201
|
Villa E, Ali ES, Sahu U, Ben-Sahra I. Cancer Cells Tune the Signaling Pathways to Empower de Novo Synthesis of Nucleotides. Cancers (Basel) 2019; 11:E688. [PMID: 31108873 PMCID: PMC6562601 DOI: 10.3390/cancers11050688] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer cells exhibit a dynamic metabolic landscape and require a sufficient supply of nucleotides and other macromolecules to grow and proliferate. To meet the metabolic requirements for cell growth, cancer cells must stimulate de novo nucleotide synthesis to obtain adequate nucleotide pools to support nucleic acid and protein synthesis along with energy preservation, signaling activity, glycosylation mechanisms, and cytoskeletal function. Both oncogenes and tumor suppressors have recently been identified as key molecular determinants for de novo nucleotide synthesis that contribute to the maintenance of homeostasis and the proliferation of cancer cells. Inactivation of tumor suppressors such as TP53 and LKB1 and hyperactivation of the mTOR pathway and of oncogenes such as MYC, RAS, and AKT have been shown to fuel nucleotide synthesis in tumor cells. The molecular mechanisms by which these signaling hubs influence metabolism, especially the metabolic pathways for nucleotide synthesis, continue to emerge. Here, we focus on the current understanding of the molecular mechanisms by which oncogenes and tumor suppressors modulate nucleotide synthesis in cancer cells and, based on these insights, discuss potential strategies to target cancer cell proliferation.
Collapse
Affiliation(s)
- Elodie Villa
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL 60611, USA; (E.V.); (E.S.A.); (U.S.)
| | - Eunus S. Ali
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL 60611, USA; (E.V.); (E.S.A.); (U.S.)
| | - Umakant Sahu
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL 60611, USA; (E.V.); (E.S.A.); (U.S.)
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL 60611, USA; (E.V.); (E.S.A.); (U.S.)
- Robert H. Lurie Cancer Center, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
202
|
Li M, Lu Y, Li Y, Tong L, Gu XC, Meng J, Zhu Y, Wu L, Feng M, Tian N, Zhang P, Xu T, Lin SH, Tong X. Transketolase Deficiency Protects the Liver from DNA Damage by Increasing Levels of Ribose 5-Phosphate and Nucleotides. Cancer Res 2019; 79:3689-3701. [PMID: 31101762 DOI: 10.1158/0008-5472.can-18-3776] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/27/2019] [Accepted: 05/09/2019] [Indexed: 11/16/2022]
Abstract
De novo nucleotide biosynthesis is essential for maintaining cellular nucleotide pools, the suppression of which leads to genome instability. The metabolic enzyme transketolase (TKT) in the nonoxidative branch of the pentose phosphate pathway (PPP) regulates ribose 5-phosphate (R5P) levels and de novo nucleotide biosynthesis. TKT is required for maintaining cell proliferation in human liver cancer cell lines, yet the role of TKT in liver injury and cancer initiation remains to be elucidated. In this study, we generated a liver-specific TKT knockout mouse strain by crossing TKTflox/flox mice with albumin-Cre mice. Loss of TKT in hepatocytes protected the liver from diethylnitrosamine (DEN)-induced DNA damage without altering DEN metabolism. DEN treatment of TKT-null liver increased levels of R5P and promoted de novo nucleotide synthesis. More importantly, supplementation of dNTPs in primary hepatocytes alleviated DEN-induced DNA damage, cell death, inflammatory response, and cell proliferation. Furthermore, DEN and high-fat diet (HFD)-induced liver carcinogenesis was reduced in TKTflox/floxAlb-Cre mice compared with control littermates. Mechanistically, loss of TKT in the liver increased apoptosis, reduced cell proliferation, decreased TNFα, IL6, and STAT3 levels, and alleviated DEN/HFD-induced hepatic steatosis and fibrosis. Together, our data identify a key role for TKT in promoting genome instability during liver injury and tumor initiation. SIGNIFICANCE: These findings identify transketolase as a novel metabolic target to maintain genome stability and reduce liver carcinogenesis.
