1
|
Eltayeb K, Alfieri R, Fumarola C, Bonelli M, Galetti M, Cavazzoni A, Digiacomo G, Galvani F, Vacondio F, Lodola A, Mor M, Minari R, Tiseo M, La Monica S, Giorgio Petronini P. Targeting metabolic adaptive responses induced by glucose starvation inhibits cell proliferation and enhances cell death in osimertinib-resistant non-small cell lung cancer (NSCLC) cell lines. Biochem Pharmacol 2024; 228:116161. [PMID: 38522556 DOI: 10.1016/j.bcp.2024.116161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/12/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Osimertinib, a tyrosine kinase inhibitor targeting mutant EGFR, has received approval for initial treatment in patients with Non-Small Cell Lung Cancer (NSCLC). While effective in both first- and second-line treatments, patients eventually develop acquired resistance. Metabolic reprogramming represents a strategy through which cancer cells may resist and adapt to the selective pressure exerted by the drug. In the current study, we investigated the metabolic adaptations associated with osimertinib-resistance in NSCLC cells under low glucose culture conditions. We demonstrated that, unlike osimertinib-sensitive cells, osimertinib-resistant cells were able to survive under low glucose conditions by increasing the rate of glucose and glutamine uptake and by shifting towards mitochondrial metabolism. Inhibiting glucose/pyruvate contribution to mitochondrial respiration, glutamine deamination to glutamate, and oxidative phosphorylation decreased the proliferation and survival abilities of osimertinib-resistant cells to glucose starvation. Our findings underscore the remarkable adaptability of osimertinib-resistant NSCLC cells in a low glucose environment and highlight the pivotal role of mitochondrial metabolism in mediating this adaptation. Targeting the metabolic adaptive responses triggered by glucose shortage emerges as a promising strategy, effectively inhibiting cell proliferation and promoting cell death in osimertinib-resistant cells.
Collapse
Affiliation(s)
- Kamal Eltayeb
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Roberta Alfieri
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy.
| | - Claudia Fumarola
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Mara Bonelli
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Maricla Galetti
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, INAIL-Italian Workers' Compensation Authority, Monte Porzio Catone, 00078 Rome, Italy
| | - Andrea Cavazzoni
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Graziana Digiacomo
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Francesca Galvani
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Federica Vacondio
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Alessio Lodola
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Marco Mor
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Roberta Minari
- Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Silvia La Monica
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | | |
Collapse
|
2
|
Feng L, Chen Y, Mei X, Wang L, Zhao W, Yao J. Prognostic Signature in Osteosarcoma Based on Amino Acid Metabolism-Associated Genes. Cancer Biother Radiopharm 2024; 39:517-531. [PMID: 38512709 DOI: 10.1089/cbr.2024.0002] [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] [Indexed: 03/23/2024] Open
Abstract
Background: Osteosarcoma (OS) is undeniably a formidable bone malignancy characterized by a scarcity of effective treatment options. Reprogramming of amino acid (AA) metabolism has been associated with OS development. The present study was designed to identify metabolism-associated genes (MAGs) that are differentially expressed in OS and to construct a MAG-based prognostic risk signature for this disease. Methods: Expression profiles and clinicopathological data were downloaded from Gene Expression Omnibus (GEO) and UCSC Xena databases. A set of AA MAGs was obtained from the MSigDB database. Differentially expressed genes (DEGs) in GEO dataset were identified using "limma." Prognostic MAGs from UCSC Xena database were determined through univariate Cox regression and used in the prognostic signature development. This signature was validated using another dataset from GEO database. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, single sample gene set enrichment analysis, and GDSC2 analyses were performed to explore the biological functions of the MAGs. A MAG-based nomogram was established to predict 1-, 3-, and 5-year survival. Real-time quantitative polymerase chain reaction, Western blot, and immunohistochemical staining confirmed the expression of MAGs in primary OS and paired adjacent normal tissues. Results: A total of 790 DEGs and 62 prognostic MAGs were identified. A MAG-based signature was constructed based on four MAGs: PIPOX, PSMC2, SMOX, and PSAT1. The prognostic value of this signature was successfully validated, with areas under the receiver operating characteristic curves for 1-, 3-, and 5-year survival of 0.714, 0.719, and 0.715, respectively. This MAG-based signature was correlated with the infiltration of CD56dim natural killer cells and resistance to several antiangiogenic agents. The nomogram was accurate in predictions, with a C-index of 0.77. The expression of MAGs verified by experiment was consistent with the trends observed in GEO database. Conclusion: Four AA MAGs were prognostic of survival in OS patients. This MAG-based signature has the potential to offer valuable insights into the development of treatments for OS.
Collapse
Affiliation(s)
- Liwen Feng
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuting Chen
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangping Mei
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Wang
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenjing Zhao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jiannan Yao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Li K, Gilberti AL, Marden JA, Akula HK, Pollard AC, Guo S, Hu B, Tonge PJ, Qu W. Synthesis and Biological Evaluation of Fluorine-18 and Deuterium Labeled l-Fluoroalanines as Positron Emission Tomography Imaging Agents for Cancer Detection. J Med Chem 2024; 67:10293-10305. [PMID: 38838188 PMCID: PMC11258582 DOI: 10.1021/acs.jmedchem.4c00774] [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] [Indexed: 06/07/2024]
Abstract
To fully explore the potential of 18F-labeled l-fluoroalanine for imaging cancer and other chronic diseases, a simple and mild radiosynthesis method has been established to produce optically pure l-3-[18F]fluoroalanine (l-[18F]FAla), using a serine-derivatized, five-membered-ring sulfamidate as the radiofluorination precursor. A deuterated analogue, l-3-[18F]fluoroalanine-d3 (l-[18F]FAla-d3), was also prepared to improve metabolic stability. Both l-[18F]FAla and l-[18F]FAla-d3 were rapidly taken up by 9L/lacZ, MIA PaCa-2, and U87MG cells and were shown to be substrates for the alanine-serine-cysteine (ASC) amino acid transporter. The ability of l-[18F]FAla, l-[18F]FAla-d3, and the d-enantiomer, d-[18F]FAla-d3, to image tumors was evaluated in U87MG tumor-bearing mice. Despite the significant bone uptake was observed for both l-[18F]FAla and l-[18F]FAla-d3, the latter had enhanced tumor uptake compared to l-[18F]FAla, and d-[18F]FAla-d3 was not specifically taken up by the tumors. The enhanced tumor uptake of l-[18F]FAla-d3 compared with its nondeuterated counterpart, l-[18F]FAla, warranted the further biological investigation of this radiotracer as a potential cancer imaging agent.
Collapse
Affiliation(s)
- Kaixuan Li
- Center for Advanced Study of Drug Action, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Chemistry, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Alexa L. Gilberti
- Center for Advanced Study of Drug Action, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Chemistry, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Jocelyn A. Marden
- Department of Psychiatry and Behavioral Health, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
| | - Hari K. Akula
- Department of Psychiatry and Behavioral Health, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
- PET Research Core, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
| | - Alyssa C. Pollard
- Center for Advanced Study of Drug Action, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Chemistry, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Shuwen Guo
- Center for Advanced Study of Drug Action, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Chemistry, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Bao Hu
- Department of Psychiatry and Behavioral Health, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
- PET Research Core, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
| | - Peter J. Tonge
- Center for Advanced Study of Drug Action, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Chemistry, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Radiology, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
- Stony Brook Cancer Center, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
| | - Wenchao Qu
- Department of Chemistry, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Psychiatry and Behavioral Health, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
- PET Research Core, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
| |
Collapse
|
4
|
Lv S, Zhang Z, Li Z, Ke Q, Ma X, Li N, Zhao X, Zou Q, Sun L, Song T. TFE3-SLC36A1 axis promotes resistance to glucose starvation in kidney cancer cells. J Biol Chem 2024; 300:107270. [PMID: 38599381 PMCID: PMC11098960 DOI: 10.1016/j.jbc.2024.107270] [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: 12/19/2023] [Revised: 03/14/2024] [Accepted: 03/25/2024] [Indexed: 04/12/2024] Open
Abstract
Higher demand for nutrients including glucose is characteristic of cancer. "Starving cancer" has been pursued to curb tumor progression. An intriguing regime is to inhibit glucose transporter GLUT1 in cancer cells. In addition, during cancer progression, cancer cells may suffer from insufficient glucose supply. Yet, cancer cells can somehow tolerate glucose starvation. Uncovering the underlying mechanisms shall shed insight into cancer progression and benefit cancer therapy. TFE3 is a transcription factor known to activate autophagic genes. Physiological TFE3 activity is regulated by phosphorylation-triggered translocation responsive to nutrient status. We recently reported TFE3 constitutively localizes to the cell nucleus and promotes cell proliferation in kidney cancer even under nutrient replete condition. It remains unclear whether and how TFE3 responds to glucose starvation. In this study, we show TFE3 promotes kidney cancer cell resistance to glucose starvation by exposing cells to physiologically relevant glucose concentration. We find glucose starvation triggers TFE3 protein stabilization through increasing its O-GlcNAcylation. Furthermore, through an unbiased functional genomic study, we identify SLC36A1, a lysosomal amino acid transporter, as a TFE3 target gene sensitive to TFE3 protein level. We find SLC36A1 is overexpressed in kidney cancer, which promotes mTOR activity and kidney cancer cell proliferation. Importantly, SLC36A1 level is induced by glucose starvation through TFE3, which enhances cellular resistance to glucose starvation. Suppressing TFE3 or SLC36A1 significantly increases cellular sensitivity to GLUT1 inhibitor in kidney cancer cells. Collectively, we uncover a functional TFE3-SLC36A1 axis that responds to glucose starvation and enhances starvation tolerance in kidney cancer.
Collapse
Affiliation(s)
- Suli Lv
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zongbiao Zhang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenyong Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Ke
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianyun Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Neng Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuefeng Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingli Zou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lidong Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Cell Architecture Research Institute, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Tanjing Song
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Cell Architecture Research Institute, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
5
|
Kryvenko V, Vadász I. Alveolar-capillary endocytosis and trafficking in acute lung injury and acute respiratory distress syndrome. Front Immunol 2024; 15:1360370. [PMID: 38533500 PMCID: PMC10963603 DOI: 10.3389/fimmu.2024.1360370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/29/2024] [Indexed: 03/28/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is associated with high morbidity and mortality but lacks specific therapeutic options. Diverse endocytic processes play a key role in all phases of acute lung injury (ALI), including the initial insult, development of respiratory failure due to alveolar flooding, as a consequence of altered alveolar-capillary barrier function, as well as in the resolution or deleterious remodeling after injury. In particular, clathrin-, caveolae-, endophilin- and glycosylphosphatidyl inositol-anchored protein-mediated endocytosis, as well as, macropinocytosis and phagocytosis have been implicated in the setting of acute lung damage. This manuscript reviews our current understanding of these endocytic pathways and subsequent intracellular trafficking in various phases of ALI, and also aims to identify potential therapeutic targets for patients with ARDS.
