pathology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Kohei Taniguchi
- Division of Translational Research, Center for Medical Research & Development, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| |
Collapse
|
7
|
Jin HR, Wang J, Wang ZJ, Xi MJ, Xia BH, Deng K, Yang JL. Lipid metabolic reprogramming in tumor microenvironment: from mechanisms to therapeutics. J Hematol Oncol 2023; 16:103. [PMID: 37700339 PMCID: PMC10498649 DOI: 10.1186/s13045-023-01498-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
Lipid metabolic reprogramming is an emerging hallmark of cancer. In order to sustain uncontrolled proliferation and survive in unfavorable environments that lack oxygen and nutrients, tumor cells undergo metabolic transformations to exploit various ways of acquiring lipid and increasing lipid oxidation. In addition, stromal cells and immune cells in the tumor microenvironment also undergo lipid metabolic reprogramming, which further affects tumor functional phenotypes and immune responses. Given that lipid metabolism plays a critical role in supporting cancer progression and remodeling the tumor microenvironment, targeting the lipid metabolism pathway could provide a novel approach to cancer treatment. This review seeks to: (1) clarify the overall landscape and mechanisms of lipid metabolic reprogramming in cancer, (2) summarize the lipid metabolic landscapes within stromal cells and immune cells in the tumor microenvironment, and clarify their roles in tumor progression, and (3) summarize potential therapeutic targets for lipid metabolism, and highlight the potential for combining such approaches with other anti-tumor therapies to provide new therapeutic opportunities for cancer patients.
Collapse
Affiliation(s)
- Hao-Ran Jin
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Wang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zi-Jing Wang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Ming-Jia Xi
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Bi-Han Xia
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Deng
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China.
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Jin-Lin Yang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China.
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
8
|
Zhou Y, Jiang X, Wang X, Huang J, Li T, Jin H, He J. Promise of spatially resolved omics for tumor research. J Pharm Anal 2023; 13:851-861. [PMID: 37719191 PMCID: PMC10499658 DOI: 10.1016/j.jpha.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 07/01/2023] [Accepted: 07/06/2023] [Indexed: 09/19/2023] Open
Abstract
Tumors are spatially heterogeneous tissues that comprise numerous cell types with intricate structures. By interacting with the microenvironment, tumor cells undergo dynamic changes in gene expression and metabolism, resulting in spatiotemporal variations in their capacity for proliferation and metastasis. In recent years, the rapid development of histological techniques has enabled efficient and high-throughput biomolecule analysis. By preserving location information while obtaining a large number of gene and molecular data, spatially resolved metabolomics (SRM) and spatially resolved transcriptomics (SRT) approaches can offer new ideas and reliable tools for the in-depth study of tumors. This review provides a comprehensive introduction and summary of the fundamental principles and research methods used for SRM and SRT techniques, as well as a review of their applications in cancer-related fields.
Collapse
Affiliation(s)
- Yanhe Zhou
- 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, China
| | - Xinyi Jiang
- 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, China
| | - Xiangyi Wang
- 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, China
| | - Jianpeng Huang
- 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, China
| | - Tong Li
- 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, China
| | - Hongtao Jin
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- NMPA Key Laboratory for Safety Research and Evaluation of Innovative Drug, Beijing, 10050, China
| | - Jiuming He
- 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, China
- NMPA Key Laboratory for Safety Research and Evaluation of Innovative Drug, Beijing, 10050, China
| |
Collapse
|
9
|
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
|
10
|
Zhou L, Zhang Q, Zhu Q, Zhan Y, Li Y, Huang X. Role and therapeutic targeting of glutamine metabolism in non‑small cell lung cancer (Review). Oncol Lett 2023; 25:159. [PMID: 36936031 PMCID: PMC10017915 DOI: 10.3892/ol.2023.13745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/13/2023] [Indexed: 03/09/2023] Open
Abstract
The Warburg effect indicates that cancer cells survive through glycolysis under aerobic conditions; as such, the topic of cancer metabolism has aroused interest. It is requisite to further explore cancer metabolism, as it helps to simultaneously explain the process of carcinogenesis and guide therapy. The flexible metabolism of cancer cells, which is the result of metabolic reprogramming, can meet the basic needs of cells, even in a nutrition-deficient environment. Glutamine is the most abundant non-essential amino acid in the circulation, and along with glucose, comprise the two basic nutrients of cancer cell metabolism. Glutamine is crucial in non-small cell lung cancer (NSCLC) cells and serves an important role in supporting cell growth, activating signal transduction and maintaining redox homeostasis. In this perspective, the present review aims to provide a new therapeutic strategy of NSCLC through inhibiting the metabolism of glutamine. This review not only summarizes the significance of glutamine metabolism in NSCLC cells, but also enumerates traditional glutamine inhibitors along with new targets. It also puts forward the concept of combination therapy and patient stratification with the aim of comprehensively showing the effect and prospect of targeted glutamine metabolism in NSCLC therapy. This review was completed by searching for keywords including 'glutamine', 'NSCLC' and 'therapy' on PubMed, and screening out articles.
Collapse
Affiliation(s)
- Lei Zhou
- The First Clinical Medical College, Nanchang University, Nanchang, Jiangxi 330036, P.R. China
| | - Qi Zhang
- The National Engineering Research Center for Bioengineering Drugs and The Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330036, P.R. China
| | - Qing Zhu
- The National Engineering Research Center for Bioengineering Drugs and The Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330036, P.R. China
| | - Yuan Zhan
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yong Li
- Department of Anesthesiology, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Correspondence to: Dr Yong Li, Department of Anesthesiology, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Donghu, Nanchang, Jiangxi 330006, P.R. China, E-mail:
| | - Xuan Huang
- The National Engineering Research Center for Bioengineering Drugs and The Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330036, P.R. China
- Dr Xuan Huang, The National Engineering Research Center for Bioengineering Drugs and The Technologies, Institute of Translational Medicine, Nanchang University, 1299 Xuefu Road, Honggutan, Nanchang, Jiangxi 330036, P.R. China, E-mail:
| |
Collapse
|
11
|
Mazzara S, Travaini L, Botta F, Granata C, Motta G, Melle F, Fiori S, Tabanelli V, Vanazzi A, Ramadan S, Radice T, Raimondi S, Lo Presti G, Ferrari ME, Jereczek-Fossa BA, Tarella C, Ceci F, Pileri S, Derenzini E. Gene expression profiling and FDG-PET radiomics uncover radiometabolic signatures associated with outcome in DLBCL. Blood Adv 2023; 7:630-643. [PMID: 36806558 PMCID: PMC9979764 DOI: 10.1182/bloodadvances.2022007825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/06/2022] [Indexed: 02/23/2023] Open
Abstract
Emerging evidence indicates that chemoresistance is closely related to altered metabolism in cancer. Here, we hypothesized that distinct metabolic gene expression profiling (GEP) signatures might be correlated with outcome and with specific fluorodeoxyglucose positron emission tomography (FDG-PET) radiomic profiles in diffuse large B-cell lymphoma (DLBCL). We retrospectively analyzed a discovery cohort of 48 consecutive patients with DLBCL treated at our center with standard first-line chemoimmunotherapy by performing targeted GEP (T-GEP)- and FDG-PET radiomic analyses on the same target lesions at baseline. T-GEP-based metabolic profiling identified a 6-gene signature independently associated with outcomes in univariate and multivariate analyses. This signature included genes regulating mitochondrial oxidative metabolism (SCL25A1, PDK4, PDPR) that were upregulated and was inversely associated with genes involved in hypoxia and glycolysis (MAP2K1, HIF1A, GBE1) that were downregulated. These data were validated in 2 large publicly available cohorts. By integrating FDG-PET radiomics and T-GEP, we identified a radiometabolic signature (RadSig) including 4 radiomic features (histo kurtosis, histo energy, shape sphericity, and neighboring gray level dependence matrix contrast), significantly associated with the metabolic GEP-based signature (r = 0.43, P = .0027) and with progression-free survival (P = .028). These results were confirmed using different target lesions, an alternative segmentation method, and were validated in an independent cohort of 64 patients. RadSig retained independent prognostic value in relation to the International Prognostic Index score and metabolic tumor volume (MTV). Integration of RadSig and MTV further refined prognostic stratification. This study provides the proof of principle for the use of FDG-PET radiomics as a tool for noninvasive assessment of cancer metabolism and prognostic stratification in DLBCL.
Collapse
Affiliation(s)
- Saveria Mazzara
- Haematopathology Division, European Institute of Oncology (IEO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | | | | | | | - Giovanna Motta
- Haematopathology Division, European Institute of Oncology (IEO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Federica Melle
- Haematopathology Division, European Institute of Oncology (IEO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Stefano Fiori
- Haematopathology Division, European Institute of Oncology (IEO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Valentina Tabanelli
- Haematopathology Division, European Institute of Oncology (IEO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Anna Vanazzi
- Oncohematology Division, IEO IRCCS, Milan, Italy
| | - Safaa Ramadan
- Oncohematology Division, IEO IRCCS, Milan, Italy
- NCI-Cairo University, Cairo, Egypt
| | | | - Sara Raimondi
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, IEO IRCCS, Milan, Italy
| | - Giuliana Lo Presti
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, IEO IRCCS, Milan, Italy
| | | | - Barbara Alicja Jereczek-Fossa
- Division of Radiation Oncology, IEO IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | - Francesco Ceci
- Nuclear Medicine Division, IEO IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Stefano Pileri
- Haematopathology Division, European Institute of Oncology (IEO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Enrico Derenzini
- Oncohematology Division, IEO IRCCS, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| |
Collapse
|
12
|
Lewis CTA, Mascall KS, Wilson HM, Murray F, Kerr KM, Gibson G, Buchan K, Small GR, Nixon GF. An endogenous inhibitor of angiogenesis downregulated by hypoxia in human aortic valve stenosis promotes disease pathogenesis. J Mol Cell Cardiol 2023; 174:25-37. [PMID: 36336008 DOI: 10.1016/j.yjmcc.2022.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/21/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Aortic valve stenosis is the most common valve disease in the western world. Central to the pathogenesis of this disease is the growth of new blood vessels (angiogenesis) within the aortic valve allowing infiltration of immune cells and development of intra-valve inflammation. Identifying the cellular mediators involved in this angiogenesis is important as this may reveal new therapeutic targets which could ultimately prevent the progression of aortic valve stenosis. Aortic valves from patients undergoing surgery for aortic valve replacement or dilation of the aortic arch were examined both ex vivo and in vitro. We now demonstrate that the anti-angiogenic protein, soluble fms-like tyrosine kinase 1 (sFlt1), a non-signalling soluble receptor for vascular endothelial growth factor, is constitutively expressed in non-diseased valves. sFlt-1 expression was, however, significantly reduced in aortic valve tissue from patients with aortic valve stenosis while protein markers of hypoxia were simultaneously increased. Exposure of primary-cultured valve interstitial cells to hypoxia resulted in a decrease in the expression of sFlt-1. We further reveal using a bioassay that siRNA knock-down of sFlt1 in valve interstitial cells directly results in a pro-angiogenic environment. Finally, incubation of aortic valves with sphingosine 1-phosphate, a bioactive lipid-mediator, increased sFlt-1 expression and inhibited angiogenesis within valve tissue. In conclusion, this study demonstrates that sFlt1 expression is directly correlated with angiogenesis in aortic valves and the observed decrease in sFlt-1 expression in aortic valve stenosis could increase valve inflammation, promoting disease progression. This could be a viable therapeutic target in treating this disease.