Collapse
Affiliation(s)
- Minle Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ying Lu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yakui Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingfeng Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Chuan Gu
- Department of Orthopedics, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jian Meng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yemin Zhu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lifang Wu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Feng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Na Tian
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianle Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu-Hai Lin
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xuemei Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
203
|
The Diverse Functions of Non-Essential Amino Acids in Cancer. Cancers (Basel) 2019; 11:cancers11050675. [PMID: 31096630 PMCID: PMC6562791 DOI: 10.3390/cancers11050675] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/07/2019] [Accepted: 05/10/2019] [Indexed: 01/31/2023] Open
Abstract
Far beyond simply being 11 of the 20 amino acids needed for protein synthesis, non-essential amino acids play numerous important roles in tumor metabolism. These diverse functions include providing precursors for the biosynthesis of macromolecules, controlling redox status and antioxidant systems, and serving as substrates for post-translational and epigenetic modifications. This functional diversity has sparked great interest in targeting non-essential amino acid metabolism for cancer therapy and has motivated the development of several therapies that are either already used in the clinic or are currently in clinical trials. In this review, we will discuss the important roles that each of the 11 non-essential amino acids play in cancer, how their metabolic pathways are linked, and how researchers are working to overcome the unique challenges of targeting non-essential amino acid metabolism for cancer therapy.
Collapse
|
204
|
Abstract
Altered cell metabolism is ubiquitous in cancer cells; however, it remains challenging to exploit these alterations for cancer therapy. A new study reveals that metabolic alterations to the urea cycle promote tumor growth but unexpectedly also trigger mutations that mark cancer cells for recognition by immunotherapy.
Collapse
Affiliation(s)
- Lucas B Sullivan
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| |
Collapse
|
205
|
Wang K, Sun Y, Guo C, Liu T, Fei X, Chang C. Androgen receptor regulates ASS1P3/miR-34a-5p/ASS1 signaling to promote renal cell carcinoma cell growth. Cell Death Dis 2019; 10:339. [PMID: 31000693 PMCID: PMC6472417 DOI: 10.1038/s41419-019-1330-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 12/20/2018] [Accepted: 01/04/2019] [Indexed: 02/06/2023]
Abstract
Recent studies have demonstrated that the androgen receptor (AR) could play important roles to promote renal cell carcinoma (RCC) cell proliferation, and other studies have also indicated that suppressing the argininosuccinate synthase 1 (ASS1) could promote proliferation of various tumors. The potential of AR promoting cell proliferation in RCC via altering ASS1, however, remains unclear. Here we found that the expression of ASS1 was lower in RCC tissues than in adjacent normal renal tissues, and a lower ASS1 expression was linked to a worse prognosis in RCC patients. Mechanism dissection showed that AR could decrease ASS1 expression to promote RCC cell proliferation via ASS1P3, a pseudogene of ASS1. The results of RIP assay and AGO2 assay revealed that AR could bind ASS1P3 to increase RCC cell proliferation via altering miR-34a-5p function, which could bind to the 3'UTR of ASS1 to suppress its protein expression. ASS1P3 could function as a miRNA decoy for miR-34a-5p to regulate ASS1 in RCC. Preclinical study also supports the in vitro data. Together, these results demonstrated that ASS1P3 could function as a competing endogenous RNA to suppress RCC cell progression, and targeting this newly identified AR-mediated ASS1P3/miR-34a-5p/ASS1 signaling might help in blocking proliferation.
Collapse
Affiliation(s)
- Kefeng Wang
- Department of Urology, Shengjing Hospital, China Medical University, 110004, Shenyang, China.,George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Yin Sun
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Changcheng Guo
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Tao Liu
- Department of Urology, Shengjing Hospital, China Medical University, 110004, Shenyang, China
| | - Xiang Fei
- Department of Urology, Shengjing Hospital, China Medical University, 110004, Shenyang, China.
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA. .,Sex Hormone Research Center, China Medical University/Hospital, 404, Taichung, Taiwan.
| |
Collapse
|
206
|
Sullivan MR, Danai LV, Lewis CA, Chan SH, Gui DY, Kunchok T, Dennstedt EA, Vander Heiden MG, Muir A. Quantification of microenvironmental metabolites in murine cancers reveals determinants of tumor nutrient availability. eLife 2019; 8:44235. [PMID: 30990168 PMCID: PMC6510537 DOI: 10.7554/elife.44235] [Citation(s) in RCA: 388] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/04/2019] [Indexed: 02/06/2023] Open
Abstract
Cancer cell metabolism is heavily influenced by microenvironmental factors, including nutrient availability. Therefore, knowledge of microenvironmental nutrient levels is essential to understand tumor metabolism. To measure the extracellular nutrient levels available to tumors, we utilized quantitative metabolomics methods to measure the absolute concentrations of >118 metabolites in plasma and tumor interstitial fluid, the extracellular fluid that perfuses tumors. Comparison of nutrient levels in tumor interstitial fluid and plasma revealed that the nutrients available to tumors differ from those present in circulation. Further, by comparing interstitial fluid nutrient levels between autochthonous and transplant models of murine pancreatic and lung adenocarcinoma, we found that tumor type, anatomical location and animal diet affect local nutrient availability. These data provide a comprehensive characterization of the nutrients present in the tumor microenvironment of widely used models of lung and pancreatic cancer and identify factors that influence metabolite levels in tumors. In the body, cancer cells can rely on different nutrients than normal cells, and they can use these nutrients in a different way. What cancer cells consume also depends on what is available in their immediate environment. In a tumor, cells grab nutrients from the ‘interstitial’ fluid that surrounds them, but what is present in this liquid may vary within tumors arising in different locations. Understanding what nutrients are ‘on the menu’ in specific tumors would help to target diseased cells while sparing healthy ones, but this knowledge has been difficult to obtain. To investigate this, Sullivan et al. used a technique called mass spectrometry to measure the amounts of 120 nutrients present in the interstitial fluid of mouse pancreas and lung tumors. Different levels of nutrients were found in the two types of tumors, and analyses showed that what was present in the interstitial fluid depended on the type of cancer cells, where the tumor was located, and what the animals ate. This suggests that cancer cells may have different needs because they are limited in what they have access to. It remains to be seen whether the nutrients levels found in mouse tumors are the same as those in humans. Armed with this knowledge, it may then be possible to feed cancer cells grown in the laboratory with the nutrient menu that they would have access to in the body. This could help identify new cancer treatments.