Collapse
Affiliation(s)
- Vitalii Kryvenko
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- The Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - István Vadász
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- The Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| |
Collapse
|
6
|
Peter RM, Chou PJ, Shannar A, Patel K, Pan Y, Dave PD, Xu J, Sarwar MS, Kong ANT. An Update on Potential Molecular Biomarkers of Dietary Phytochemicals Targeting Lung Cancer Interception and Prevention. Pharm Res 2023; 40:2699-2714. [PMID: 37726406 DOI: 10.1007/s11095-023-03595-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023]
Abstract
Since ancient times, dietary phytochemicals are known for their medicinal properties. They are broadly classified into polyphenols, terpenoids, alkaloids, phytosterols, and organosulfur compounds. Currently, there is considerable interest in their potential health effects against various diseases, including lung cancer. Lung cancer is the leading cause of cancer deaths with an average of five-year survival rate of lung cancer patients limited to just 14%. Identifying potential early molecular biomarkers of pre-malignant lung cancer cells may provide a strong basis to develop early cancer detection and interception methods. In this review, we will discuss molecular changes, including genetic alterations, inflammation, signal transduction pathways, redox imbalance, epigenetic and proteomic signatures associated with initiation and progression of lung carcinoma. We will also highlight molecular targets of phytochemicals during lung cancer development. These targets mainly consist of cellular signaling pathways, epigenetic regulators and metabolic reprogramming. With growing interest in natural products research, translation of these compounds into new cancer prevention approaches to medical care will be urgently needed. In this context, we will also discuss the overall pharmacokinetic challenges of phytochemicals in translating to humans. Lastly, we will discuss clinical trials of phytochemicals in lung cancer patients.
Collapse
Affiliation(s)
- Rebecca Mary Peter
- Graduate Program in Pharmaceutical Science, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Pochung Jordan Chou
- Graduate Program in Pharmaceutical Science, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Ahmad Shannar
- Graduate Program in Pharmaceutical Science, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Komal Patel
- Graduate Program in Pharmaceutical Science, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Yuxin Pan
- Graduate Program in Pharmaceutical Science, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Parv Dushyant Dave
- Graduate Program in Pharmaceutical Science, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Jiawei Xu
- Graduate Program in Pharmaceutical Science, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Md Shahid Sarwar
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Ah-Ng Tony Kong
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
| |
Collapse
|
7
|
Boso D, Tognon M, Curtarello M, Minuzzo S, Piga I, Brillo V, Lazzarini E, Carlet J, Marra L, Trento C, Rasola A, Masgras I, Caporali L, Del Ben F, Brisotto G, Turetta M, Pastorelli R, Brunelli L, Navaglia F, Esposito G, Grassi A, Indraccolo S. Anti-VEGF therapy selects for clones resistant to glucose starvation in ovarian cancer xenografts. J Exp Clin Cancer Res 2023; 42:196. [PMID: 37550722 PMCID: PMC10405561 DOI: 10.1186/s13046-023-02779-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/25/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Genetic and metabolic heterogeneity are well-known features of cancer and tumors can be viewed as an evolving mix of subclonal populations, subjected to selection driven by microenvironmental pressures or drug treatment. In previous studies, anti-VEGF therapy was found to elicit rewiring of tumor metabolism, causing marked alterations in glucose, lactate ad ATP levels in tumors. The aim of this study was to evaluate whether differences in the sensitivity to glucose starvation existed at the clonal level in ovarian cancer cells and to investigate the effects induced by anti-VEGF therapy on this phenotype by multi-omics analysis. METHODS Clonal populations, obtained from both ovarian cancer cell lines (IGROV-1 and SKOV3) and tumor xenografts upon glucose deprivation, were defined as glucose deprivation resistant (GDR) or glucose deprivation sensitive (GDS) clones based on their in vitro behaviour. GDR and GDS clones were characterized using a multi-omics approach, including genetic, transcriptomic and metabolic analysis, and tested for their tumorigenic potential and reaction to anti-angiogenic therapy. RESULTS Two clonal populations, GDR and GDS, with strikingly different viability following in vitro glucose starvation, were identified in ovarian cancer cell lines. GDR clones survived and overcame glucose starvation-induced stress by enhancing mitochondrial oxidative phosphorylation (OXPHOS) and both pyruvate and lipids uptake, whereas GDS clones were less able to adapt and died. Treatment of ovarian cancer xenografts with the anti-VEGF drug bevacizumab positively selected for GDR clones that disclosed increased tumorigenic properties in NOD/SCID mice. Remarkably, GDR clones were more sensitive than GDS clones to the mitochondrial respiratory chain complex I inhibitor metformin, thus suggesting a potential therapeutic strategy to target the OXPHOS-metabolic dependency of this subpopulation. CONCLUSION A glucose-deprivation resistant population of ovarian cancer cells showing druggable OXPHOS-dependent metabolic traits is enriched in experimental tumors treated by anti-VEGF therapy.
Collapse
Affiliation(s)
- Daniele Boso
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, via Gattamelata 64, 35128, Padova, Italy
| | - Martina Tognon
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Matteo Curtarello
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Sonia Minuzzo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova, 35124, Italy
| | - Ilaria Piga
- Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova, 35124, Italy
| | | | - Elisabetta Lazzarini
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, via Gattamelata 64, 35128, Padova, Italy
| | - Jessica Carlet
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Ludovica Marra
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Chiara Trento
- Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova, 35124, Italy
| | - Andrea Rasola
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ionica Masgras
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Institute of Neuroscience, National Research Council, Padova, Italy
| | - Leonardo Caporali
- Department of Biomedical and Neuromotor Sciences - DIBINEM, University of Bologna, Bologna, Italy
| | - Fabio Del Ben
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO)-IRCCS, Aviano, Italy
| | - Giulia Brisotto
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO)-IRCCS, Aviano, Italy
| | - Matteo Turetta
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO)-IRCCS, Aviano, Italy
| | - Roberta Pastorelli
- Laboratory of Mass Spectrometry, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Laura Brunelli
- Laboratory of Mass Spectrometry, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Filippo Navaglia
- Laboratory Medicine, Department of Medicine-DIMED, University Hospital of Padova, Padova, Italy
| | - Giovanni Esposito
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Angela Grassi
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Stefano Indraccolo
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, via Gattamelata 64, 35128, Padova, Italy.
- Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova, 35124, Italy.
| |
Collapse
|
8
|
Wang C, Chen Q, Tang M, Wei T, Zou J. Effects of TLR2/4 signalling pathway in western mosquitofish (Gambusia affinis) after Edwardsiella tarda infection. JOURNAL OF FISH DISEASES 2023; 46:299-307. [PMID: 36811195 DOI: 10.1111/jfd.13744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 06/18/2023]
Abstract
Gambusia affinis is regarded as an important animal model. Edwardsiella tarda is one of the most serious pathogens affecting aquaculture. The study explores the effects of TLR2/4 partial signalling pathway in G. affinis of E. tarda infection. The study collected the brain, liver, and intestine after E. tarda LD50 and 0.85% NaCl solution challenge at different times (0 h, 3 h, 9 h, 18 h, 24 h, and 48 h). In these three tissues, the mRNA levels of PI3K, AKT3, IRAK4, TAK1, IKKβ, and IL-1β were substantially enhanced (p < .05) then returned to normal levels. Additionally, Rac1 and MyD88 in liver had different trend with other genes in brain and intestine, which displayed significantly indifference. The overexpression of IKKβ, and IL-1β indicated that E. tarda also caused immune reaction in intestine and liver, which would be consistent with delayed edwardsiellosis, which causes intestinal lesions and liver and kidney necrosis. Additionally, MyD88 plays a smaller role than IRAK4 and TAK1 in this signalling pathways. This study could enrich the understanding of the immune mechanism of the TLR2/4 signalling pathway in fish and might help to prescribe preventive measures against E. tarda to prevent infectious diseases in fish.
Collapse
Affiliation(s)
- Chong Wang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Qingshi Chen
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Manfei Tang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Tianli Wei
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Jixing Zou
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| |
Collapse
|
9
|
Salloum G, Bresnick AR, Backer JM. Macropinocytosis: mechanisms and regulation. Biochem J 2023; 480:335-362. [PMID: 36920093 DOI: 10.1042/bcj20210584] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/16/2023]
Abstract
Macropinocytosis is defined as an actin-dependent but coat- and dynamin-independent endocytic uptake process, which generates large intracellular vesicles (macropinosomes) containing a non-selective sampling of extracellular fluid. Macropinocytosis provides an important mechanism of immune surveillance by dendritic cells and macrophages, but also serves as an essential nutrient uptake pathway for unicellular organisms and tumor cells. This review examines the cell biological mechanisms that drive macropinocytosis, as well as the complex signaling pathways - GTPases, lipid and protein kinases and phosphatases, and actin regulatory proteins - that regulate macropinosome formation, internalization, and disposition.
Collapse
Affiliation(s)
- Gilbert Salloum
- Department of Molecular Pharamacology, Albert Einstein College of Medicine, Bronx, NY, U.S.A
| | - Anne R Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, U.S.A
| | - Jonathan M Backer
- Department of Molecular Pharamacology, Albert Einstein College of Medicine, Bronx, NY, U.S.A
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, U.S.A
| |
Collapse
|
10
|
Inhibition of Macropinocytosis Enhances the Sensitivity of Osteosarcoma Cells to Benzethonium Chloride. Cancers (Basel) 2023; 15:cancers15030961. [PMID: 36765917 PMCID: PMC9913482 DOI: 10.3390/cancers15030961] [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: 01/08/2023] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Osteosarcoma (OS) is a primary malignant tumor of bone. Chemotherapy is one of the crucial approaches to prevent its metastasis and improve prognosis. Despite continuous improvements in the clinical treatment of OS, tumor resistance and metastasis remain dominant clinical challenges. Macropinocytosis, a form of non-selective nutrient endocytosis, has received increasing attention as a novel target for cancer therapy, yet its role in OS cells remains obscure. Benzethonium chloride (BZN) is an FDA-approved antiseptic and bactericide with broad-spectrum anticancer effects. Here, we described that BZN suppressed the proliferation, migration, and invasion of OS cells in vitro and in vivo, but simultaneously promoted the massive accumulation of cytoplasmic vacuoles as well. Mechanistically, BZN repressed the ERK1/2 signaling pathway, and the ERK1/2 activator partially neutralized the inhibitory effect of BZN on OS cells. Subsequently, we demonstrated that vacuoles originated from macropinocytosis and indicated that OS cells might employ macropinocytosis as a compensatory survival mechanism in response to BZN. Remarkably, macropinocytosis inhibitors enhanced the anti-OS effect of BZN in vitro and in vivo. In conclusion, our results suggest that BZN may inhibit OS cells by repressing the ERK1/2 signaling pathway and propose a potential strategy to enhance the BZN-induced inhibitory effect by suppressing macropinocytosis.