Collapse
Affiliation(s)
- Christopher T A Lewis
- Aberdeen Cardiovascular and Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, UK
| | - Keith S Mascall
- Aberdeen Cardiovascular and Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, UK
| | - Heather M Wilson
- Aberdeen Cardiovascular and Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, UK
| | - Fiona Murray
- Aberdeen Cardiovascular and Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, UK
| | - Keith M Kerr
- Department of Pathology, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen and Aberdeen Royal Infirmary, UK
| | - George Gibson
- Department of Cardiothoracic Surgery, Aberdeen Royal Infirmary, UK
| | - Keith Buchan
- Department of Cardiothoracic Surgery, Aberdeen Royal Infirmary, UK
| | - Gary R Small
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Graeme F Nixon
- Aberdeen Cardiovascular and Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, UK.
| |
Collapse
|
13
|
Warburg effect in colorectal cancer: the emerging roles in tumor microenvironment and therapeutic implications. J Hematol Oncol 2022; 15:160. [PMID: 36319992 PMCID: PMC9628128 DOI: 10.1186/s13045-022-01358-5] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 09/26/2022] [Indexed: 11/07/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer-related death worldwide. Countless CRC patients undergo disease progression. As a hallmark of cancer, Warburg effect promotes cancer metastasis and remodels the tumor microenvironment, including promoting angiogenesis, immune suppression, cancer-associated fibroblasts formation and drug resistance. Targeting Warburg metabolism would be a promising method for the treatment of CRC. In this review, we summarize information about the roles of Warburg effect in tumor microenvironment to elucidate the mechanisms governing Warburg effect in CRC and to identify novel targets for therapy.
Collapse
|
14
|
Jacquet P, Stéphanou A. Searching for the Metabolic Signature of Cancer: A Review from Warburg's Time to Now. Biomolecules 2022; 12:biom12101412. [PMID: 36291621 PMCID: PMC9599674 DOI: 10.3390/biom12101412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/18/2022] [Accepted: 09/29/2022] [Indexed: 11/29/2022] Open
Abstract
This review focuses on the evolving understanding that we have of tumor cell metabolism, particularly glycolytic and oxidative metabolism, and traces back its evolution through time. This understanding has developed since the pioneering work of Otto Warburg, but the understanding of tumor cell metabolism continues to be hampered by misinterpretation of his work. This has contributed to the use of the new concepts of metabolic switch and metabolic reprogramming, that are out of step with reality. The Warburg effect is often considered to be a hallmark of cancer, but is it really? More generally, is there a metabolic signature of cancer? We draw the conclusion that the signature of cancer cannot be reduced to a single factor, but is expressed at the tissue level in terms of the capacity of cells to dynamically explore a vast metabolic landscape in the context of significant environmental heterogeneities.
Collapse
|
15
|
Tiwari S, Sapkota N, Han Z. Effect of fasting on cancer: A narrative review of scientific evidence. Cancer Sci 2022; 113:3291-3302. [PMID: 35848874 PMCID: PMC9530862 DOI: 10.1111/cas.15492] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/28/2022] [Accepted: 07/03/2022] [Indexed: 11/30/2022] Open
Abstract
Emerging evidence suggests that fasting could play a key role in cancer treatment by fostering conditions that limit cancer cells' adaptability, survival, and growth. Fasting could increase the effectiveness of cancer treatments and limit adverse events. Yet, we lack an integrated mechanistic model for how these two complicated systems interact, limiting our ability to understand, prevent, and treat cancer using fasting. Here, we review recent findings at the interface of oncology and fasting metabolism, with an emphasis on human clinical studies of intermittent fasting. We recommend combining prolonged periodic fasting with a standard conventional therapeutic approach to promote cancer-free survival, treatment efficacy and reduce side effects in cancer patients.
Collapse
Affiliation(s)
- Sagun Tiwari
- Department of Neurology and RehabilitationSeventh People's Hospital of Shanghai University of TCMShanghaiChina
- Shanghai University of TCMShanghaiChina
- Life Care HospitalBagmatiNepal
| | - Namrata Sapkota
- University of Chinese Academy of SciencesBeijingChina
- Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
- Net Fresh HospitalBagmatiNepal
| | - Zhenxiang Han
- Department of Neurology and RehabilitationSeventh People's Hospital of Shanghai University of TCMShanghaiChina
| |
Collapse
|
16
|
Senavirathna L, Ma C, Chen R, Pan S. Spectral Library-Based Single-Cell Proteomics Resolves Cellular Heterogeneity. Cells 2022; 11:cells11152450. [PMID: 35954294 PMCID: PMC9368228 DOI: 10.3390/cells11152450] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 02/07/2023] Open
Abstract
Dissecting the proteome of cell types and states at single-cell resolution, while being highly challenging, has significant implications in basic science and biomedicine. Mass spectrometry (MS)-based single-cell proteomics represents an emerging technology for system-wide, unbiased profiling of proteins in single cells. However, significant challenges remain in analyzing an extremely small amount of proteins collected from a single cell, as a proteome-wide amplification of proteins is not currently feasible. Here, we report an integrated spectral library-based single-cell proteomics (SLB-SCP) platform that is ultrasensitive and well suited for a large-scale analysis. To overcome the low MS/MS signal intensity intrinsically associated with a single-cell analysis, this approach takes an alternative approach by extracting a breadth of information that specifically defines the physicochemical characteristics of a peptide from MS1 spectra, including monoisotopic mass, isotopic distribution, and retention time (hydrophobicity), and uses a spectral library for proteomic identification. This conceptually unique MS platform, coupled with the DIRECT sample preparation method, enabled identification of more than 2000 proteins in a single cell to distinguish different proteome landscapes associated with cellular types and heterogeneity. We characterized individual normal and cancerous pancreatic ductal cells (HPDE and PANC-1, respectively) and demonstrated the substantial difference in the proteomes between HPDE and PANC-1 at the single-cell level. A significant upregulation of multiple protein networks in cancer hallmarks was identified in the PANC-1 cells, functionally discriminating the PANC-1 cells from the HPDE cells. This integrated platform can be built on high-resolution MS and widely accepted proteomic software, making it possible for community-wide applications.
Collapse
Affiliation(s)
- Lakmini Senavirathna
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Cheng Ma
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ru Chen
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sheng Pan
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
17
|
Lapin A, Perfahl H, Jain HV, Reuss M. Integrating a dynamic central metabolism model of cancer cells with a hybrid 3D multiscale model for vascular hepatocellular carcinoma growth. Sci Rep 2022; 12:12373. [PMID: 35858953 PMCID: PMC9300625 DOI: 10.1038/s41598-022-15767-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/29/2022] [Indexed: 11/09/2022] Open
Abstract
We develop here a novel modelling approach with the aim of closing the conceptual gap between tumour-level metabolic processes and the metabolic processes occurring in individual cancer cells. In particular, the metabolism in hepatocellular carcinoma derived cell lines (HEPG2 cells) has been well characterized but implementations of multiscale models integrating this known metabolism have not been previously reported. We therefore extend a previously published multiscale model of vascular tumour growth, and integrate it with an experimentally verified network of central metabolism in HEPG2 cells. This resultant combined model links spatially heterogeneous vascular tumour growth with known metabolic networks within tumour cells and accounts for blood flow, angiogenesis, vascular remodelling and nutrient/growth factor transport within a growing tumour, as well as the movement of, and interactions between normal and cancer cells. Model simulations report for the first time, predictions of spatially resolved time courses of core metabolites in HEPG2 cells. These simulations can be performed at a sufficient scale to incorporate clinically relevant features of different tumour systems using reasonable computational resources. Our results predict larger than expected temporal and spatial heterogeneity in the intracellular concentrations of glucose, oxygen, lactate pyruvate, f16bp and Acetyl-CoA. The integrated multiscale model developed here provides an ideal quantitative framework in which to study the relationship between dosage, timing, and scheduling of anti-neoplastic agents and the physiological effects of tumour metabolism at the cellular level. Such models, therefore, have the potential to inform treatment decisions when drug response is dependent on the metabolic state of individual cancer cells.