Collapse
Affiliation(s)
- Mark R Sullivan
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Laura V Danai
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, United States
| | - Caroline A Lewis
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, United States
| | - Sze Ham Chan
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, United States
| | - Dan Y Gui
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Tenzin Kunchok
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, United States
| | - Emily A Dennstedt
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Dana-Farber Cancer Institute, Boston, United States
| | - Alexander Muir
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Ben May Department for Cancer Research, University of Chicago, Chicago, United States
| |
Collapse
|
207
|
Lu A, Disoma C, Zhou Y, Chen Z, Zhang L, Shen Y, Zhou M, Du A, Zheng R, Li S, Alsaadawe M, Li S, Li J, Wang W, Jiang T, Peng J, Xia Z. Protein interactome of the deamidase phosphoribosylformylglycinamidine synthetase (PFAS) by LC-MS/MS. Biochem Biophys Res Commun 2019; 513:746-752. [PMID: 30987822 DOI: 10.1016/j.bbrc.2019.04.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 04/04/2019] [Indexed: 12/27/2022]
Abstract
Phosphoribosylformylglycinamidine synthase (PFAS) is an essential enzyme in de novo synthesis of purine. Previously, PFAS has been reported to modulate RIG-I activation during viral infection via deamidation. In this study, we sought to identify potential substrates that PFAS can deamidate. Flag-PFAS was transfected into HEK-293T cells and PFAS associated proteins were purified with anti-Flag M2 magnetic beads. PFAS associated proteins were identified using mass spectrometry and were analyzed using bioinformatics tools including KEGG pathway analysis, gene ontology annotation, and protein interaction network analysis. A total of 441 proteins is suggested to potentially interact with PFAS. Of this number, 12 were previously identified and 429 are newly identified. The interactions of PFAS with CAD, CCT2, PRDX1, and PHGDH were confirmed by co-immunoprecipitation and western blotting. This study is first to report the interaction of PFAS with several proteins which play physiological roles in tumor development including CAD, CCT2, PRDX1, and PHGDH. Furthermore, we show here that PFAS is able to deamidate PHGDH, and induce other posttranslational modification into CAD, CCT2 and PRDX1. The present data provide insight on the biological function of PFAS. Further study to explore the role of these protein interactions in tumorigenesis and other diseases is recommended.
Collapse
Affiliation(s)
- Ai Lu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Cyrollah Disoma
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yuzheng Zhou
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Zongpeng Chen
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Liming Zhang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yilun Shen
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Mei Zhou
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Ashuai Du
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Rong Zheng
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Sijia Li
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Moyed Alsaadawe
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Shiqin Li
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Jiada Li
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Weilan Wang
- School of Life Sciences and Technology, Xinjiang University, Urumqi, Xinjiang, China
| | - Taijiao Jiang
- Center of System Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Suzhou Institute of Systems Medicine, Suzhou, China
| | - Jian Peng
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zanxian Xia
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China.