Collapse
|
11
|
Gu W, Pan Y, Zhao W, Liu J, Meng Y. Metabolic signatures of lymphangioleiomyomatosis in biofluids: nuclear magnetic resonance (NMR)-based metabonomics of blood plasma: a case-control study. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:76. [PMID: 36819539 PMCID: PMC9929845 DOI: 10.21037/atm-22-6420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/10/2023] [Indexed: 01/18/2023]
Abstract
Background Our aim was to analyze and compare the characteristics and differences of blood metabolites between lymphangioleiomyomatosis (LAM) patients and healthy controls, in order to find biomarkers that can be used for the diagnosis and classification of LAM. Methods Between January 2020 to January 2022, 61 eligible LAM patients [51 sporadic LAM (S-LAM) and 10 tuberous sclerosis complex LAM (TSC-LAM)] from the First Affiliated Hospital of Guangzhou Medical University and 30 healthy controls were enrolled. Blood samples were taken for nuclear magnetic resonance (NMR) detection. Data analysis was performed by the umbrella program, and Wilcoxon analysis was used for comparisons between groups. The difference indicators were modeled by logistic regression. Diagnostic accuracy of the best predictive parameters was evaluated by the area under the receiver operating characteristic (ROC) curve (AUC), and the sensitivity and specificity were calculated. Results The indexes differed between LAM patients and healthy controls, S-LAM patients and healthy controls, and between TSC-LAM patients and healthy controls. There were two different metabolic indexes between S-LAM and TSC-LAM patients. After logistic regression modeling and ROC analysis, methionine (AUC =0.929, sensitivity =73.8%, specificity =100%, cut-off value =0.011 mmol/L) and acetic acid (AUC =0.966, sensitivity =95.1%, specificity =90%, cut-off value =0.006 mmol/L) had the highest diagnostic efficiency in LAM patients, and could be used to distinguish between affected and healthy people. Methionine was significantly associated with pneumothorax (P<0.05), and creatinine was significantly correlated with hysteromyoma (P<0.05). Conclusions Methionine and acetic acid in the plasma of LAM patients are potential biomarkers. Methionine was also associated with pneumothorax in LAM patients. Also, acetone and creatinine were promising metabolic markers to distinguish S-LAM from TSC-LAM. NMR as a new non-invasive diagnostic method had a good discriminatory power for LAM.
Collapse
Affiliation(s)
- Weili Gu
- Departments of Respiratory and Critical Care Medicine, Chronic Airways Diseases Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, China;,The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Yingxin Pan
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Wei Zhao
- Tianjin Key Laboratory of Clinical Multiomics, Tianjin, China
| | - Jie Liu
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Ying Meng
- Departments of Respiratory and Critical Care Medicine, Chronic Airways Diseases Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Huang Z, Chen CW, Buj R, Tangudu NK, Fang RS, Leon KE, Dahl ES, Varner EL, von Krusenstiern E, Cole AR, Snyder NW, Aird KM. ATM inhibition drives metabolic adaptation via induction of macropinocytosis. J Cell Biol 2023; 222:e202007026. [PMID: 36399181 PMCID: PMC9679964 DOI: 10.1083/jcb.202007026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 05/30/2022] [Accepted: 10/06/2022] [Indexed: 11/19/2022] Open
Abstract
Macropinocytosis is a nonspecific endocytic process that may enhance cancer cell survival under nutrient-poor conditions. Ataxia-Telangiectasia mutated (ATM) is a tumor suppressor that has been previously shown to play a role in cellular metabolic reprogramming. We report that the suppression of ATM increases macropinocytosis to promote cancer cell survival in nutrient-poor conditions. Combined inhibition of ATM and macropinocytosis suppressed proliferation and induced cell death both in vitro and in vivo. Supplementation of ATM-inhibited cells with amino acids, branched-chain amino acids (BCAAs) in particular, abrogated macropinocytosis. Analysis of ATM-inhibited cells in vitro demonstrated increased BCAA uptake, and metabolomics of ascites and interstitial fluid from tumors indicated decreased BCAAs in the microenvironment of ATM-inhibited tumors. These data reveal a novel basis of ATM-mediated tumor suppression whereby loss of ATM stimulates protumorigenic uptake of nutrients in part via macropinocytosis to promote cancer cell survival and reveal a potential metabolic vulnerability of ATM-inhibited cells.
Collapse
Affiliation(s)
- Zhentai Huang
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Chi-Wei Chen
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Raquel Buj
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Naveen Kumar Tangudu
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Richard S. Fang
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kelly E. Leon
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Biomedical Sciences Graduate Program, Penn State College of Medicine, Hershey, PA
| | - Erika S. Dahl
- Biomedical Sciences Graduate Program, Penn State College of Medicine, Hershey, PA
| | - Erika L. Varner
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University, Philadelphia, PA
| | - Eliana von Krusenstiern
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University, Philadelphia, PA
| | - Aidan R. Cole
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Nathaniel W. Snyder
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University, Philadelphia, PA
| | - Katherine M. Aird
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
13
|
MRI measurement of alanine uptake in a mouse xenograft model of U-87 MG glioblastoma. Magn Reson Imaging 2022; 93:189-194. [PMID: 36029935 DOI: 10.1016/j.mri.2022.08.015] [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: 06/03/2022] [Revised: 08/09/2022] [Accepted: 08/18/2022] [Indexed: 11/22/2022]
Abstract
The potential use of alanine as an MRI contrast agent was investigated. The relaxation properties of alanine solutions were measured at 9.4 T. The T2 relaxivity caused by the chemical exchange (R2ex) between amine protons and water protons was 0.10 mM-1 s-1 at 37 °C. As a demonstration, alanine uptake in a mouse xenograft model of U-87 MG glioblastoma was measured using MRI, and was compared with immunohistochemistry staining of ASCT2, a transporter that imports amino acids into cancer cells. Statistically significant (p = 0.0079) differences in ASCT2 distribution were found between regions that show strong and weak alanine uptake in MRI. To better understand the influence of perfusion, the effect of ASCT2 inhibition on the alanine uptake in MRI was investigated, and dynamic contrast enhanced MRI was compared with alanine MRI.
Collapse
|
14
|
Qiu Z, Liu W, Zhu Q, Ke K, Zhu Q, Jin W, Yu S, Yang Z, Li L, Sun X, Ren S, Liu Y, Zhu Z, Zeng J, Huang X, Huang Y, Wei L, Ma M, Lu J, Chen X, Mou Y, Xie T, Sui X. The Role and Therapeutic Potential of Macropinocytosis in Cancer. Front Pharmacol 2022; 13:919819. [PMID: 36046825 PMCID: PMC9421435 DOI: 10.3389/fphar.2022.919819] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/17/2022] [Indexed: 11/20/2022] Open
Abstract
Macropinocytosis, a unique endocytosis pathway characterized by nonspecific internalization, has a vital role in the uptake of extracellular substances and antigen presentation. It is known to have dual effects on cancer cells, depending on cancer type and certain microenvironmental conditions. It helps cancer cells survive in nutrient-deficient environments, enhances resistance to anticancer drugs, and promotes invasion and metastasis. Conversely, overexpression of the RAS gene alongside drug treatment can lead to methuosis, a novel mode of cell death. The survival and proliferation of cancer cells is closely related to macropinocytosis in the tumor microenvironment (TME), but identifying how these cells interface with the TME is crucial for creating drugs that can limit cancer progression and metastasis. Substantial progress has been made in recent years on designing anticancer therapies that utilize the effects of macropinocytosis. Both the induction and inhibition of macropinocytosis are useful strategies for combating cancer cells. This article systematically reviews the general mechanisms of macropinocytosis, its specific functions in tumor cells, its occurrence in nontumor cells in the TME, and its application in tumor therapies. The aim is to elucidate the role and therapeutic potential of macropinocytosis in cancer treatment.
Collapse
Affiliation(s)
- Zejing Qiu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Wencheng Liu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Qianru Zhu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Kun Ke
- Department of Gastrointestinal-Pancreatic Surgery, General Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Qicong Zhu
- Department of Gastrointestinal-Pancreatic Surgery, General Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Weiwei Jin
- Department of Gastrointestinal-Pancreatic Surgery, General Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Shuxian Yu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Zuyi Yang
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Lin Li
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Xiaochen Sun
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Shuyi Ren
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Yanfen Liu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Zhiyu Zhu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Jiangping Zeng
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Xiaoyu Huang
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Yan Huang
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Lu Wei
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Mengmeng Ma
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Jun Lu
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Xiaoyang Chen
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Yiping Mou
- Department of Gastrointestinal-Pancreatic Surgery, General Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
- *Correspondence: Yiping Mou, ; Tian Xie, ; Xinbing Sui,
| | - Tian Xie
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
- *Correspondence: Yiping Mou, ; Tian Xie, ; Xinbing Sui,
| | - Xinbing Sui
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
- *Correspondence: Yiping Mou, ; Tian Xie, ; Xinbing Sui,
| |
Collapse
|
15
|
Evers M, Song J, Shriwas P, Greenbaum HS, Chen X. From Transcriptomics, Metabolomics to Functional Studies: Extracellular ATP Induces TGF-β-Like Epithelial Mesenchymal Transition in Lung Cancer Cells. Front Oncol 2022; 12:912065. [PMID: 35847855 PMCID: PMC9282887 DOI: 10.3389/fonc.2022.912065] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/25/2022] [Indexed: 12/02/2022] Open
Abstract
We and others previously showed that extracellular ATP (eATP) is implicated in epithelial mesenchymal transition (EMT). However, the mechanisms by which eATP induces EMT and ATP’s relationship to TGF-β, a well-known EMT inducer, are largely unclear. Also, eATP-induced EMT has never been studied at transcriptomic and metabolomics levels. Based on our previous studies, we hypothesized that eATP acts as a specific inducer and regulator of EMT at all levels in cancer cells. RNAseq and metabolomics analyses were performed on human non-small cell lung cancer (NSCLC) A549 cells treated with either eATP or TGF-β. Bio-functional assays, such as invasion, intracellular ATP, cell proliferation, cytoskeleton remodeling, and others were conducted in NSCLC A549 and H1299 cells to validate changes observed from RNAseq and metabolomics studies. In the RNAseq study, eATP significantly enriched expressions of genes involved in EMT similarly to TGF-β after 2 and 6 hours of treatment. Samples treated with eATP for 2 hours share 131 upregulated EMT genes with those of TGF-β treated samples, and 42 genes at 6 hours treatment. Eleven genes, with known or unknown functions in EMT, are significantly upregulated by both inducers at both time points, have been identified. BLOC1S6, one of the 11 genes, was selected for further study. eATP induced numerous EMT-related changes in metabolic pathways, including cytoskeleton rearrangement, glycolysis, glutaminolysis, ROS, and individual metabolic changes similar to those induced by TGF-β. Functional bioassays verified the findings from RNAseq and metabolomics that eATP EMT-like changes in A549 and H1299 cells similarly to TGF-β. BLOC1S6 was found to be implicated in EMT. In these studies, eATP-induced EMT, at all levels examined, is similar but non-identical to that induced by TGF-β, and functions in such a way that exogenous addition of TGF-β is unnecessary for the induction. The study of BLOC1S6 further verified its potential roles in EMT and the RNAseq analysis results. All these strongly indicate that eATP is a multi-functional and multi-locational inducer and regulator of EMT, changing our thinking on how EMT is induced and regulated and pointing to new directions for inhibiting EMT in cancer.