Collapse
Affiliation(s)
- Alexey Lapin
- Stuttgart Research Center Systems Biology, University Stuttgart, Stuttgart, Germany
- Institute of Chemical Process Engineering, University Stuttgart, Stuttgart, Germany
| | - Holger Perfahl
- Stuttgart Research Center Systems Biology, University Stuttgart, Stuttgart, Germany
| | - Harsh Vardhan Jain
- Department of Mathematics and Statistics, University of Minnesota Duluth, Duluth, MN, USA
| | - Matthias Reuss
- Stuttgart Research Center Systems Biology, University Stuttgart, Stuttgart, Germany.
| |
Collapse
|
18
|
Zhang C, Wang X, He J, Huang Y, Huang Q, Qin C, Qin J, Chen L. Neural excitotoxicity and the toxic mechanism induced by acute hypoxia in Chinese mitten crab (Eriocheir sinensis). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 245:106131. [PMID: 35255275 DOI: 10.1016/j.aquatox.2022.106131] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/05/2022] [Accepted: 02/27/2022] [Indexed: 06/14/2023]
Abstract
Hypoxia can induce neural excitotoxicity in mammals, but this adverse effect has not been investigated in aquatic animals to date, especially in crustaceans. This study explored the induction effect and toxic mechanism of acute hypoxia stress (1.0 ± 0.1 mg dissolved oxygen /L) for 24 h on neural excitotoxicity in juvenile Chinese mitten crab, Eriocheir sinensis. The results showed that hemolymph glucose and serum lactic acid content were significantly increased, and the mRNA expression of crustacean hyperglycemic hormone and hypoxia-inducible factor 1α were significantly up-regulated in the hypoxia group compared with control. RNA-Seq results confirmed that acute hypoxia stress had a more significant impact on carbohydrate metabolism than lipid and protein metabolism. In addition, the TUNEL assay showed that the apoptosis rate of nerve cells was significantly higher in the hypoxia group than in the control, and similar trends were observed in the expression of apoptosis-related genes. RNA-Seq results also showed that acute hypoxia stress-induced neuronal apoptosis by regulating multiple apoptosis-related pathways. Moreover, free glutamate and GABA contents in the nerve tissue of thoracic ganglia were significantly higher in the hypoxia group than in the control group. Furthermore, the mRNA expression of NMDA related receptors was significantly up-regulated in the hypoxia group compared with the control. Similar trends were observed in the expression of calcium-dependent degrading enzymes and endogenous antioxidant-related proteins or enzymes. Meanwhile, the mRNA expression level of high-affinity neuronal glutamate transporter in the hypoxia group was significantly up-regulated compared with the control, whereas the vesicular glutamate transporter was significantly down-regulated. Furthermore, NMDA-R antagonists (MK-801 and Ro25-6981) injection showed that NMDA-R served as the bridge and core position of glutamate-induced neural neurotoxicity. This study provides a new perspective and theoretical guidance for exploring the regulation of hypoxic tolerance in E. sinensis.
Collapse
Affiliation(s)
- Cong Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Xiaodan Wang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Jiaqi He
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Yuxing Huang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Qincheng Huang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Chuanjie Qin
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Sichuan, 641100, PR China
| | - Jianguang Qin
- College of Science and Engineering, Flinders University, Adelaide, SA, 5001, Australia
| | - Liqiao Chen
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China.
| |
Collapse
|
19
|
Germain N, Dhayer M, Dekiouk S, Marchetti P. Current Advances in 3D Bioprinting for Cancer Modeling and Personalized Medicine. Int J Mol Sci 2022; 23:3432. [PMID: 35408789 PMCID: PMC8998835 DOI: 10.3390/ijms23073432] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 02/01/2023] Open
Abstract
Tumor cells evolve in a complex and heterogeneous environment composed of different cell types and an extracellular matrix. Current 2D culture methods are very limited in their ability to mimic the cancer cell environment. In recent years, various 3D models of cancer cells have been developed, notably in the form of spheroids/organoids, using scaffold or cancer-on-chip devices. However, these models have the disadvantage of not being able to precisely control the organization of multiple cell types in complex architecture and are sometimes not very reproducible in their production, and this is especially true for spheroids. Three-dimensional bioprinting can produce complex, multi-cellular, and reproducible constructs in which the matrix composition and rigidity can be adapted locally or globally to the tumor model studied. For these reasons, 3D bioprinting seems to be the technique of choice to mimic the tumor microenvironment in vivo as closely as possible. In this review, we discuss different 3D-bioprinting technologies, including bioinks and crosslinkers that can be used for in vitro cancer models and the techniques used to study cells grown in hydrogels; finally, we provide some applications of bioprinted cancer models.
Collapse
Affiliation(s)
- Nicolas Germain
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
- Banque de Tissus, Centre de Biologie-Pathologie, CHU Lille, F-59000 Lille, France
| | - Melanie Dhayer
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
| | - Salim Dekiouk
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
| | - Philippe Marchetti
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
- Banque de Tissus, Centre de Biologie-Pathologie, CHU Lille, F-59000 Lille, France
| |
Collapse
|
20
|
Zhou L, Zhang Z, Nice E, Huang C, Zhang W, Tang Y. Circadian rhythms and cancers: the intrinsic links and therapeutic potentials. J Hematol Oncol 2022; 15:21. [PMID: 35246220 PMCID: PMC8896306 DOI: 10.1186/s13045-022-01238-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
The circadian rhythm is an evolutionarily conserved time-keeping system that comprises a wide variety of processes including sleep-wake cycles, eating-fasting cycles, and activity-rest cycles, coordinating the behavior and physiology of all organs for whole-body homeostasis. Acute disruption of circadian rhythm may lead to transient discomfort, whereas long-term irregular circadian rhythm will result in the dysfunction of the organism, therefore increasing the risks of numerous diseases especially cancers. Indeed, both epidemiological and experimental evidence has demonstrated the intrinsic link between dysregulated circadian rhythm and cancer. Accordingly, a rapidly increasing understanding of the molecular mechanisms of circadian rhythms is opening new options for cancer therapy, possibly by modulating the circadian clock. In this review, we first describe the general regulators of circadian rhythms and their functions on cancer. In addition, we provide insights into the mechanisms underlying how several types of disruption of the circadian rhythm (including sleep-wake, eating-fasting, and activity-rest) can drive cancer progression, which may expand our understanding of cancer development from the clock perspective. Moreover, we also summarize the potential applications of modulating circadian rhythms for cancer treatment, which may provide an optional therapeutic strategy for cancer patients.
Collapse
Affiliation(s)
- Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Edouard Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Acupuncture and Chronobiology Laboratory of Sichuan Province, Chengdu, 610075, China.
| |
Collapse
|
21
|
Tarabichi M, Demetter P, Craciun L, Maenhaut C, Detours V. Thyroid cancer under the scope of emerging technologies. Mol Cell Endocrinol 2022; 541:111491. [PMID: 34740746 DOI: 10.1016/j.mce.2021.111491] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 10/08/2021] [Accepted: 10/18/2021] [Indexed: 01/03/2023]
Abstract
The vast majority of thyroid cancers originate from follicular cells. We outline outstanding issues at each step along the path of cancer patient care, from prevention to post-treatment follow-up and highlight how emerging technologies will help address them in the coming years. Three directions will dominate the coming technological landscape. Genomics will reveal tumoral evolutionary history and shed light on how these cancers arise from the normal epithelium and the genomics alteration driving their progression. Transcriptomics will gain cellular and spatial resolution providing a full account of intra-tumor heterogeneity and opening a window on the microenvironment supporting thyroid tumor growth. Artificial intelligence will set morphological analysis on an objective quantitative ground laying the foundations of a systematic thyroid tumor classification system. It will also integrate into unified representations the molecular and morphological perspectives on thyroid cancer.
Collapse
Affiliation(s)
- Maxime Tarabichi
- Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium.
| | - Pieter Demetter
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Ligia Craciun
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Carine Maenhaut
- Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium.
| | - Vincent Detours
- Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
22
|
Jacquemin V, Antoine M, Dom G, Detours V, Maenhaut C, Dumont JE. Dynamic Cancer Cell Heterogeneity: Diagnostic and Therapeutic Implications. Cancers (Basel) 2022; 14:280. [PMID: 35053446 PMCID: PMC8773841 DOI: 10.3390/cancers14020280] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Though heterogeneity of cancers is recognized and has been much discussed in recent years, the concept often remains overlooked in different routine examinations. Indeed, in clinical or biological articles, reviews, and textbooks, cancers and cancer cells are generally presented as evolving distinct entities rather than as an independent heterogeneous cooperative cell population with its self-oriented biology. There are, therefore, conceptual gaps which can mislead the interpretations/diagnostic and therapeutic approaches. In this short review, we wish to summarize and discuss various aspects of this dynamic evolving heterogeneity and its biological, pathological, clinical, diagnostic, and therapeutic implications, using thyroid carcinoma as an illustrative example.
Collapse
Affiliation(s)
- Valerie Jacquemin
- Correspondence: (V.J.); (J.E.D.); Tel.: +32-2-555-32-26 (V.J.); +32-2-555-41-34 (J.E.D.)
| | | | | | | | | | - Jacques E. Dumont
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles, 1070 Brussels, Belgium; (M.A.); (G.D.); (V.D.); (C.M.)
| |
Collapse
|
23
|
Jarboe T, Tuli NY, Chakraborty S, Maniyar RR, DeSouza N, Xiu-Min Li, Moscatello A, Geliebter J, Tiwari RK. Inflammatory Components of the Thyroid Cancer Microenvironment: An Avenue for Identification of Novel Biomarkers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1350:1-31. [PMID: 34888842 DOI: 10.1007/978-3-030-83282-7_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The incidence of thyroid cancer in the United States is on the rise with an appreciably high disease recurrence rate of 20-30%. Anaplastic thyroid cancer (ATC), although rare in occurrence, is an aggressive form of cancer with limited treatment options and bleak cure rates. This chapter uses discussions of in vitro models that are representative of papillary, anaplastic, and follicular thyroid cancer to evaluate the crosstalk between specific cells of the tumor microenvironment (TME), which serves as a highly heterogeneous realm of signaling cascades and metabolism that are associated with tumorigenesis. The cellular constituents of the TME carry out varying characteristic immunomodulatory functions that are discussed throughout this chapter. The aforementioned cell types include cancer-associated fibroblasts (CAFs), endothelial cells (ECs), and cancer stem cells (CSCs), as well as specific immune cells, including natural killer (NK) cells, dendritic cells (DCs), mast cells, T regulatory (Treg) cells, CD8+ T cells, and tumor-associated macrophages (TAMs). TAM-mediated inflammation is associated with a poor prognosis of thyroid cancer, and the molecular basis of the cellular crosstalk between macrophages and thyroid cancer cells with respect to inducing a metastatic phenotype is not yet known. The dynamic nature of the physiological transition to pathological metastatic phenotypes when establishing the TME encompasses a wide range of characteristics that are further explored within this chapter, including the roles of somatic mutations and epigenetic alterations that drive the genetic heterogeneity of cancer cells, allowing for selective advantages that aid in their proliferation. Induction of these proliferating cells is typically accomplished through inflammatory induction, whereby chronic inflammation sets up a constant physiological state of inflammatory cell recruitment. The secretions of these inflammatory cells can alter the genetic makeup of proliferating cells, which can in turn, promote tumor growth.This chapter also presents an in-depth analysis of molecular interactions within the TME, including secretory cytokines and exosomes. Since the exosomal cargo of a cell is a reflection and fingerprint of the originating parental cells, the profiling of exosomal miRNA derived from thyroid cancer cells and macrophages in the TME may serve as an important step in biomarker discovery. Identification of a distinct set of tumor suppressive miRNAs downregulated in ATC-secreted exosomes indicates their role in the regulation of tumor suppressive genes that may increase the metastatic propensity of ATC. Additionally, the high expression of pro-inflammatory cytokines in studies looking at thyroid cancer and activated macrophage conditioned media suggests the existence of an inflammatory TME in thyroid cancer. New findings are suggestive of the presence of a metastatic niche in ATC tissues that is influenced by thyroid tumor microenvironment secretome-induced epithelial to mesenchymal transition (EMT), mediated by a reciprocal interaction between the pro-inflammatory M1 macrophages and the thyroid cancer cells. Thus, targeting the metastatic thyroid carcinoma microenvironment could offer potential therapeutic benefits and should be explored further in preclinical and translational models of human metastatic thyroid cancer.