Collapse
Affiliation(s)
- Maria Evers
- Honors Tutorial College, Ohio University, Athens, OH, United States
- The Ben May Department for Cancer Research, University of Chicago, Chicago, IL, United States
| | - Jingwen Song
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- The Molecular and Cellular Biology Program, Ohio University, Athens, OH, United States
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Pratik Shriwas
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Harrison S. Greenbaum
- The Ben May Department for Cancer Research, University of Chicago, Chicago, IL, United States
| | - Xiaozhuo Chen
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- The Molecular and Cellular Biology Program, Ohio University, Athens, OH, United States
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- Department of Biomedical Sciences, The Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
- *Correspondence: Xiaozhuo Chen,
| |
Collapse
|
16
|
Vanhove K, Derveaux E, Mesotten L, Thomeer M, Criel M, Mariën H, Adriaensens P. Unraveling the Rewired Metabolism in Lung Cancer Using Quantitative NMR Metabolomics. Int J Mol Sci 2022; 23:ijms23105602. [PMID: 35628415 PMCID: PMC9146819 DOI: 10.3390/ijms23105602] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 11/16/2022] Open
Abstract
Lung cancer cells are well documented to rewire their metabolism and energy production networks to enable proliferation and survival in a nutrient-poor and hypoxic environment. Although metabolite profiling of blood plasma and tissue is still emerging in omics approaches, several techniques have shown potential in cancer diagnosis. In this paper, the authors describe the alterations in the metabolic phenotype of lung cancer patients. In addition, we focus on the metabolic cooperation between tumor cells and healthy tissue. Furthermore, the authors discuss how metabolomics could improve the management of lung cancer patients.
Collapse
Affiliation(s)
- Karolien Vanhove
- Applied and Analytical Chemistry, Institute for Materials Research, Hasselt University, Agoralaan 1-Building D, B-3590 Diepenbeek, Belgium;
- Department of Respiratory Medicine, AZ Vesalius, Hazelereik 51, B-3700 Tongeren, Belgium
- Correspondence:
| | - Elien Derveaux
- Faculty of Medicine and Life Sciences, Hasselt University, Martelarenlaan 42, B-3500 Hasselt, Belgium; (E.D.); (H.M.)
| | - Liesbet Mesotten
- Department of Nuclear Medicine, Ziekenhuis Oost-Limburg, Schiepse Bos 6, B-3600 Genk, Belgium;
| | - Michiel Thomeer
- Department of Respiratory Medicine, Ziekenhuis Oost-Limburg, Schiepse Bos 6, B-3600 Genk, Belgium; (M.T.); (M.C.)
| | - Maarten Criel
- Department of Respiratory Medicine, Ziekenhuis Oost-Limburg, Schiepse Bos 6, B-3600 Genk, Belgium; (M.T.); (M.C.)
| | - Hanne Mariën
- Faculty of Medicine and Life Sciences, Hasselt University, Martelarenlaan 42, B-3500 Hasselt, Belgium; (E.D.); (H.M.)
| | - Peter Adriaensens
- Applied and Analytical Chemistry, Institute for Materials Research, Hasselt University, Agoralaan 1-Building D, B-3590 Diepenbeek, Belgium;
| |
Collapse
|
17
|
Abstract
Macropinocytosis is an evolutionarily conserved endocytic pathway that mediates the nonselective acquisition of extracellular material via large endocytic vesicles known as macropinosomes. In addition to other functions, this uptake pathway supports cancer cell metabolism through the uptake of nutrients. Cells harboring oncogene or tumor suppressor mutations are known to display heightened macropinocytosis, which confers to the cancer cells the ability to survive and proliferate despite the nutrient-scarce conditions of the tumor microenvironment. Thus, macropinocytosis is associated with cancer malignancy. Macropinocytic uptake can be induced in cancer cells by different stress stimuli, acting as an adaptive mechanism for the cells to resist stresses in the tumor milieu. Here, we review the cellular stresses that are known to promote macropinocytosis, as well as the underlying molecular mechanisms that drive this process.
Collapse
Affiliation(s)
- Guillem Lambies
- Cell and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Cosimo Commisso
- Cell and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
18
|
Seguin L. KRAS Addiction Promotes Cancer Cell Adaptation in Harsh Microenvironment Through Macropinocytosis. Subcell Biochem 2022; 98:189-204. [PMID: 35378709 DOI: 10.1007/978-3-030-94004-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
KRAS is the most frequently mutated oncogene in cancer and despite intensive studies, attempts to develop effective therapies targeting KRAS or its downstream signaling have failed mostly due to the complexity of KRAS activation and function in cancer initiation and progression. Over the years, KRAS has been involved in several biological processes including cell survival, proliferation, and metabolism by promoting not only a favorable tumor environment but also a cell-microenvironment dialog to allow cancer cells to adapt to tumor microenvironment scarcity. One of the mechanisms involved in this adaption is KRAS-mediated macropinocytosis. Macropinocytosis is an evolutionarily conserved, large-scale, and nonselective form of endocytosis involving actin-driven cell membrane remodeling to engulf large amounts of extracellular fluids and proteins from the local environment. While macropinocytosis process has been known for decades, recent gain interest due to its regulation of KRAS-driven tumor growth in adverse microenvironments. By promoting extracellular protein and other macromolecules internalization, macropinocytosis provides a survival mechanism under nutrient scarce conditions and the potential for unrestricted tumor growth. Thus, a better understanding of macropinocytotic process is needed to develop alternative therapeutic strategies.
Collapse
|
19
|
Abstract
Here we review the regulation of macropinocytosis by Wnt growth factor signaling. Canonical Wnt signaling is normally thought of as a regulator of nuclear β-catenin, but emerging results indicate that there is much more than β-catenin to the Wnt pathway. Macropinocytosis is transiently regulated by EGF-RTK-Ras-PI3K signaling. Recent studies show that Wnt signaling provides for sustained acquisition of nutrients by macropinocytosis. Endocytosis of Wnt-Lrp6-Fz receptor complexes triggers the sequestration of GSK3 and components of the cytosolic destruction complex such as Axin1 inside multivesicular bodies (MVBs) through the action of the ESCRT machinery. Wnt macropinocytosis can be induced both by the transcriptional loop of stabilized β-catenin, and by the inhibition of GSK3 even in the absence of new protein synthesis. The cell is poised for macropinocytosis, and all it requires for triggering of Pak1 and the actin machinery is the inhibition of GSK3. Striking lysosomal acidification, which requires macropinocytosis, is induced by GSK3 chemical inhibitors or Wnt protein. Wnt-induced macropinocytosis requires the ESCRT machinery that forms MVBs. In cancer cells, mutations in the tumor suppressors APC and Axin1 result in extensive macropinocytosis, which can be reversed by restoring wild-type protein. In basal cellular conditions, GSK3 functions to constitutively repress macropinocytosis.
Collapse
|
20
|
Means N, Elechalawar CK, Chen WR, Bhattacharya R, Mukherjee P. Revealing macropinocytosis using nanoparticles. Mol Aspects Med 2022; 83:100993. [PMID: 34281720 PMCID: PMC8761201 DOI: 10.1016/j.mam.2021.100993] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/12/2021] [Indexed: 02/03/2023]
Abstract
Endocytosis mechanisms are one of the methods that cells use to interact with their environments. Endocytosis mechanisms vary from the clathrin-mediated endocytosis to the receptor independent macropinocytosis. Macropinocytosis is a niche of endocytosis that is quickly becoming more relevant in various fields of research since its discovery in the 1930s. Macropinocytosis has several distinguishing factors from other receptor-mediated forms of endocytosis, including: types of extracellular material for uptake, signaling cascade, and niche uses between cell types. Nanoparticles (NPs) are an important tool for various applications, including drug delivery and disease treatment. However, surface engineering of NPs could be tailored to target them inside the cells exploiting different endocytosis pathways, such as endocytosis versus macropinocytosis. Such surface engineering of NPs mainly, size, charge, shape and the core material will allow identification of new adapter molecules regulating different endocytosis process and provide further insight into how cells tweak these pathways to meet their physiological need. In this review, we focus on the description of macropinocytosis, a lesser studied endocytosis mechanism than the conventional receptor mediated endocytosis. Additionally, we will discuss nanoparticle endocytosis (including macropinocytosis), and how the physio-chemical properties of the NP (size, charge, and surface coating) affect their intracellular uptake and exploiting them as tools to identify new adapter molecules regulating these processes.
Collapse
Affiliation(s)
- Nicolas Means
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Wei R Chen
- Stephenson School of Biomedical Engineering, Gallogly College of Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Resham Bhattacharya
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Priyabrata Mukherjee
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
21
|
Abstract
Macropinocytosis is a critical route of nutrient acquisition in pancreatic cancer cells. Constitutive macropinocytosis is promoted by mutant KRAS, which activates the PI3Kα lipid kinase and RAC1, to drive membrane ruffling, macropinosome uptake and processing. However, our recent study on the KRASG12R mutant indicated the presence of a KRAS-independent mode of macropinocytosis in pancreatic cancer cell lines, thereby increasing the complexity of this process. We found that KRASG12R-mutant cell lines promote macropinocytosis independent of KRAS activity using PI3Kγ and RAC1, highlighting the convergence of regulation on RAC signaling. While macropinocytosis has been proposed to be a therapeutic target for the treatment of pancreatic cancer, our studies have underscored how little we understand about the activation and regulation of this metabolic process. Therefore, this review seeks to highlight the differences in macropinocytosis regulation in the two cellular subtypes while also highlighting the features that make the KRASG12R mutant atypical.
Collapse
Affiliation(s)
- G Aaron Hobbs
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA.
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA.
| | - Channing J Der
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
22
|
Hanada K, Kawada K, Nishikawa G, Toda K, Maekawa H, Nishikawa Y, Masui H, Hirata W, Okamoto M, Kiyasu Y, Honma S, Ogawa R, Mizuno R, Itatani Y, Miyoshi H, Sasazuki T, Shirasawa S, Taketo MM, Obama K, Sakai Y. Dual blockade of macropinocytosis and asparagine bioavailability shows synergistic anti-tumor effects on KRAS-mutant colorectal cancer. Cancer Lett 2021; 522:129-141. [PMID: 34543685 DOI: 10.1016/j.canlet.2021.09.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 02/07/2023]
Abstract
Mutations of KRAS gene are found in various types of cancer, including colorectal cancer (CRC). Despite intense efforts, no pharmacological approaches are expected to be effective against KRAS-mutant cancers. Macropinocytosis is an evolutionarily conserved actin-dependent endocytic process that internalizes extracellular fluids into large vesicles called macropinosomes. Recent studies have revealed macropinocytosis's important role in metabolic adaptation to nutrient stress in cancer cells harboring KRAS mutations. Here we showed that KRAS-mutant CRC cells enhanced macropinocytosis for tumor growth under nutrient-depleted conditions. We also demonstrated that activation of Rac1 and phosphoinositide 3-kinase were involved in macropinocytosis of KRAS-mutant CRC cells. Furthermore, we found that macropinocytosis was closely correlated with asparagine metabolism. In KRAS-mutant CRC cells engineered with knockdown of asparagine synthetase, macropinocytosis was accelerated under glutamine-depleted condition, and albumin addition could restore the glutamine depletion-induced growth suppression by recovering the intracellular asparagine level. Finally, we discovered that the combination of macropinocytosis inhibition and asparagine depletion dramatically suppressed the tumor growth of KRAS-mutant CRC cells in vivo. These results indicate that dual blockade of macropinocytosis and asparagine bioavailability could be a novel therapeutic strategy for KRAS-mutant cancers.
Collapse
Affiliation(s)
- Keita Hanada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenji Kawada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Gen Nishikawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kosuke Toda
- Department of Surgery, Otsu City Hospital, Otsu, Japan
| | - Hisatsugu Maekawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuyo Nishikawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideyuki Masui
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Wataru Hirata
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Michio Okamoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshiyuki Kiyasu
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shusaku Honma
- Department of Surgery, Kobe City Medical Center West Hospital, Kobe, Japan
| | - Ryotaro Ogawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Rei Mizuno
- Department of Surgery, Uji Tokushukai Medical Center, Kyoto, Japan
| | - Yoshiro Itatani
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Miyoshi
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | | | - Senji Shirasawa
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - M Mark Taketo
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Kazutaka Obama
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshiharu Sakai
- Department of Surgery, Osaka Red Cross Hospital, Osaka, Japan
| |
Collapse
|
23
|
Kerk SA, Papagiannakopoulos T, Shah YM, Lyssiotis CA. Metabolic networks in mutant KRAS-driven tumours: tissue specificities and the microenvironment. Nat Rev Cancer 2021; 21:510-525. [PMID: 34244683 DOI: 10.1038/s41568-021-00375-9] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Oncogenic mutations in KRAS drive common metabolic programmes that facilitate tumour survival, growth and immune evasion in colorectal carcinoma, non-small-cell lung cancer and pancreatic ductal adenocarcinoma. However, the impacts of mutant KRAS signalling on malignant cell programmes and tumour properties are also dictated by tumour suppressor losses and physiological features specific to the cell and tissue of origin. Here we review convergent and disparate metabolic networks regulated by oncogenic mutant KRAS in colon, lung and pancreas tumours, with an emphasis on co-occurring mutations and the role of the tumour microenvironment. Furthermore, we explore how these networks can be exploited for therapeutic gain.
Collapse
Affiliation(s)
- Samuel A Kerk
- Doctoral Program in Cancer Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Thales Papagiannakopoulos
- Department of Pathology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Yatrik M Shah
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
24
|
Endicott M, Jones M, Hull J. Amino acid metabolism as a therapeutic target in cancer: a review. Amino Acids 2021; 53:1169-1179. [PMID: 34292410 PMCID: PMC8325646 DOI: 10.1007/s00726-021-03052-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/16/2021] [Indexed: 02/04/2023]
Abstract
Malignant cells often demonstrate a proliferative advantage when compared to non-malignant cells. However, the rapid growth and metabolism required for survival can also highlight vulnerabilities specific to these malignant cells. One such vulnerability exhibited by cancer is an increased demand for amino acids (AAs), which often results in a dependency on exogenous sources of AAs or requires upregulation of de novo synthesis. These metabolic alterations can be exploited by therapy, which aims to improve treatment outcome and decrease relapse and reoccurrence. One clinically utilised strategy targeting AA dependency is the use of asparaginase in the treatment of acute lymphoblastic leukaemia (ALL), which results in a depletion of exogenous asparagine and subsequent cancer cell death. Examples of other successful strategies include the exploitation of arginine deiminase and methioninase, nutrient restriction of methionine and the inhibition of glutaminase. In this review, we summarise these treatment strategies into three promising avenues: AA restriction, enzymatic depletion and inhibition of metabolism. This review provides an insight into the complexity of metabolism in cancer, whilst highlighting these three current research avenues that have support in both preclinical and clinical settings.
Collapse
Affiliation(s)
- Molly Endicott
- Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Michael Jones
- Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Jonathon Hull
- Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.
| |
Collapse
|
25
|
Abstract
Wnt signaling has multiple functions beyond the transcriptional effects of β-catenin stabilization. We review recent investigations that uncover new cell physiological effects through the regulation of Wnt receptor endocytosis, Wnt-induced stabilization of proteins (Wnt-STOP), macropinocytosis, increase in lysosomal activity, and metabolic changes. Many of these growth-promoting effects of canonical Wnt occur within minutes and are independent of new protein synthesis. A key element is the sequestration of glycogen synthase kinase 3 (GSK3) inside multivesicular bodies and lysosomes. Twenty percent of human proteins contain consecutive GSK3 phosphorylation motifs, which in the absence of Wnt can form phosphodegrons for polyubiquitination and proteasomal degradation. Wnt signaling by either the pharmacological inhibition of GSK3 or the loss of tumor-suppressor proteins, such as adenomatous polyposis coli (APC) and Axin1, increases lysosomal acidification, anabolic metabolites, and macropinocytosis, which is normally repressed by the GSK3-Axin1-APC destruction complex. The combination of these cell physiological effects drives cell growth. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Lauren V Albrecht
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California 90095-1662, USA;
| | - Nydia Tejeda-Muñoz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California 90095-1662, USA;
| | - Edward M De Robertis
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California 90095-1662, USA;
| |
Collapse
|
26
|
Wang Y, Song M, Liu M, Zhang G, Zhang X, Li MO, Ma X, Zhang JJ, Huang XY. Fascin inhibitor increases intratumoral dendritic cell activation and anti-cancer immunity. Cell Rep 2021; 35:108948. [PMID: 33826900 PMCID: PMC8050791 DOI: 10.1016/j.celrep.2021.108948] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 12/21/2020] [Accepted: 03/15/2021] [Indexed: 12/21/2022] Open
Abstract
Fascin protein is the main actin-bundling protein in filopodia and invadopodia, which are critical for tumor cell migration, invasion, and metastasis. Small-molecule fascin inhibitors block tumor invasion and metastasis and increase the overall survival of tumor-bearing mice. Here, we report a finding that fascin blockade additionally reinvigorates anti-tumor immune response in syngeneic mouse models of various cancers. Fascin protein levels are increased in conventional dendritic cells (cDCs) in the tumor microenvironment. Mechanistically, fascin inhibitor NP-G2-044 increases the number of intratumoral-activated cDCs and enhances the antigen uptake by cDCs. Furthermore, together with PD-1 blocking antibody, NP-G2-044 markedly increases the number of activated CD8+ T cells in the otherwise anti-PD-1 refractory tumors. Reduction of fascin levels in cDCs, but not fascin gene knockout in tumor cells, mimics the anti-tumor immune effect of NP-G2-044. These data demonstrate that fascin inhibitor NP-G2-044 simultaneously limits tumor metastasis and reinvigorates anti-tumor immune responses.
Collapse
Affiliation(s)
- Yufeng Wang
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Mei Song
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Ming Liu
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Guoan Zhang
- Proteomics and Metabolomics Core Facility, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Xian Zhang
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ming O Li
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Xiaojing Ma
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine of Cornell University, New York, NY 10065, USA
| | | | - Xin-Yun Huang
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine of Cornell University, New York, NY 10065, USA.
| |
Collapse
|
27
|
Trkulja CL, Jungholm O, Davidson M, Jardemark K, Marcus MM, Hägglund J, Karlsson A, Karlsson R, Bruton J, Ivarsson N, Srinivasa SP, Cavallin A, Svensson P, Jeffries GDM, Christakopoulou MN, Reymer A, Ashok A, Willman G, Papadia D, Johnsson E, Orwar O. Rational antibody design for undruggable targets using kinetically controlled biomolecular probes. SCIENCE ADVANCES 2021; 7:7/16/eabe6397. [PMID: 33863724 PMCID: PMC8051879 DOI: 10.1126/sciadv.abe6397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 02/26/2021] [Indexed: 05/07/2023]
Abstract
Several important drug targets, e.g., ion channels and G protein-coupled receptors, are extremely difficult to approach with current antibody technologies. To address these targets classes, we explored kinetically controlled proteases as structural dynamics-sensitive druggability probes in native-state and disease-relevant proteins. By using low-Reynolds number flows, such that a single or a few protease incisions are made, we could identify antibody binding sites (epitopes) that were translated into short-sequence antigens for antibody production. We obtained molecular-level information of the epitope-paratope region and could produce high-affinity antibodies with programmed pharmacological function against difficult-to-drug targets. We demonstrate the first stimulus-selective monoclonal antibodies targeting the transient receptor potential vanilloid 1 (TRPV1) channel, a clinically validated pain target widely considered undruggable with antibodies, and apoptosis-inducing antibodies selectively mediating cytotoxicity in KRAS-mutated cells. It is our hope that this platform will widen the scope of antibody therapeutics for the benefit of patients.