Collapse
Affiliation(s)
- Tara Jarboe
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Neha Y Tuli
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Sanjukta Chakraborty
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA.,Weill Cornell Medicine, New York, NY, USA
| | - Rachana R Maniyar
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA.,Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicole DeSouza
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Xiu-Min Li
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | | | - Jan Geliebter
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Raj K Tiwari
- Departments of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
24
|
Duraj T, Carrión-Navarro J, Seyfried TN, García-Romero N, Ayuso-Sacido A. Metabolic therapy and bioenergetic analysis: The missing piece of the puzzle. Mol Metab 2021; 54:101389. [PMID: 34749013 PMCID: PMC8637646 DOI: 10.1016/j.molmet.2021.101389] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Aberrant metabolism is recognized as a hallmark of cancer, a pillar necessary for cellular proliferation. Regarding bioenergetics (ATP generation), most cancers display a preference not only toward aerobic glycolysis ("Warburg effect") and glutaminolysis (mitochondrial substrate level-phosphorylation) but also toward other metabolites such as lactate, pyruvate, and fat-derived sources. These secondary metabolites can assist in proliferation but cannot fully cover ATP demands. SCOPE OF REVIEW The concept of a static metabolic profile is challenged by instances of heterogeneity and flexibility to meet fuel/anaplerotic demands. Although metabolic therapies are a promising tool to improve therapeutic outcomes, either via pharmacological targets or press-pulse interventions, metabolic plasticity is rarely considered. Lack of bioenergetic analysis in vitro and patient-derived models is hindering translational potential. Here, we review the bioenergetics of cancer and propose a simple analysis of major metabolic pathways, encompassing both affordable and advanced techniques. A comprehensive compendium of Seahorse XF bioenergetic measurements is presented for the first time. MAJOR CONCLUSIONS Standardization of principal readouts might help researchers to collect a complete metabolic picture of cancer using the most appropriate methods depending on the sample of interest.
Collapse
Affiliation(s)
- Tomás Duraj
- Faculty of Medicine, Institute for Applied Molecular Medicine (IMMA), CEU San Pablo University, 28668, Madrid, Spain.
| | - Josefa Carrión-Navarro
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223, Madrid, Spain; Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043, Madrid, Spain.
| | - Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, MA, 02467, USA.
| | - Noemí García-Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223, Madrid, Spain; Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043, Madrid, Spain.
| | - Angel Ayuso-Sacido
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223, Madrid, Spain; Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043, Madrid, Spain; Faculty of Medicine, Universidad Francisco de Vitoria, 28223, Madrid, Spain.
| |
Collapse
|
25
|
Alvarez R, Mandal D, Chittiboina P. Canonical and Non-Canonical Roles of PFKFB3 in Brain Tumors. Cells 2021; 10:cells10112913. [PMID: 34831136 PMCID: PMC8616071 DOI: 10.3390/cells10112913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 12/27/2022] Open
Abstract
PFKFB3 is a bifunctional enzyme that modulates and maintains the intracellular concentrations of fructose-2,6-bisphosphate (F2,6-P2), essentially controlling the rate of glycolysis. PFKFB3 is a known activator of glycolytic rewiring in neoplastic cells, including central nervous system (CNS) neoplastic cells. The pathologic regulation of PFKFB3 is invoked via various microenvironmental stimuli and oncogenic signals. Hypoxia is a primary inducer of PFKFB3 transcription via HIF-1alpha. In addition, translational modifications of PFKFB3 are driven by various intracellular signaling pathways that allow PFKFB3 to respond to varying stimuli. PFKFB3 synthesizes F2,6P2 through the phosphorylation of F6P with a donated PO4 group from ATP and has the highest kinase activity of all PFKFB isoenzymes. The intracellular concentration of F2,6P2 in cancers is maintained primarily by PFKFB3 allowing cancer cells to evade glycolytic suppression. PFKFB3 is a primary enzyme responsible for glycolytic tumor metabolic reprogramming. PFKFB3 protein levels are significantly higher in high-grade glioma than in non-pathologic brain tissue or lower grade gliomas, but without relative upregulation of transcript levels. High PFKFB3 expression is linked to poor survival in brain tumors. Solitary or concomitant PFKFB3 inhibition has additionally shown great potential in restoring chemosensitivity and radiosensitivity in treatment-resistant brain tumors. An improved understanding of canonical and non-canonical functions of PFKFB3 could allow for the development of effective combinatorial targeted therapies for brain tumors.
Collapse
Affiliation(s)
- Reinier Alvarez
- Department of Neurological Surgery, University of Colorado School of Medicine, Aurora, CO 80045, USA;
- Neurosurgery Unit for Pituitary and Inheritable Disorders, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20824, USA;
| | - Debjani Mandal
- Neurosurgery Unit for Pituitary and Inheritable Disorders, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20824, USA;
| | - Prashant Chittiboina
- Neurosurgery Unit for Pituitary and Inheritable Disorders, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20824, USA;
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20824, USA
- Correspondence:
| |
Collapse
|
26
|
Patra S, Elahi N, Armorer A, Arunachalam S, Omala J, Hamid I, Ashton AW, Joyce D, Jiao X, Pestell RG. Mechanisms Governing Metabolic Heterogeneity in Breast Cancer and Other Tumors. Front Oncol 2021; 11:700629. [PMID: 34631530 PMCID: PMC8495201 DOI: 10.3389/fonc.2021.700629] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/30/2021] [Indexed: 12/14/2022] Open
Abstract
Reprogramming of metabolic priorities promotes tumor progression. Our understanding of the Warburg effect, based on studies of cultured cancer cells, has evolved to a more complex understanding of tumor metabolism within an ecosystem that provides and catabolizes diverse nutrients provided by the local tumor microenvironment. Recent studies have illustrated that heterogeneous metabolic changes occur at the level of tumor type, tumor subtype, within the tumor itself, and within the tumor microenvironment. Thus, altered metabolism occurs in cancer cells and in the tumor microenvironment (fibroblasts, immune cells and fat cells). Herein we describe how these growth advantages are obtained through either “convergent” genetic changes, in which common metabolic properties are induced as a final common pathway induced by diverse oncogene factors, or “divergent” genetic changes, in which distinct factors lead to subtype-selective phenotypes and thereby tumor heterogeneity. Metabolic heterogeneity allows subtyping of cancers and further metabolic heterogeneity occurs within the same tumor mass thought of as “microenvironmental metabolic nesting”. Furthermore, recent findings show that mutations of metabolic genes arise in the majority of tumors providing an opportunity for the development of more robust metabolic models of an individual patient’s tumor. The focus of this review is on the mechanisms governing this metabolic heterogeneity in breast cancer.
Collapse
Affiliation(s)
- Sayani Patra
- Pensylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA, United States.,Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Naveed Elahi
- Pensylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA, United States.,Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Aaron Armorer
- Pensylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA, United States.,Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Swathi Arunachalam
- Pensylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA, United States.,Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Joshua Omala
- Pensylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA, United States.,Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Iman Hamid
- Pensylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA, United States.,Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Anthony W Ashton
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba.,Program in Cardiovascular Medicine, Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | - David Joyce
- Medical School, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Xuanmao Jiao
- Pensylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA, United States.,Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Richard G Pestell
- Pensylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA, United States.,Xavier University School of Medicine at Aruba, Oranjestad, Aruba.,Cancer Center, Wistar Institute, Philadelphia, PA, United States
| |
Collapse
|
27
|
Pappa KI, Daskalakis G, Anagnou NP. Metabolic rewiring is associated with HPV-specific profiles in cervical cancer cell lines. Sci Rep 2021; 11:17718. [PMID: 34489482 PMCID: PMC8421399 DOI: 10.1038/s41598-021-96038-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 07/29/2021] [Indexed: 01/10/2023] Open
Abstract
Both HPV-positive and HPV-negative cervical cancers are associated with aberrant metabolism, although the oncogenic drivers remain elusive. Here we show the assessment of the metabolomic profiles of four distinct cervical cell lines, a normal and three cancer cell lines, one HPV-negative (C33A) and two HPV-positive (SiHa HPV16+, HeLa HPV18+), employing an ultra performance liquid chromatography and a high resolution mass spectrometry. Out of the total 462 metabolites, 248 to 326 exhibited statistically significant differences, while Random Forests analysis identified unique molecules for each cell line. The two HPV+ cell lines exhibited features of Warburg metabolism, consistent with the role of the HPV E6 protein. SiHa and HeLa cells displayed purine salvage pathway activity, while C33A cells revealed synthesis of cytidine, via a novel mechanism. These data document a highly dynamic HPV-specific rewiring of metabolic pathways occurring in cervical cancer. Therefore, this approach can eventually provide novel mechanistic insights into cervical carcinogenesis.
Collapse
Affiliation(s)
- Kalliopi I Pappa
- Cell and Gene Therapy Laboratory, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.,First Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - George Daskalakis
- First Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Nicholas P Anagnou
- Cell and Gene Therapy Laboratory, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece. .,Laboratory of Biology, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| |
Collapse
|
28
|
Jaymand M, Davatgaran Taghipour Y, Rezaei A, Derakhshankhah H, Foad Abazari M, Samadian H, Hamblin MR. Radiolabeled carbon-based nanostructures: New radiopharmaceuticals for cancer therapy? Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213974] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
LSD1 defines erythroleukemia metabolism by controlling the lineage-specific transcription factors GATA1 and C/EBPα. Blood Adv 2021; 5:2305-2318. [PMID: 33929501 DOI: 10.1182/bloodadvances.2020003521] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/17/2021] [Indexed: 12/18/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogenous malignancy characterized by distinct lineage subtypes and various genetic/epigenetic alterations. As with other neoplasms, AML cells have well-known aerobic glycolysis, but metabolic variations depending on cellular lineages also exist. Lysine-specific demethylase-1 (LSD1) has been reported to be crucial for human leukemogenesis, which is currently one of the emerging therapeutic targets. However, metabolic roles of LSD1 and lineage-dependent factors remain to be elucidated in AML cells. Here, we show that LSD1 directs a hematopoietic lineage-specific metabolic program in AML subtypes. Erythroid leukemia (EL) cells particularly showed activated glycolysis and high expression of LSD1 in both AML cell lines and clinical samples. Transcriptome, chromatin immunoprecipitation-sequencing, and metabolomic analyses revealed that LSD1 was essential not only for glycolysis but also for heme synthesis, the most characteristic metabolic pathway of erythroid origin. Notably, LSD1 stabilized the erythroid transcription factor GATA1, which directly enhanced the expression of glycolysis and heme synthesis genes. In contrast, LSD1 epigenetically downregulated the granulo-monocytic transcription factor C/EBPα. Thus, the use of LSD1 knockdown or chemical inhibitor dominated C/EBPα instead of GATA1 in EL cells, resulting in metabolic shifts and growth arrest. Furthermore, GATA1 suppressed the gene encoding C/EBPα that then acted as a repressor of GATA1 target genes. Collectively, we conclude that LSD1 shapes metabolic phenotypes in EL cells by balancing these lineage-specific transcription factors and that LSD1 inhibitors pharmacologically cause lineage-dependent metabolic remodeling.
Collapse
|
30
|
Kuai XY, Lei ZY, Liu XS, Shao XY. The Interaction of GLUT1 and FOXM1 Leads to a Poor Prognosis in Colorectal Cancer. Anticancer Agents Med Chem 2021; 20:941-950. [PMID: 32188390 DOI: 10.2174/1871520620666200318094618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND Colorectal Cancer (CRC) is one of the most common fatal diseases with high morbidity. Alteration of glucose metabolism is one of the hallmarks in the development of CRC. Glucose Transporter 1 (GLUT1) is a key rate-limiting protein in hyperactive glucose metabolism and up-regulated in CRC, however, the underlying mechanism of the altered metabolism in CRC is still unknown. METHODS In this study, immunohistochemical staining was used to evaluate the expression of GLUT1 and FOXM1 in 135 paired CRC and adjacent normal tissues. The association between the expression of GLUT1/FOXM1 and clinicopathological factors was determined and the correlation between GLUT1 and FOXM1 in CRC was investigated. RESULTS Our results revealed that regardless of tumor location, GLUT1 and FOXM1 were overexpressed in CRC tissues, especially in patients with positive lymph node metastasis and TNM stage III-IV. Furthermore, GLUT1 showed a significantly strong link with FOXM1 in CRC tissue. CONCLUSION Overexpression of GLUT1 and FOXM1 may play critical roles in CRC leading to a poor prognosis.
Collapse
Affiliation(s)
- Xiao-Yi Kuai
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Zhi-Yi Lei
- Department of Radiology, The Qinghai Provincial People's Hospital, XiNing, QingHai, China
| | - Xiao-Shuang Liu
- Department of Colorectal Surgery, Changhai Hospital, Shanghai, China
| | - Xin-Yu Shao
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
31
|
Cancer Cell Metabolism in Hypoxia: Role of HIF-1 as Key Regulator and Therapeutic Target. Int J Mol Sci 2021; 22:ijms22115703. [PMID: 34071836 PMCID: PMC8199012 DOI: 10.3390/ijms22115703] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 12/13/2022] Open
Abstract
In order to meet the high energy demand, a metabolic reprogramming occurs in cancer cells. Its role is crucial in promoting tumor survival. Among the substrates in demand, oxygen is fundamental for bioenergetics. Nevertheless, tumor microenvironment is frequently characterized by low-oxygen conditions. Hypoxia-inducible factor 1 (HIF-1) is a pivotal modulator of the metabolic reprogramming which takes place in hypoxic cancer cells. In the hub of cellular bioenergetics, mitochondria are key players in regulating cellular energy. Therefore, a close crosstalk between mitochondria and HIF-1 underlies the metabolic and functional changes of cancer cells. Noteworthy, HIF-1 represents a promising target for novel cancer therapeutics. In this review, we summarize the molecular mechanisms underlying the interplay between HIF-1 and energetic metabolism, with a focus on mitochondria, of hypoxic cancer cells.
Collapse
|
32
|
Podsednik A, Jiang J, Jacob A, Li LZ, Xu HN. Optical Redox Imaging of Treatment Responses to Nampt Inhibition and Combination Therapy in Triple-Negative Breast Cancer Cells. Int J Mol Sci 2021; 22:ijms22115563. [PMID: 34070254 PMCID: PMC8197351 DOI: 10.3390/ijms22115563] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 01/02/2023] Open
Abstract
We evaluated the utility of optical redox imaging (ORI) to identify the therapeutic response of triple-negative breast cancers (TNBC) under various drug treatments. Cultured HCC1806 and MDA-MB-231 cells treated with FK866 (nicotinamide phosphoribosyltransferase (Nampt) inhibitor), FX11 (lactate dehydrogenase A inhibitor), paclitaxel, and their combinations were subjected to ORI, followed by imaging fluorescently labeled reactive oxygen species (ROS). Cell growth inhibition was measured by a cell viability assay. We found that both cell lines experienced significant NADH decrease and redox ratio (Fp/(NADH+Fp)) increase due to FK866 treatment; however, HCC1806 was much more responsive than MDA-MB-231. We further studied HCC1806 with the main findings: (i) nicotinamide riboside (NR) partially restored NADH in FK866-treated cells; (ii) FX11 induced an over 3-fold NADH increase in FK866 or FK866+NR pretreated cells; (iii) FK866 combined with paclitaxel caused synergistic increases in both Fp and the redox ratio; (iv) FK866 sensitized cells to paclitaxel treatments, which agrees with the redox changes detected by ORI; (v) Fp and the redox ratio positively correlated with cell growth inhibition; and (vi) Fp and NADH positively correlated with ROS level. Our study supports the utility of ORI for detecting the treatment responses of TNBC to Nampt inhibition and the sensitization effects on standard chemotherapeutics.
Collapse
|
33
|
Park JK, Coffey NJ, Limoges A, Le A. The Heterogeneity of Lipid Metabolism in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1311:39-56. [PMID: 34014533 DOI: 10.1007/978-3-030-65768-0_3] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The study of cancer cell metabolism has traditionally focused on glycolysis and glutaminolysis. However, lipidomic technologies have matured considerably over the last decade and broadened our understanding of how lipid metabolism is relevant to cancer biology [1-3]. Studies now suggest that the reprogramming of cellular lipid metabolism contributes directly to malignant transformation and progression [4, 5]. For example, de novo lipid synthesis can supply proliferating tumor cells with phospholipid components that comprise the plasma and organelle membranes of new daughter cells [6, 7]. Moreover, the upregulation of mitochondrial β-oxidation can support tumor cell energetics and redox homeostasis [8], while lipid-derived messengers can regulate major signaling pathways or coordinate immunosuppressive mechanisms [9-11]. Lipid metabolism has, therefore, become implicated in a variety of oncogenic processes, including metastatic colonization, drug resistance, and cell differentiation [10, 12-16]. However, whether we can safely and effectively modulate the underlying mechanisms of lipid metabolism for cancer therapy is still an open question.
Collapse
Affiliation(s)
- Joshua K Park
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nathan J Coffey
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Aaron Limoges
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Anne Le
- Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, USA.
| |
Collapse
|
34
|
Broadfield LA, Pane AA, Talebi A, Swinnen JV, Fendt SM. Lipid metabolism in cancer: New perspectives and emerging mechanisms. Dev Cell 2021; 56:1363-1393. [PMID: 33945792 DOI: 10.1016/j.devcel.2021.04.013] [Citation(s) in RCA: 245] [Impact Index Per Article: 81.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/15/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022]
Abstract
Tumors undergo metabolic transformations to sustain uncontrolled proliferation, avoid cell death, and seed in secondary organs. An increased focus on cancer lipid metabolism has unveiled a number of mechanisms that promote tumor growth and survival, many of which are independent of classical cellular bioenergetics. These mechanisms include modulation of ferroptotic-mediated cell death, support during tumor metastasis, and interactions with the cells of the tumor microenvironment. As such, targeting lipid metabolism for anti-cancer therapies is attractive, with recent work on small-molecule inhibitors identifying compounds to target lipid metabolism. Here, we discuss these topics and identify open questions.
Collapse
Affiliation(s)
- Lindsay A Broadfield
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Antonino Alejandro Pane
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Ali Talebi
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute (LKI), KU Leuven, University of Leuven, Leuven, Belgium
| | - Johannes V Swinnen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute (LKI), KU Leuven, University of Leuven, Leuven, Belgium
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium.
| |
Collapse
|
35
|
Pfohl U, Pflaume A, Regenbrecht M, Finkler S, Graf Adelmann Q, Reinhard C, Regenbrecht CRA, Wedeken L. Precision Oncology Beyond Genomics: The Future Is Here-It Is Just Not Evenly Distributed. Cells 2021; 10:928. [PMID: 33920536 PMCID: PMC8072767 DOI: 10.3390/cells10040928] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is a multifactorial disease with increasing incidence. There are more than 100 different cancer types, defined by location, cell of origin, and genomic alterations that influence oncogenesis and therapeutic response. This heterogeneity between tumors of different patients and also the heterogeneity within the same patient's tumor pose an enormous challenge to cancer treatment. In this review, we explore tumor heterogeneity on the longitudinal and the latitudinal axis, reviewing current and future approaches to study this heterogeneity and their potential to support oncologists in tailoring a patient's treatment regimen. We highlight how the ideal of precision oncology is reaching far beyond the knowledge of genetic variants to inform clinical practice and discuss the technologies and strategies already available to improve our understanding and management of heterogeneity in cancer treatment. We will focus on integrating multi-omics technologies with suitable in vitro models and their proficiency in mimicking endogenous tumor heterogeneity.