Collapse
Affiliation(s)
| | - Oscar Jungholm
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Max Davidson
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden
| | - Kent Jardemark
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Monica M Marcus
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Jessica Hägglund
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Anders Karlsson
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden
- Nanoxis Consulting AB, SE-40016 Gothenburg, Sweden
| | - Roger Karlsson
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden
- Nanoxis Consulting AB, SE-40016 Gothenburg, Sweden
| | - Joseph Bruton
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Niklas Ivarsson
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | - Alexandra Cavallin
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Peder Svensson
- Integrative Research Laboratories, SE-41346, Gothenburg, Sweden
| | | | | | - Anna Reymer
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-405 30 Gothenburg, Sweden
| | | | | | | | - Emma Johnsson
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden
| | - Owe Orwar
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden.
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
28
|
Selvaraj G, Kaliamurthi S, Peslherbe GH, Wei DQ. Identifying potential drug targets and candidate drugs for COVID-19: biological networks and structural modeling approaches. F1000Res 2021; 10:127. [PMID: 33968364 PMCID: PMC8080978 DOI: 10.12688/f1000research.50850.2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 11/08/2024] Open
Abstract
Background: Coronavirus (CoV) is an emerging human pathogen causing severe acute respiratory syndrome (SARS) around the world. Earlier identification of biomarkers for SARS can facilitate detection and reduce the mortality rate of the disease. Thus, by integrated network analysis and structural modeling approach, we aimed to explore the potential drug targets and the candidate drugs for coronavirus medicated SARS. Methods: Differentially expression (DE) analysis of CoV infected host genes (HGs) expression profiles was conducted by using the Limma. Highly integrated DE-CoV-HGs were selected to construct the protein-protein interaction (PPI) network. Results: Using the Walktrap algorithm highly interconnected modules include module 1 (202 nodes); module 2 (126 nodes) and module 3 (121 nodes) modules were retrieved from the PPI network. MYC, HDAC9, NCOA3, CEBPB, VEGFA, BCL3, SMAD3, SMURF1, KLHL12, CBL, ERBB4, and CRKL were identified as potential drug targets (PDTs), which are highly expressed in the human respiratory system after CoV infection. Functional terms growth factor receptor binding, c-type lectin receptor signaling, interleukin-1 mediated signaling, TAP dependent antigen processing and presentation of peptide antigen via MHC class I, stimulatory T cell receptor signaling, and innate immune response signaling pathways, signal transduction and cytokine immune signaling pathways were enriched in the modules. Protein-protein docking results demonstrated the strong binding affinity (-314.57 kcal/mol) of the ERBB4-3cLpro complex which was selected as a drug target. In addition, molecular dynamics simulations indicated the structural stability and flexibility of the ERBB4-3cLpro complex. Further, Wortmannin was proposed as a candidate drug to ERBB4 to control SARS-CoV-2 pathogenesis through inhibit receptor tyrosine kinase-dependent macropinocytosis, MAPK signaling, and NF-kb singling pathways that regulate host cell entry, replication, and modulation of the host immune system. Conclusion: We conclude that CoV drug target "ERBB4" and candidate drug "Wortmannin" provide insights on the possible personalized therapeutics for emerging COVID-19.
Collapse
Affiliation(s)
- Gurudeeban Selvaraj
- Centre for Research in Molecular Modeling, Concordia University, Montreal, Quebec, H4B 1R6, Canada
- Centre of Interdisciplinary Science-Computational Life Sciences, College of Chemistry and Chemical Engineering,, Henan University of Technology, Zhengzhou, Henan, 450001, China
| | - Satyavani Kaliamurthi
- Centre for Research in Molecular Modeling, Concordia University, Montreal, Quebec, H4B 1R6, Canada
- Centre of Interdisciplinary Science-Computational Life Sciences, College of Chemistry and Chemical Engineering,, Henan University of Technology, Zhengzhou, Henan, 450001, China
| | - Gilles H. Peslherbe
- Centre for Research in Molecular Modeling, Concordia University, Montreal, Quebec, H4B 1R6, Canada
| | - Dong-Qing Wei
- Centre of Interdisciplinary Science-Computational Life Sciences, College of Chemistry and Chemical Engineering,, Henan University of Technology, Zhengzhou, Henan, 450001, China
- The State Key Laboratory of Microbial Metabolism, College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, Shanghai, 200240, China
- IASIA (International Association of Scientists in the Interdisciplinary Areas), 125 Boul. de Bromont, Quebec, J2L 2K7, Canada
| |
Collapse
|
29
|
Grasmann G, Mondal A, Leithner K. Flexibility and Adaptation of Cancer Cells in a Heterogenous Metabolic Microenvironment. Int J Mol Sci 2021; 22:1476. [PMID: 33540663 PMCID: PMC7867260 DOI: 10.3390/ijms22031476] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023] Open
Abstract
The metabolic microenvironment, comprising all soluble and insoluble nutrients and co-factors in the extracellular milieu, has a major impact on cancer cell proliferation and survival. A large body of evidence from recent studies suggests that tumor cells show a high degree of metabolic flexibility and adapt to variations in nutrient availability. Insufficient vascular networks and an imbalance of supply and demand shape the metabolic tumor microenvironment, which typically contains a lower concentration of glucose compared to normal tissues. The present review sheds light on the recent literature on adaptive responses in cancer cells to nutrient deprivation. It focuses on the utilization of alternative nutrients in anabolic metabolic pathways in cancer cells, including soluble metabolites and macromolecules and outlines the role of central metabolic enzymes conferring metabolic flexibility, like gluconeogenesis enzymes. Moreover, a conceptual framework for potential therapies targeting metabolically flexible cancer cells is presented.
Collapse
Affiliation(s)
- Gabriele Grasmann
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (G.G.); (A.M.)
| | - Ayusi Mondal
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (G.G.); (A.M.)
| | - Katharina Leithner
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (G.G.); (A.M.)
- BioTechMed-Graz, A-8010 Graz, Austria
| |
Collapse
|
30
|
Song S, Zhang Y, Ding T, Ji N, Zhao H. The Dual Role of Macropinocytosis in Cancers: Promoting Growth and Inducing Methuosis to Participate in Anticancer Therapies as Targets. Front Oncol 2021; 10:570108. [PMID: 33542897 PMCID: PMC7851083 DOI: 10.3389/fonc.2020.570108] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 12/01/2020] [Indexed: 02/05/2023] Open
Abstract
Macropinocytosis is an important mechanism of internalizing extracellular materials and dissolved molecules in eukaryotic cells. Macropinocytosis has a dual effect on cancer cells. On the one hand, cells expressing RAS genes (such as K-RAS, H-RAS) under the stress of nutrient deficiency can spontaneously produce constitutive macropinocytosis to promote the growth of cancer cells by internalization of extracellular nutrients (like proteins), receptors, and extracellular vesicles(EVs). On the other hand, abnormal expression of RAS genes and drug treatment (such as MOMIPP) can induce a novel cell death associated with hyperactivated macropinocytosis: methuosis. Based on the dual effect, there is immense potential for designing anticancer therapies that target macropinocytosis in cancer cells. In view of the fact that there has been little review of the dual effect of macropinocytosis in cancer cells, herein, we systematically review the general process of macropinocytosis, its specific manifestation in cancer cells, and its application in cancer treatment, including anticancer drug delivery and destruction of macropinocytosis. This review aims to serve as a reference for studying macropinocytosis in cancers and designing macropinocytosis-targeting anticancer drugs in the future.
Collapse
Affiliation(s)
- Shaojuan Song
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tingting Ding
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hang Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Xiao F, Li J, Huang K, Li X, Xiong Y, Wu M, Wu L, Kuang W, Lv S, Wu L, Zhu X, Guo H. Macropinocytosis: mechanism and targeted therapy in cancers. Am J Cancer Res 2021; 11:14-30. [PMID: 33520357 PMCID: PMC7840718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/16/2020] [Indexed: 06/12/2023] Open
Abstract
Macropinocytosis is a form of endocytosis which provides an effective way for non-selective uptakes of extracellular proteins, liquids, and particles. The endocytic process is initiated by the activation of the growth factors signaling pathways. After activation of the biochemical signal, the cell starts internalizing extracellular solutes and nutrients into the irregular endocytic vesicles, known as macropinosomes that deliver them into the lysosomes for degradation. Macropinocytosis plays an important role in the nutritional supply of cancer cells. Due to the rapid expansion of cancer cells and the abnormal vascular microenvironment, cancer cells are usually deprived of oxygen and nutrients. Therefore, they must transform their metabolism to survive and grow in this harsh microenvironment. To satisfy their energy needs, cancer cells enhance the activity of macropinocytosis. Therefore, this metabolic adaptation that is used by cancer cells can be exploited to develop new targeted cancer therapies. In this review, we discuss the molecular mechanism that actuates the process of macropinocytosis in a variety of cancers, and the novel anti-cancer therapeutics in targeting macropinocytosis.
Collapse
Affiliation(s)
- Feng Xiao
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Jingying Li
- Department of Comprehensive Intensive Care Unit, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Kai Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Xin Li
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Yaping Xiong
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Miaojing Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Lei Wu
- Department of Emergency, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Wei Kuang
- Department of Emergency, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Shigang Lv
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Lei Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Hua Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang UniversityNanchang 330006, Jiangxi, China
| |
Collapse
|
32
|
Selvaraj G, Kaliamurthi S, Peslherbe GH, Wei DQ. Identifying potential drug targets and candidate drugs for COVID-19: biological networks and structural modeling approaches. F1000Res 2021; 10:127. [PMID: 33968364 PMCID: PMC8080978 DOI: 10.12688/f1000research.50850.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/01/2021] [Indexed: 02/05/2023] Open
Abstract
Background: Coronavirus (CoV) is an emerging human pathogen causing severe acute respiratory syndrome (SARS) around the world. Earlier identification of biomarkers for SARS can facilitate detection and reduce the mortality rate of the disease. Thus, by integrated network analysis and structural modeling approach, we aimed to explore the potential drug targets and the candidate drugs for coronavirus medicated SARS. Methods: Differentially expression (DE) analysis of CoV infected host genes (HGs) expression profiles was conducted by using the Limma. Highly integrated DE-CoV-HGs were selected to construct the protein-protein interaction (PPI) network. Results: Using the Walktrap algorithm highly interconnected modules include module 1 (202 nodes); module 2 (126 nodes) and module 3 (121 nodes) modules were retrieved from the PPI network. MYC, HDAC9, NCOA3, CEBPB, VEGFA, BCL3, SMAD3, SMURF1, KLHL12, CBL, ERBB4, and CRKL were identified as potential drug targets (PDTs), which are highly expressed in the human respiratory system after CoV infection. Functional terms growth factor receptor binding, c-type lectin receptor signaling, interleukin-1 mediated signaling, TAP dependent antigen processing and presentation of peptide antigen via MHC class I, stimulatory T cell receptor signaling, and innate immune response signaling pathways, signal transduction and cytokine immune signaling pathways were enriched in the modules. Protein-protein docking results demonstrated the strong binding affinity (-314.57 kcal/mol) of the ERBB4-3cLpro complex which was selected as a drug target. In addition, molecular dynamics simulations indicated the structural stability and flexibility of the ERBB4-3cLpro complex. Further, Wortmannin was proposed as a candidate drug to ERBB4 to control SARS-CoV-2 pathogenesis through inhibit receptor tyrosine kinase-dependent macropinocytosis, MAPK signaling, and NF-kb singling pathways that regulate host cell entry, replication, and modulation of the host immune system. Conclusion: We conclude that CoV drug target "ERBB4" and candidate drug "Wortmannin" provide insights on the possible personalized therapeutics for emerging COVID-19.