Collapse
Affiliation(s)
- Ulrike Pfohl
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
- Institut für Molekulare Biowissenschaften, Goethe Universität Frankfurt am Main, Theodor-W.-Adorno-Platz 1, 60323 Frankfurt am Main, Germany
| | - Alina Pflaume
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| | - Manuela Regenbrecht
- Helios Klinikum Berlin-Buch, Schwanebecker Chaussee 50, 13125 Berlin, Germany;
| | - Sabine Finkler
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| | - Quirin Graf Adelmann
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| | - Christoph Reinhard
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| | - Christian R. A. Regenbrecht
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
- Institut für Pathologie, Universitätsklinikum Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Lena Wedeken
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| |
Collapse
|
36
|
Atif F, Yousuf S, Espinosa-Garcia C, Stein DG. Progesterone Modulates Mitochondrial Functions in Human Glioblastoma Cells. Mol Neurobiol 2021; 58:3805-3816. [PMID: 33847913 DOI: 10.1007/s12035-021-02382-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/31/2021] [Indexed: 11/25/2022]
Abstract
A substantial literature supports the notion that cancer is a metabolic disease. Mitochondria are sexually dimorphic, and progesterone (P4) plays a key regulatory role in mitochondrial functions. We investigated the effect of P4 on mitochondrial functions in three human glioblastoma multiforme (GBM) cell lines. In dose-response and time-response studies, GBM cells were exposed to different concentrations of P4 followed by mitochondrial stress-testing with a Seahorse analyzer. Data were analyzed for oxygen consumption rate (OCR), extracellular acidification rate (ECAR), and spare respiratory capacity (SRC) to determine the effects of P4 exposure on mitochondrial respiration and rate of glycolysis. We also examined the effect of P4 on mitochondrial superoxide radical generation by confocal microscopy. As early as 1h post-P4 exposure, we found a substantial dose-dependent inhibitory effect of P4 on OCR, ECAR, and SRC in all GBM cell lines. P4 treatment altered the levels of basal respiration, maximum respiration, nonmitochondrial oxygen consumption, ATP production, and proton leak. P4 given at 80-μM concentration showed the maximum inhibitory effect compared to controls. Live imaging data showed an 11-22% increase in superoxide radical generation in all three GBM cell lines following 6h exposure to a high concentration of P4. Our data show that high-dose P4 exerts an inhibitory effect on both mitochondrial respiration and glycolysis in GBM cells. These effects would lead to decreased tumor size and rate of growth, representing a potential treatment to control the spread of GBM.
Collapse
Affiliation(s)
- Fahim Atif
- Brain Research Laboratory, Department of Emergency Medicine, Emory University School of Medicine, Whitehead Biomedical Research Building, Room 655A, Atlanta, GA, 30322, USA.
| | - Seema Yousuf
- Brain Research Laboratory, Department of Emergency Medicine, Emory University School of Medicine, Whitehead Biomedical Research Building, Room 655A, Atlanta, GA, 30322, USA
| | - Claudia Espinosa-Garcia
- Brain Research Laboratory, Department of Emergency Medicine, Emory University School of Medicine, Whitehead Biomedical Research Building, Room 655A, Atlanta, GA, 30322, USA
| | - Donald G Stein
- Brain Research Laboratory, Department of Emergency Medicine, Emory University School of Medicine, Whitehead Biomedical Research Building, Room 655A, Atlanta, GA, 30322, USA
- Neuroscience and Behavioral Biology Program, Emory College of Arts and Sciences, Atlanta, GA, 30322, USA
| |
Collapse
|
37
|
Abstract
Glioblastomas (GBMs) exhibit altered metabolism to support a variety of bioenergetic and biosynthetic demands for tumor growth, invasion, and drug resistance. Changes in glycolytic flux, oxidative phosphorylation, the pentose phosphate pathway, fatty acid biosynthesis and oxidation, and nucleic acid biosynthesis are observed in GBMs to help drive tumorigenesis. Both the genetic landscape of GBMs and the unique brain tumor microenvironment shape metabolism; therefore, an understanding of how both intrinsic and extrinsic factors modulate metabolism is becoming increasingly important for finding effect targets and therapeutics for GBM.
Collapse
Affiliation(s)
- Danielle Morrow
- Department of Molecular and Medical Pharmacology, University of California Los Angeles
| | - Jenna Minami
- Department of Molecular and Medical Pharmacology, University of California Los Angeles
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, University of California Los Angeles; David Geffen School of Medicine, University of California Los Angeles.
| |
Collapse
|
38
|
Becker HM, Deitmer JW. Proton Transport in Cancer Cells: The Role of Carbonic Anhydrases. Int J Mol Sci 2021; 22:ijms22063171. [PMID: 33804674 PMCID: PMC8003680 DOI: 10.3390/ijms22063171] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
Intra- and extracellular pH regulation is a pivotal function of all cells and tissues. Net outward transport of H+ is a prerequisite for normal physiological function, since a number of intracellular processes, such as metabolism and energy supply, produce acid. In tumor tissues, distorted pH regulation results in extracellular acidification and the formation of a hostile environment in which cancer cells can outcompete healthy local host cells. Cancer cells employ a variety of H+/HCO3−-coupled transporters in combination with intra- and extracellular carbonic anhydrase (CA) isoforms, to alter intra- and extracellular pH to values that promote tumor progression. Many of the transporters could closely associate to CAs, to form a protein complex coined “transport metabolon”. While transport metabolons built with HCO3−-coupled transporters require CA catalytic activity, transport metabolons with monocarboxylate transporters (MCTs) operate independently from CA catalytic function. In this article, we assess some of the processes and functions of CAs for tumor pH regulation and discuss the role of intra- and extracellular pH regulation for cancer pathogenesis and therapeutic intervention.
Collapse
Affiliation(s)
- Holger M. Becker
- Zoology and Animal Physiology, Institute of Zoology, TU Dresden, D-01217 Dresden, Germany
- Correspondence:
| | - Joachim W. Deitmer
- Department of Biology, University of Kaiserslautern, D-67653 Kaiserslautern, Germany;
| |
Collapse
|
39
|
Horchani M, Della Sala G, Caso A, D’Aria F, Esposito G, Laurenzana I, Giancola C, Costantino V, Jannet HB, Romdhane A. Molecular Docking and Biophysical Studies for Antiproliferative Assessment of Synthetic Pyrazolo-Pyrimidinones Tethered with Hydrazide-Hydrazones. Int J Mol Sci 2021; 22:2742. [PMID: 33800505 PMCID: PMC7962976 DOI: 10.3390/ijms22052742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 12/22/2022] Open
Abstract
Chemotherapy represents the most applied approach to cancer treatment. Owing to the frequent onset of chemoresistance and tumor relapses, there is an urgent need to discover novel and more effective anticancer drugs. In the search for therapeutic alternatives to treat the cancer disease, a series of hybrid pyrazolo[3,4-d]pyrimidin-4(5H)-ones tethered with hydrazide-hydrazones, 5a-h, was synthesized from condensation reaction of pyrazolopyrimidinone-hydrazide 4 with a series of arylaldehydes in ethanol, in acid catalysis. In vitro assessment of antiproliferative effects against MCF-7 breast cancer cells, unveiled that 5a, 5e, 5g, and 5h were the most effective compounds of the series and exerted their cytotoxic activity through apoptosis induction and G0/G1 phase cell-cycle arrest. To explore their mechanism at a molecular level, 5a, 5e, 5g, and 5h were evaluated for their binding interactions with two well-known anticancer targets, namely the epidermal growth factor receptor (EGFR) and the G-quadruplex DNA structures. Molecular docking simulations highlighted high binding affinity of 5a, 5e, 5g, and 5h towards EGFR. Circular dichroism (CD) experiments suggested 5a as a stabilizer agent of the G-quadruplex from the Kirsten ras (KRAS) oncogene promoter. In the light of these findings, we propose the pyrazolo-pyrimidinone scaffold bearing a hydrazide-hydrazone moiety as a lead skeleton for designing novel anticancer compounds.
Collapse
Affiliation(s)
- Mabrouk Horchani
- Laboratory of Heterocyclic Chemistry, Natural Products and Reactivity, Medicinal Chemistry and Natural Products (LR11ES39), Faculty of Sciences of Monastir, University of Monastir, 5000 Monastir, Tunisia; (M.H.); (A.R.)
| | - Gerardo Della Sala
- Department of Marine Biotechnology, Stazione Zoologica Anton Dohrn, Villa Comunale, 80125 Naples, Italy;
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy;
| | - Alessia Caso
- The Blue Chemistry Lab, Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (A.C.); (G.E.)
| | - Federica D’Aria
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (F.D.); (C.G.)
| | - Germana Esposito
- The Blue Chemistry Lab, Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (A.C.); (G.E.)
| | - Ilaria Laurenzana
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy;
| | - Concetta Giancola
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (F.D.); (C.G.)
| | - Valeria Costantino
- The Blue Chemistry Lab, Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (A.C.); (G.E.)
| | - Hichem Ben Jannet
- Laboratory of Heterocyclic Chemistry, Natural Products and Reactivity, Medicinal Chemistry and Natural Products (LR11ES39), Faculty of Sciences of Monastir, University of Monastir, 5000 Monastir, Tunisia; (M.H.); (A.R.)
| | - Anis Romdhane
- Laboratory of Heterocyclic Chemistry, Natural Products and Reactivity, Medicinal Chemistry and Natural Products (LR11ES39), Faculty of Sciences of Monastir, University of Monastir, 5000 Monastir, Tunisia; (M.H.); (A.R.)
| |
Collapse
|
40
|
Metabolic Reprogramming, Questioning, and Implications for Cancer. BIOLOGY 2021; 10:biology10020129. [PMID: 33562201 PMCID: PMC7916061 DOI: 10.3390/biology10020129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/30/2021] [Accepted: 02/03/2021] [Indexed: 01/08/2023]
Abstract
The expression "metabolic reprogramming" has been encountered more and more in the literature since the mid-1990s. It seems to encompass several notions depending on the author, but the lack of a clear definition allows it to be used as a "catch-all" expression. Our first intention is to point out the inconsistencies in the use of the reprogramming terminology for cancer metabolism. The second is to address the over-focus of the role of mutations in metabolic adaptation. With the increased interest in metabolism and, more specifically, in the Warburg effect in cancer research, it seems appropriate to discuss this terminology and related concepts in detail.
Collapse
|
41
|
Yao Z, Zhang Q, Guo F, Guo S, Yang B, Liu B, Li P, Li J, Guan S, Liu X. Long Noncoding RNA PCED1B-AS1 Promotes the Warburg Effect and Tumorigenesis by Upregulating HIF-1α in Glioblastoma. Cell Transplant 2021; 29:963689720906777. [PMID: 32326742 PMCID: PMC7444212 DOI: 10.1177/0963689720906777] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Accumulating evidence suggests that long noncoding RNA (lncRNA) functions as a critical regulator in cancer biology. Here, we characterized the role of lncRNA PCED1B antisense RNA 1 (PCED1B-AS1) in glioblastoma (GBM). PCED1B-AS1 was notably upregulated in GBM tissues and cell lines and closely associated with larger tumor size and higher grade. Patients with high PCED1B-AS1 had shorter survival time than those with low PCED1B-AS1. Functional experiments showed that depletion of PCED1B-AS1 significantly inhibited, while overexpression of PCED1B-AS1 promoted cell proliferation, glucose uptake, and lactate release. Mechanistically, PCED1B-AS1 was able to directly bind to the 5'-UTR of HIF-1α mRNA and potentiate HIF-1α translation, leading to increased HIF-1α protein level, thereby promoting the Warburg effect and tumorigenesis. Importantly, PCED1B-AS1 lost the carcinogenic properties in the absence of HIF-1α. In addition, we also confirmed the existence of the PCED1B-AS1/HIF-1α regulatory axis in vivo. Taken together, our findings demonstrate that PCED1B-AS1 is a novel oncogenic lncRNA in GBM and functions in a HIF-1α-dependent manner, which provides a promising prognostic biomarker and druggable target for GBM.
Collapse
Affiliation(s)
- Zhiqiang Yao
- Department of Interventional Neuroradiology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Qiansheng Zhang
- Department of Neurosurgery, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, China
| | - Fuyou Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Shewei Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Bo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Binghui Liu
- Department of Interventional Neuroradiology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Panxing Li
- Department of Interventional Neuroradiology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Jinyi Li
- Department of Interventional Neuroradiology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Sheng Guan
- Department of Interventional Neuroradiology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Henan, China
| |
Collapse
|
42
|
Høgset H, Horgan CC, Armstrong JPK, Bergholt MS, Torraca V, Chen Q, Keane TJ, Bugeon L, Dallman MJ, Mostowy S, Stevens MM. In vivo biomolecular imaging of zebrafish embryos using confocal Raman spectroscopy. Nat Commun 2020; 11:6172. [PMID: 33268772 PMCID: PMC7710741 DOI: 10.1038/s41467-020-19827-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 10/28/2020] [Indexed: 12/18/2022] Open
Abstract
Zebrafish embryos provide a unique opportunity to visualize complex biological processes, yet conventional imaging modalities are unable to access intricate biomolecular information without compromising the integrity of the embryos. Here, we report the use of confocal Raman spectroscopic imaging for the visualization and multivariate analysis of biomolecular information extracted from unlabeled zebrafish embryos. We outline broad applications of this method in: (i) visualizing the biomolecular distribution of whole embryos in three dimensions, (ii) resolving anatomical features at subcellular spatial resolution, (iii) biomolecular profiling and discrimination of wild type and ΔRD1 mutant Mycobacterium marinum strains in a zebrafish embryo model of tuberculosis and (iv) in vivo temporal monitoring of the wound response in living zebrafish embryos. Overall, this study demonstrates the application of confocal Raman spectroscopic imaging for the comparative bimolecular analysis of fully intact and living zebrafish embryos.
Collapse
Affiliation(s)
- Håkon Høgset
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Conor C Horgan
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - James P K Armstrong
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Mads S Bergholt
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
- Department of Craniofacial Development & Stem Cell Biology, Kings College London, Tower Wing, Guy's Hospital, London, SE1 9RT, UK
| | - Vincenzo Torraca
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Qu Chen
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Timothy J Keane
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Laurence Bugeon
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Margaret J Dallman
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
43
|
Berndt N, Eckstein J, Heucke N, Wuensch T, Gajowski R, Stockmann M, Meierhofer D, Holzhütter HG. Metabolic heterogeneity of human hepatocellular carcinoma: implications for personalized pharmacological treatment. FEBS J 2020; 288:2332-2346. [PMID: 33030799 DOI: 10.1111/febs.15587] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/01/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022]
Abstract
Metabolic reprogramming is a characteristic feature of cancer cells, but there is no unique metabolic program for all tumors. Genetic and gene expression studies have revealed heterogeneous inter- and intratumor patterns of metabolic enzymes and membrane transporters. The functional implications of this heterogeneity remain often elusive. Here, we applied a systems biology approach to gain a comprehensive and quantitative picture of metabolic changes in individual hepatocellular carcinoma (HCC). We used protein intensity profiles determined by mass spectrometry in samples of 10 human HCCs and the adjacent noncancerous tissue to calibrate Hepatokin1, a complex mathematical model of liver metabolism. We computed the 24-h profile of 18 metabolic functions related to carbohydrate, lipid, and nitrogen metabolism. There was a general tendency among the tumors toward downregulated glucose uptake and glucose release albeit with large intertumor variability. This finding calls into question that the Warburg effect dictates the metabolic phenotype of HCC. All tumors comprised elevated β-oxidation rates. Urea synthesis was found to be consistently downregulated but without compromising the tumor's capacity for ammonia detoxification owing to increased glutamine synthesis. The largest intertumor heterogeneity was found for the uptake and release of lactate and the size of the cellular glycogen content. In line with the observed metabolic heterogeneity, the individual HCCs differed largely in their vulnerability against pharmacological treatment with metformin. Taken together, our approach provided a comprehensive and quantitative characterization of HCC metabolism that may pave the way for a computational a priori assessment of pharmacological therapies targeting metabolic processes of HCC.
Collapse
Affiliation(s)
- Nikolaus Berndt
- Institute for Imaging Science and Computational Modelling in Cardiovascular Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Johannes Eckstein
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Niklas Heucke
- Department of Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Tilo Wuensch
- Department of Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Robert Gajowski
- Mass Spectroscopy Facility, Max Planck Institute for Molecular Genetics, Berlin, Germany.,Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Germany
| | - Martin Stockmann
- Department of Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - David Meierhofer
- Mass Spectroscopy Facility, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Hermann-Georg Holzhütter
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| |
Collapse
|
44
|
Mitochondria at Center of Exchanges between Cancer Cells and Cancer-Associated Fibroblasts during Tumor Progression. Cancers (Basel) 2020; 12:cancers12103017. [PMID: 33080792 PMCID: PMC7603005 DOI: 10.3390/cancers12103017] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/14/2020] [Accepted: 10/14/2020] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Malignant cells and their supportive associated fibroblasts (CAFs) exchange various molecules that promote energy production, biosynthesis and therapy resistance by modulating mitochondrial activity and dynamics. We herein review molecular exchanges from CAFs to malignant cells that support tumor growth and therapy resistance, and we highlight the crucial role of CAFs mitochondria in this support. This implies (1) reciprocal mitochondrial control by malignant cells and (2) fibroblast activation. Finally, we discuss therapeutic strategies that could improve current therapies by targeting mitochondrial-mediated dialogue between the two cell types. Abstract Resistance of solid cancer cells to chemotherapies and targeted therapies is not only due to the mutational status of cancer cells but also to the concurring of stromal cells of the tumor ecosystem, such as immune cells, vasculature and cancer-associated fibroblasts (CAFs). The reciprocal education of cancer cells and CAFs favors tumor growth, survival and invasion. Mitochondrial function control, including the regulation of mitochondrial metabolism, oxidative stress and apoptotic stress are crucial for these different tumor progression steps. In this review, we focus on how CAFs participate in cancer progression by modulating cancer cells metabolic functions and mitochondrial apoptosis. We emphasize that mitochondria from CAFs influence their activation status and pro-tumoral effects. We thus advocate that understanding mitochondria-mediated tumor–stroma interactions provides the possibility to consider cancer therapies that improve current treatments by targeting these interactions or mitochondria directly in tumor and/or stromal cells.
Collapse
|
45
|
Kar M, Sultania M, Roy S, Padhi S, Banerjee B. 𝛽-Catenin-a Possible Prognostic Molecular Marker for Recurrence in Histopathologically Negative Surgical Margin of Oral Cancer. Indian J Surg Oncol 2020; 12:128-133. [PMID: 33994738 DOI: 10.1007/s13193-020-01217-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/14/2020] [Indexed: 10/23/2022] Open
Abstract
The locoregional recurrence in oral cancer is not predicted by the histopathological parameters solely as the normal morphological looking cells harbor the genomic instability which acts as the potential tumor cells for recurrence in future. Therefore, there is an urgent need of the biomarker for prognostic stratification of patients with high risk of disease recurrence and appropriate management. Eighty oral squamous cell carcinoma (OSCC) patients were included in the study during the period 2012 to 2014 at Apollo Hospitals and Kalinga Institute of Medical sciences, Bhubaneswar. OSCC tissue samples were collected at the time of surgical excision, and immunohistochemistry (IHC) was performed to check the expression of β-catenin in cut margin (CM) and tumor. Statistical analysis was carried out using SPSS based on clinical and pathological records. It was observed that among 80 patients, 33.75% (27 patients) developed recurrence. The recurrence rate was low for 6 out of 27 patients (22.2%) where β-catenin is positive in tumor and negative in cut margin, while it was quite high in 21 out of 27 (77.8%) when marker is negative in tumor but positive in cut margin (CM). The odds of recurrence among patients having high levels of 𝛽-catenin in CM was 3.6 times higher than the odds of recurrence among patients having lower levels of 𝛽-catenin in CM (p < 0.017). In conclusion, this study highlighted that 𝛽-catenin can be included as a prognostic molecular marker, along with routine histopathological study to influence therapeutic decisions and appropriate management of disease.