Collapse
Affiliation(s)
- Gurudeeban Selvaraj
- Centre for Research in Molecular Modeling, Concordia University, Montreal, Quebec, H4B 1R6, Canada
- Centre of Interdisciplinary Science-Computational Life Sciences, College of Chemistry and Chemical Engineering,, Henan University of Technology, Zhengzhou, Henan, 450001, China
| | - Satyavani Kaliamurthi
- Centre for Research in Molecular Modeling, Concordia University, Montreal, Quebec, H4B 1R6, Canada
- Centre of Interdisciplinary Science-Computational Life Sciences, College of Chemistry and Chemical Engineering,, Henan University of Technology, Zhengzhou, Henan, 450001, China
| | - Gilles H. Peslherbe
- Centre for Research in Molecular Modeling, Concordia University, Montreal, Quebec, H4B 1R6, Canada
| | - Dong-Qing Wei
- Centre of Interdisciplinary Science-Computational Life Sciences, College of Chemistry and Chemical Engineering,, Henan University of Technology, Zhengzhou, Henan, 450001, China
- The State Key Laboratory of Microbial Metabolism, College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, Shanghai, 200240, China
- IASIA (International Association of Scientists in the Interdisciplinary Areas), 125 Boul. de Bromont, Quebec, J2L 2K7, Canada
| |
Collapse
|
33
|
Selvaraj G, Kaliamurthi S, Peslherbe GH, Wei DQ. Identifying potential drug targets and candidate drugs for COVID-19: biological networks and structural modeling approaches. F1000Res 2021; 10:127. [PMID: 33968364 PMCID: PMC8080978 DOI: 10.12688/f1000research.50850.3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 02/05/2023] Open
Abstract
Background: Coronavirus (CoV) is an emerging human pathogen causing severe acute respiratory syndrome (SARS) around the world. Earlier identification of biomarkers for SARS can facilitate detection and reduce the mortality rate of the disease. Thus, by integrated network analysis and structural modeling approach, we aimed to explore the potential drug targets and the candidate drugs for coronavirus medicated SARS. Methods: Differentially expression (DE) analysis of CoV infected host genes (HGs) expression profiles was conducted by using the Limma. Highly integrated DE-CoV-HGs were selected to construct the protein-protein interaction (PPI) network. Results: Using the Walktrap algorithm highly interconnected modules include module 1 (202 nodes); module 2 (126 nodes) and module 3 (121 nodes) modules were retrieved from the PPI network. MYC, HDAC9, NCOA3, CEBPB, VEGFA, BCL3, SMAD3, SMURF1, KLHL12, CBL, ERBB4, and CRKL were identified as potential drug targets (PDTs), which are highly expressed in the human respiratory system after CoV infection. Functional terms growth factor receptor binding, c-type lectin receptor signaling, interleukin-1 mediated signaling, TAP dependent antigen processing and presentation of peptide antigen via MHC class I, stimulatory T cell receptor signaling, and innate immune response signaling pathways, signal transduction and cytokine immune signaling pathways were enriched in the modules. Protein-protein docking results demonstrated the strong binding affinity (-314.57 kcal/mol) of the ERBB4-3cLpro complex which was selected as a drug target. In addition, molecular dynamics simulations indicated the structural stability and flexibility of the ERBB4-3cLpro complex. Further, Wortmannin was proposed as a candidate drug to ERBB4 to control SARS-CoV-2 pathogenesis through inhibit receptor tyrosine kinase-dependent macropinocytosis, MAPK signaling, and NF-kb singling pathways that regulate host cell entry, replication, and modulation of the host immune system. Conclusion: We conclude that CoV drug target "ERBB4" and candidate drug "Wortmannin" provide insights on the possible personalized therapeutics for emerging COVID-19.
Collapse
Affiliation(s)
- Gurudeeban Selvaraj
- Centre for Research in Molecular Modeling, Concordia University, Montreal, Quebec, H4B 1R6, Canada
- Centre of Interdisciplinary Science-Computational Life Sciences, College of Chemistry and Chemical Engineering,, Henan University of Technology, Zhengzhou, Henan, 450001, China
| | - Satyavani Kaliamurthi
- Centre for Research in Molecular Modeling, Concordia University, Montreal, Quebec, H4B 1R6, Canada
- Centre of Interdisciplinary Science-Computational Life Sciences, College of Chemistry and Chemical Engineering,, Henan University of Technology, Zhengzhou, Henan, 450001, China
| | - Gilles H. Peslherbe
- Centre for Research in Molecular Modeling, Concordia University, Montreal, Quebec, H4B 1R6, Canada
| | - Dong-Qing Wei
- Centre of Interdisciplinary Science-Computational Life Sciences, College of Chemistry and Chemical Engineering,, Henan University of Technology, Zhengzhou, Henan, 450001, China
- The State Key Laboratory of Microbial Metabolism, College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, Shanghai, 200240, China
- IASIA (International Association of Scientists in the Interdisciplinary Areas), 125 Boul. de Bromont, Quebec, J2L 2K7, Canada
| |
Collapse
|
34
|
Albrecht LV, Tejeda-Muñoz N, De Robertis EM. Protocol for Probing Regulated Lysosomal Activity and Function in Living Cells. STAR Protoc 2020; 1:100132. [PMID: 33377026 PMCID: PMC7757114 DOI: 10.1016/j.xpro.2020.100132] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Lysosomes are the catabolic center of the cell. Limitations of many lysosomal tracers include low specificity and lack of reliable physiological readouts for changes in growth factor-regulated lysosomal activity. The imaging-based protocols described here provide insights at the cellular level to quantify functions essential to lysosomal biology, including β-glucosidase enzymatic cleavage, active Cathepsin D, and pH regulation in real time. These optimized protocols, applied in different cell types and pathophysiologic contexts, provide useful tools to study lysosome function in cultured living cells. For complete details on the use and execution of this protocol, please refer to Albrecht et al. (2020).
Collapse
Affiliation(s)
- L V Albrecht
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles 90095-1662, USA
| | - N Tejeda-Muñoz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles 90095-1662, USA
| | - E M De Robertis
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles 90095-1662, USA
| |
Collapse
|
35
|
Oncology Therapeutics Targeting the Metabolism of Amino Acids. Cells 2020; 9:cells9081904. [PMID: 32824193 PMCID: PMC7463463 DOI: 10.3390/cells9081904] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/19/2022] Open
Abstract
Amino acid metabolism promotes cancer cell proliferation and survival by supporting building block synthesis, producing reducing agents to mitigate oxidative stress, and generating immunosuppressive metabolites for immune evasion. Malignant cells rewire amino acid metabolism to maximize their access to nutrients. Amino acid transporter expression is upregulated to acquire amino acids from the extracellular environment. Under nutrient depleted conditions, macropinocytosis can be activated where proteins from the extracellular environment are engulfed and degraded into the constituent amino acids. The demand for non-essential amino acids (NEAAs) can be met through de novo synthesis pathways. Cancer cells can alter various signaling pathways to boost amino acid usage for the generation of nucleotides, reactive oxygen species (ROS) scavenging molecules, and oncometabolites. The importance of amino acid metabolism in cancer proliferation makes it a potential target for therapeutic intervention, including via small molecules and antibodies. In this review, we will delineate the targets related to amino acid metabolism and promising therapeutic approaches.
Collapse
|
36
|
Albrecht LV, Tejeda-Muñoz N, Bui MH, Cicchetto AC, Di Biagio D, Colozza G, Schmid E, Piccolo S, Christofk HR, De Robertis EM. GSK3 Inhibits Macropinocytosis and Lysosomal Activity through the Wnt Destruction Complex Machinery. Cell Rep 2020; 32:107973. [PMID: 32726636 PMCID: PMC7666578 DOI: 10.1016/j.celrep.2020.107973] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/29/2020] [Accepted: 07/08/2020] [Indexed: 12/17/2022] Open
Abstract
Canonical Wnt signaling is emerging as a major regulator of endocytosis. Here, we report that Wnt-induced macropinocytosis is regulated through glycogen synthase kinase 3 (GSK3) and the β-catenin destruction complex. We find that mutation of Axin1, a tumor suppressor and component of the destruction complex, results in the activation of macropinocytosis. Surprisingly, inhibition of GSK3 by lithium chloride (LiCl), CHIR99021, or dominant-negative GSK3 triggers macropinocytosis. GSK3 inhibition causes a rapid increase in acidic endolysosomes that is independent of new protein synthesis. GSK3 inhibition or Axin1 mutation increases lysosomal activity, which can be followed with tracers of active cathepsin D, β-glucosidase, and ovalbumin degradation. Microinjection of LiCl into the blastula cavity of Xenopus embryos causes a striking increase in dextran macropinocytosis. The effects of GSK3 inhibition on protein degradation in endolysosomes are blocked by the macropinocytosis inhibitors EIPA or IPA-3, suggesting that increases in membrane trafficking drive lysosomal activity.
Collapse
Affiliation(s)
- Lauren V Albrecht
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Nydia Tejeda-Muñoz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Maggie H Bui
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Andrew C Cicchetto
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Daniele Di Biagio
- Department of Molecular Medicine, University of Padua School of Medicine, viale Colombo 3, 35126 Padua, Italy
| | - Gabriele Colozza
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Ernst Schmid
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua School of Medicine, viale Colombo 3, 35126 Padua, Italy
| | - Heather R Christofk
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Edward M De Robertis
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA.
| |
Collapse
|
37
|
Caiola E, Colombo M, Sestito G, Lupi M, Marabese M, Pastorelli R, Broggini M, Brunelli L. Glutaminase Inhibition on NSCLC Depends on Extracellular Alanine Exploitation. Cells 2020; 9:cells9081766. [PMID: 32718002 PMCID: PMC7465377 DOI: 10.3390/cells9081766] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 12/12/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) cell lines vary in their sensitivity to glutaminase inhibitors, so it is important to identify the metabolic assets underling their efficacy in cancer cells. Even though specific genetic lesions such as in KRAS and LKB1 have been associated with reliance on glutamine for their metabolic needs, we found no distinction between glutaminase inhibitor CB-839 sensitivity and resistant phenotypes in NSCLC cells with or without these genetic alterations. We demonstrated the close relationship between environmental alanine uptake and catabolism. This response depended on the individual cell’s ability to employ alanine aminotransferase (GPT2) to compensate the reduced glutamate availability. It may, therefore, be useful to determine GPT2 levels to predict which NSCLC patients would benefit most from glutaminase inhibitor treatment.