Collapse
Affiliation(s)
- Madhabananda Kar
- Department of Surgical Oncology, All India Institute of Medical Sciences, Bhubaneswar, Odisha 751019 India
| | - Mahesh Sultania
- Department of Surgical Oncology, All India Institute of Medical Sciences, Bhubaneswar, Odisha 751019 India
| | - Souvick Roy
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, Odisha 751024 India
| | - Swatishree Padhi
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, Odisha 751024 India
| | - Birendranath Banerjee
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, Odisha 751024 India
| |
Collapse
|
46
|
Varghese E, Samuel SM, Líšková A, Samec M, Kubatka P, Büsselberg D. Targeting Glucose Metabolism to Overcome Resistance to Anticancer Chemotherapy in Breast Cancer. Cancers (Basel) 2020; 12:E2252. [PMID: 32806533 PMCID: PMC7464784 DOI: 10.3390/cancers12082252] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 01/10/2023] Open
Abstract
Breast cancer (BC) is the most prevalent cancer in women. BC is heterogeneous, with distinct phenotypical and morphological characteristics. These are based on their gene expression profiles, which divide BC into different subtypes, among which the triple-negative breast cancer (TNBC) subtype is the most aggressive one. The growing interest in tumor metabolism emphasizes the role of altered glucose metabolism in driving cancer progression, response to cancer treatment, and its distinct role in therapy resistance. Alterations in glucose metabolism are characterized by increased uptake of glucose, hyperactivated glycolysis, decreased oxidative phosphorylation (OXPHOS) component, and the accumulation of lactate. These deviations are attributed to the upregulation of key glycolytic enzymes and transporters of the glucose metabolic pathway. Key glycolytic enzymes such as hexokinase, lactate dehydrogenase, and enolase are upregulated, thereby conferring resistance towards drugs such as cisplatin, paclitaxel, tamoxifen, and doxorubicin. Besides, drug efflux and detoxification are two energy-dependent mechanisms contributing to resistance. The emergence of resistance to chemotherapy can occur at an early or later stage of the treatment, thus limiting the success and outcome of the therapy. Therefore, understanding the aberrant glucose metabolism in tumors and its link in conferring therapy resistance is essential. Using combinatory treatment with metabolic inhibitors, for example, 2-deoxy-D-glucose (2-DG) and metformin, showed promising results in countering therapy resistance. Newer drug designs such as drugs conjugated to sugars or peptides that utilize the enhanced expression of tumor cell glucose transporters offer selective and efficient drug delivery to cancer cells with less toxicity to healthy cells. Last but not least, naturally occurring compounds of plants defined as phytochemicals manifest a promising approach for the eradication of cancer cells via suppression of essential enzymes or other compartments associated with glycolysis. Their benefits for human health open new opportunities in therapeutic intervention, either alone or in combination with chemotherapeutic drugs. Importantly, phytochemicals as efficacious instruments of anticancer therapy can suppress events leading to chemoresistance of cancer cells. Here, we review the current knowledge of altered glucose metabolism in contributing to resistance to classical anticancer drugs in BC treatment and various ways to target the aberrant metabolism that will serve as a promising strategy for chemosensitizing tumors and overcoming resistance in BC.
Collapse
Affiliation(s)
- Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (E.V.); (S.M.S.)
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (E.V.); (S.M.S.)
| | - Alena Líšková
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (A.L.); (M.S.)
| | - Marek Samec
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (A.L.); (M.S.)
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (E.V.); (S.M.S.)
| |
Collapse
|
47
|
Logotheti S, Marquardt S, Gupta SK, Richter C, Edelhäuser BA, Engelmann D, Brenmoehl J, Söhnchen C, Murr N, Alpers M, Singh KP, Wolkenhauer O, Heckl D, Spitschak A, Pützer BM. LncRNA-SLC16A1-AS1 induces metabolic reprogramming during Bladder Cancer progression as target and co-activator of E2F1. Am J Cancer Res 2020; 10:9620-9643. [PMID: 32863950 PMCID: PMC7449907 DOI: 10.7150/thno.44176] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/23/2020] [Indexed: 12/15/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as integral components of E2F1-regulated gene regulatory networks (GRNs), but their implication in advanced or treatment-refractory malignancy is unknown. Methods: We combined high-throughput transcriptomic approaches with bioinformatics and structure modeling to search for lncRNAs that participate in E2F1-activated prometastatic GRNs and their phenotypic targets in the highly-relevant case of E2F1-driven aggressive bladder cancer (BC). RNA immunoprecipitation was performed to verify RNA-protein interactions. Functional analyses including qRT-PCR, immunoblotting, luciferase assays and measurement of extracellular fluxes were conducted to validate expression and target gene regulation. Results: We identified E2F1-responsive lncRNA-SLC16A1-AS1 and its associated neighboring protein-coding gene, SLC16A1/MCT1, which both promote cancer invasiveness. Mechanistically, upon E2F1-mediated co-transactivation of the gene pair, SLC16A1-AS1 associates with E2F1 in a structure-dependent manner and forms an RNA-protein complex that enhances SLC16A1/MCT1 expression through binding to a composite SLC16A1-AS1:E2F1-responsive promoter element. Moreover, SLC16A1-AS1 increases aerobic glycolysis and mitochondrial respiration and fuels ATP production by fatty acid β-oxidation. These metabolic changes are accompanied by alterations in the expression of the SLC16A1-AS1:E2F1-responsive gene PPARA, a key mediator of fatty acid β-oxidation. Conclusions: Our results unveil a new gene regulatory program by which E2F1-induced lncRNA-SLC16A1-AS1 forms a complex with its transcription factor that promotes cancer metabolic reprogramming towards the acquisition of a hybrid oxidative phosphorylation/glycolysis cell phenotype favoring BC invasiveness.
Collapse
|
48
|
Reiter RJ, Sharma R, Ma Q, Rorsales-Corral S, de Almeida Chuffa LG. Melatonin inhibits Warburg-dependent cancer by redirecting glucose oxidation to the mitochondria: a mechanistic hypothesis. Cell Mol Life Sci 2020; 77:2527-2542. [PMID: 31970423 PMCID: PMC11104865 DOI: 10.1007/s00018-019-03438-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/16/2019] [Accepted: 12/23/2019] [Indexed: 12/16/2022]
Abstract
Melatonin has the ability to intervene in the initiation, progression and metastasis of some experimental cancers. A large variety of potential mechanisms have been advanced to describe the metabolic and molecular events associated with melatonin's interactions with cancer cells. There is one metabolic perturbation that is common to a large number of solid tumors and accounts for the ability of cancer cells to actively proliferate, avoid apoptosis, and readily metastasize, i.e., they use cytosolic aerobic glycolysis (the Warburg effect) to rapidly generate the necessary ATP required for the high metabolic demands of the cancer cells. There are several drugs, referred to as glycolytic agents, that cause cancer cells to abandon aerobic glycolysis and shift to the more conventional mitochondrial oxidative phosphorylation for ATP synthesis as in normal cells. In doing so, glycolytic agents also inhibit cancer growth. Herein, we hypothesize that melatonin also functions as an inhibitor of cytosolic glycolysis in cancer cells using mechanisms, i.e., downregulation of the enzyme (pyruvate dehydrogenase kinase) that interferes with the conversion of pyruvate to acetyl CoA in the mitochondria, as do other glycolytic drugs. In doing so, melatonin halts the proliferative activity of cancer cells, reduces their metastatic potential and causes them to more readily undergo apoptosis. This hypothesis is discussed in relation to the previously published reports. Whereas melatonin is synthesized in the mitochondria of normal cells, we hypothesize that this synthetic capability is not present in cancer cell mitochondria because of the depressed acetyl CoA; acetyl CoA is necessary for the rate limiting enzyme in melatonin synthesis, arylalkylamine-N-acetyltransferase. Finally, the ability of melatonin to switch glucose oxidation from the cytosol to the mitochondria also explains how tumors that become resistant to conventional chemotherapies are re-sensitized to the same treatment when melatonin is applied.
Collapse
Affiliation(s)
- Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA.
| | - Ramaswamy Sharma
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Qiang Ma
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Sergio Rorsales-Corral
- Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Mexico
| | | |
Collapse
|
49
|
Chulpanova DS, Kitaeva KV, Rutland CS, Rizvanov AA, Solovyeva VV. Mouse Tumor Models for Advanced Cancer Immunotherapy. Int J Mol Sci 2020; 21:E4118. [PMID: 32526987 PMCID: PMC7312663 DOI: 10.3390/ijms21114118] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 12/21/2022] Open
Abstract
Recent advances in the development of new methods of cancer immunotherapy require the production of complex cancer animal models that reliably reflect the complexity of the tumor and its microenvironment. Mice are good animals to create tumor models because they are low cost, have a short reproductive cycle, exhibit high tumor growth rates, and can be easily genetically modified. However, the obvious problem of these models is the high failure rate observed in human clinical trials after promising results obtained in mouse models. In order to increase the reliability of the results obtained in mice, the tumor model should reflect the heterogeneity of the tumor, contain components of the tumor microenvironment, in particular immune cells, to which the action of immunotherapeutic drugs are directed. This review discusses the current immunocompetent and immunocompromised mouse models of human tumors that are used to evaluate the effectiveness of immunotherapeutic agents, in particular chimeric antigen receptor (CAR) T-cells and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Daria S. Chulpanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (K.V.K.); (A.A.R.)
| | - Kristina V. Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (K.V.K.); (A.A.R.)
| | - Catrin S. Rutland
- Faculty of Medicine and Health Sciences, University of Medicine, Nottingham NG7 2HA, UK;
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (K.V.K.); (A.A.R.)
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (K.V.K.); (A.A.R.)
| |
Collapse
|
50
|
Abstract
The rediscovery and reinterpretation of the Warburg effect in the year 2000 occulted for almost a decade the key functions exerted by mitochondria in cancer cells. Until recent times, the scientific community indeed focused on constitutive glycolysis as a hallmark of cancer cells, which it is not, largely ignoring the contribution of mitochondria to the malignancy of oxidative and glycolytic cancer cells, being Warburgian or merely adapted to hypoxia. In this review, we highlight that mitochondria are not only powerhouses in some cancer cells, but also dynamic regulators of life, death, proliferation, motion and stemness in other types of cancer cells. Similar to the cells that host them, mitochondria are capable to adapt to tumoral conditions, and probably to evolve to ‘oncogenic mitochondria' capable of transferring malignant capacities to recipient cells. In the wider quest of metabolic modulators of cancer, treatments have already been identified targeting mitochondria in cancer cells, but the field is still in infancy.
Collapse
Affiliation(s)
- Debora Grasso
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Luca X Zampieri
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Tânia Capelôa
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Justine A Van de Velde
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|