Collapse
Affiliation(s)
- Elisa Caiola
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (E.C.); (M.C.); (M.B.)
| | - Marika Colombo
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (E.C.); (M.C.); (M.B.)
| | - Giovanna Sestito
- Laboratory of Mass Spectrometry, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (G.S.); (R.P.)
| | - Monica Lupi
- Laboratory of Antitumor Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy;
| | - Mirko Marabese
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (E.C.); (M.C.); (M.B.)
- Correspondence: (M.M.); (L.B.); Tel.: +39-0239014236 (M.M.); +39-0239014742 (L.B.)
| | - Roberta Pastorelli
- Laboratory of Mass Spectrometry, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (G.S.); (R.P.)
| | - Massimo Broggini
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (E.C.); (M.C.); (M.B.)
| | - Laura Brunelli
- Laboratory of Mass Spectrometry, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (G.S.); (R.P.)
- Correspondence: (M.M.); (L.B.); Tel.: +39-0239014236 (M.M.); +39-0239014742 (L.B.)
| |
Collapse
|
38
|
Nikolaou S, Machesky LM. The stressful tumour environment drives plasticity of cell migration programmes, contributing to metastasis. J Pathol 2020; 250:612-623. [PMID: 32057095 PMCID: PMC7216910 DOI: 10.1002/path.5395] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/21/2020] [Accepted: 02/10/2020] [Indexed: 12/19/2022]
Abstract
Tumours evolve to cope with environmental stresses or challenges such as nutrient starvation, depletion of survival factors, and unbalanced mechanical forces. The uncontrolled growth and aberrant deregulation of core cell homeostatic pathways induced by genetic mutations create an environment of stress. Here, we explore how the adaptations of tumours to the changing environment can drive changes in the motility machinery of cells, affecting migration, invasion, and metastasis. Tumour cells can invade individually or collectively, or they can be extruded out of the surrounding epithelium. These mechanisms are thought to be modifications of normal processes occurring during development or tissue repair. Therefore, tumours may activate these pathways in response to environmental stresses, enabling them to survive in hostile environments and spread to distant sites. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Savvas Nikolaou
- Division of Cancer Metastasis and RecurrenceCRUK Beatson InstituteGlasgowUK
| | - Laura M Machesky
- Division of Cancer Metastasis and RecurrenceCRUK Beatson InstituteGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
| |
Collapse
|
39
|
Macropinocytosis confers resistance to therapies targeting cancer anabolism. Nat Commun 2020; 11:1121. [PMID: 32111826 PMCID: PMC7048872 DOI: 10.1038/s41467-020-14928-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 02/06/2020] [Indexed: 01/07/2023] Open
Abstract
Macropinocytic cancer cells scavenge amino acids from extracellular proteins. Here, we show that consuming necrotic cell debris via macropinocytosis (necrocytosis) offers additional anabolic benefits. A click chemistry-based flux assay reveals that necrocytosis provides not only amino acids, but sugars, fatty acids and nucleotides for biosynthesis, conferring resistance to therapies targeting anabolic pathways. Indeed, necrotic cell debris allow macropinocytic breast and prostate cancer cells to proliferate, despite fatty acid synthase inhibition. Standard therapies such as gemcitabine, 5-fluorouracil (5-FU), doxorubicin and gamma-irradiation directly or indirectly target nucleotide biosynthesis, creating stress that is relieved by scavenged nucleotides. Strikingly, necrotic debris also render macropinocytic, but not non-macropinocytic, pancreas and breast cancer cells resistant to these treatments. Selective, genetic inhibition of macropinocytosis confirms that necrocytosis both supports tumor growth and limits the effectiveness of 5-FU in vivo. Therefore, this study establishes necrocytosis as a mechanism for drug resistance. Macropinocytosis allows cancer cells to cope with nutrient stress. Here, the authors use a selective, genetic approach to inhibit macropinocytosis and show that consuming necrotic cell debris via macropinocytosis—necrocytosis—affords resistance to many therapies that target biosynthesis.
Collapse
|
40
|
Cooke M, Baker MJ, Kazanietz MG. Rac-GEF/Rac Signaling and Metastatic Dissemination in Lung Cancer. Front Cell Dev Biol 2020; 8:118. [PMID: 32158759 PMCID: PMC7051914 DOI: 10.3389/fcell.2020.00118] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 02/11/2020] [Indexed: 12/17/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, with non-small cell lung cancer (NSCLC) representing ∼85% of new diagnoses. The disease is often detected in an advanced metastatic stage, with poor prognosis and clinical outcome. In order to escape from the primary tumor, cancer cells acquire highly motile and invasive phenotypes that involve the dynamic reorganization of the actin cytoskeleton. These processes are tightly regulated by Rac1, a small G-protein that participates in the formation of actin-rich membrane protrusions required for cancer cell motility and for the secretion of extracellular matrix (ECM)-degrading proteases. In this perspective article we focus on the mechanisms leading to aberrant Rac1 signaling in NSCLC progression and metastasis, highlighting the role of Rac Guanine nucleotide Exchange Factors (GEFs). A plausible scenario is that specific Rac-GEFs activate discrete intracellular pools of Rac1, leading to unique functional responses in the context of specific oncogenic drivers, such as mutant EGFR or mutant KRAS. The identification of dysregulated Rac signaling regulators may serve to predict critical biomarkers for metastatic disease in lung cancer patients, ultimately aiding in refining patient prognosis and decision-making in the clinical setting.
Collapse
Affiliation(s)
- Mariana Cooke
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Marcelo G. Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
41
|
Metabolic Reprogramming and Vulnerabilities in Cancer. Cancers (Basel) 2019; 12:cancers12010090. [PMID: 31905922 PMCID: PMC7016671 DOI: 10.3390/cancers12010090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 12/17/2022] Open
Abstract
Metabolic programs are rewired in tumors to support growth, progression, and immune evasion. A wealth of work in the past decade has delineated how these metabolic rearrangements are facilitated by signaling pathways downstream of oncogene activation and tumor suppressor loss. More recently, this field has expanded to include metabolic interactions among the diverse cell types that exist within a tumor and how this impacts the immune system. In this special issue, 17 review articles discuss these phenomena, and, alongside four original research manuscripts, the vulnerabilities associated with deregulated metabolic programming are highlighted and examined.
Collapse
|
42
|
Salloum G, Jakubik CT, Erami Z, Heitz SD, Bresnick AR, Backer JM. PI3Kβ is selectively required for growth factor-stimulated macropinocytosis. J Cell Sci 2019; 132:jcs.231639. [PMID: 31409694 DOI: 10.1242/jcs.231639] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/16/2019] [Indexed: 12/16/2022] Open
Abstract
Macropinocytosis is an actin-dependent but clathrin-independent endocytic process by which cells nonselectively take up large aliquots of extracellular material. Macropinocytosis is used for immune surveillance by dendritic cells, as a route of infection by viruses and protozoa, and as a nutrient uptake pathway in tumor cells. In this study, we explore the role of class I phosphoinositide 3-kinases (PI3Ks) during ligand-stimulated macropinocytosis. We find that macropinocytosis in response to receptor tyrosine kinase activation is strikingly dependent on a single class I PI3K isoform, namely PI3Kβ (containing the p110β catalytic subunit encoded by PIK3CB). Loss of PI3Kβ expression or activity blocks macropinocytosis at early steps, before the formation of circular dorsal ruffles, but also plays a role in later steps, downstream from Rac1 activation. PI3Kβ is also required for the elevated levels of constitutive macropinocytosis found in tumor cells that are defective for the PTEN tumor suppressor. Our data shed new light on PI3K signaling during macropinocytosis, and suggest new therapeutic uses for pharmacological inhibitors of PI3Kβ.
Collapse
Affiliation(s)
- Gilbert Salloum
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Charles T Jakubik
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Zahra Erami
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Samantha D Heitz
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Anne R Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jonathan M Backer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA .,Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
43
|
Gluconeogenesis in cancer cells - Repurposing of a starvation-induced metabolic pathway? Biochim Biophys Acta Rev Cancer 2019; 1872:24-36. [PMID: 31152822 DOI: 10.1016/j.bbcan.2019.05.006] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/15/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022]
Abstract
Cancer cells constantly face a fluctuating nutrient supply and interference with adaptive responses might be an effective therapeutic approach. It has been discovered that in the absence of glucose, cancer cells can synthesize crucial metabolites by expressing phosphoenolpyruvate carboxykinase (PEPCK, PCK1 or PCK2) using abbreviated forms of gluconeogenesis. Gluconeogenesis, which in essence is the reverse pathway of glycolysis, uses lactate or amino acids to feed biosynthetic pathways branching from glycolysis. PCK1 and PCK2 have been shown to be critical for the growth of certain cancers. In contrast, fructose-1,6-bisphosphatase 1 (FBP1), a downstream gluconeogenesis enzyme, inhibits glycolysis and tumor growth, partly by non-enzymatic mechanisms. This review sheds light on the current knowledge of cancer cell gluconeogenesis and its role in metabolic reprogramming, cancer cell plasticity, and tumor growth.
Collapse
|
44
|
RAC1 Takes the Lead in Solid Tumors. Cells 2019; 8:cells8050382. [PMID: 31027363 PMCID: PMC6562738 DOI: 10.3390/cells8050382] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/18/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022] Open
Abstract
Three GTPases, RAC, RHO, and Cdc42, play essential roles in coordinating many cellular functions during embryonic development, both in healthy cells and in disease conditions like cancers. We have presented patterns of distribution of the frequency of RAC1-alteration(s) in cancers as obtained from cBioPortal. With this background data, we have interrogated the various functions of RAC1 in tumors, including proliferation, metastasis-associated phenotypes, and drug-resistance with a special emphasis on solid tumors in adults. We have reviewed the activation and regulation of RAC1 functions on the basis of its sub-cellular localization in tumor cells. Our review focuses on the role of RAC1 in cancers and summarizes the regulatory mechanisms, inhibitory efficacy, and the anticancer potential of RAC1-PAK targeting agents.
Collapse
|