| |
Collapse
|
208
|
Metabolic reaction network-based recursive metabolite annotation for untargeted metabolomics. Nat Commun 2019; 10:1516. [PMID: 30944337 PMCID: PMC6447530 DOI: 10.1038/s41467-019-09550-x] [Citation(s) in RCA: 229] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 03/13/2019] [Indexed: 12/31/2022] Open
Abstract
Large-scale metabolite annotation is a challenge in liquid chromatogram-mass spectrometry (LC-MS)-based untargeted metabolomics. Here, we develop a metabolic reaction network (MRN)-based recursive algorithm (MetDNA) that expands metabolite annotations without the need for a comprehensive standard spectral library. MetDNA is based on the rationale that seed metabolites and their reaction-paired neighbors tend to share structural similarities resulting in similar MS2 spectra. MetDNA characterizes initial seed metabolites using a small library of MS2 spectra, and utilizes their experimental MS2 spectra as surrogate spectra to annotate their reaction-paired neighbor metabolites, which subsequently serve as the basis for recursive analysis. Using different LC-MS platforms, data acquisition methods, and biological samples, we showcase the utility and versatility of MetDNA and demonstrate that about 2000 metabolites can cumulatively be annotated from one experiment. Our results demonstrate that MetDNA substantially expands metabolite annotation, enabling quantitative assessment of metabolic pathways and facilitating integrative multi-omics analysis.
Collapse
|
209
|
Watch What You (Self-) Eat: Autophagic Mechanisms that Modulate Metabolism. Cell Metab 2019; 29:803-826. [PMID: 30943392 PMCID: PMC6450419 DOI: 10.1016/j.cmet.2019.03.003] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/07/2019] [Accepted: 03/04/2019] [Indexed: 02/07/2023]
Abstract
Autophagy is an evolutionarily conserved lysosome- or vacuole-dependent catabolic pathway in eukaryotes. Autophagy functions basally for cellular quality control and is induced to act as an alternative source of basic metabolites during nutrient deprivation. These functions of autophagy are intimately connected to the regulation of metabolism, and the metabolic status of the cell in turn controls the nature and extent of autophagic induction. Here, we highlight the co-regulation of autophagy and metabolism with a special focus on selective autophagy that, along with bulk autophagy, plays a central role in regulating and rewiring metabolic circuits. We outline the metabolic signals that activate these pathways, the mechanisms involved, and the downstream effects and implications while recognizing yet unanswered questions. We also discuss the role of autophagy in the development and maintenance of adipose tissue, an emerging player in systemic metabolic homeostasis, and describe what is currently known about the complex relationship between autophagy and cancer.
Collapse
|
210
|
Darnell AM, Subramaniam AR, O'Shea EK. Translational Control through Differential Ribosome Pausing during Amino Acid Limitation in Mammalian Cells. Mol Cell 2019; 71:229-243.e11. [PMID: 30029003 DOI: 10.1016/j.molcel.2018.06.041] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/07/2018] [Accepted: 06/26/2018] [Indexed: 12/22/2022]
Abstract
Limitation for amino acids is thought to regulate translation in mammalian cells primarily by signaling through the kinases mTORC1 and GCN2. We find that a selective loss of arginine tRNA charging during limitation for arginine regulates translation through ribosome pausing at two of six arginine codons. Surprisingly, limitation for leucine, an essential and abundant amino acid in protein, results in little or no ribosome pausing. Chemical and genetic perturbation of mTORC1 and GCN2 signaling revealed that their robust response to leucine limitation prevents ribosome pausing, while an insufficient response to arginine limitation leads to loss of tRNA charging and ribosome pausing. Ribosome pausing decreases protein production and triggers premature ribosome termination without reducing mRNA levels. Together, our results suggest that amino acids that are not optimally sensed by the mTORC1 and GCN2 pathways still regulate translation through an evolutionarily conserved mechanism based on codon-specific ribosome pausing.
Collapse
Affiliation(s)
- Alicia M Darnell
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Arvind R Subramaniam
- Basic Sciences Division and Computational Biology Program of Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Erin K O'Shea
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA; Department of Chemistry and Chemical Biology and Faculty of Arts and Sciences Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
211
|
Li L, Mao Y, Zhao L, Li L, Wu J, Zhao M, Du W, Yu L, Jiang P. p53 regulation of ammonia metabolism through urea cycle controls polyamine biosynthesis. Nature 2019; 567:253-256. [PMID: 30842655 DOI: 10.1038/s41586-019-0996-7] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 02/05/2019] [Indexed: 12/20/2022]
Abstract
Cancer cells exhibit altered and usually increased metabolic processes to meet their high biogenetic demands1,2. Under these conditions, ammonia is concomitantly produced by the increased metabolic processing. However, it is unclear how tumour cells dispose of excess ammonia and what outcomes might be caused by the accumulation of ammonia. Here we report that the tumour suppressor p53, the most frequently mutated gene in human tumours, regulates ammonia metabolism by repressing the urea cycle. Through transcriptional downregulation of CPS1, OTC and ARG1, p53 suppresses ureagenesis and elimination of ammonia in vitro and in vivo, leading to the inhibition of tumour growth. Conversely, downregulation of these genes reciprocally activates p53 by MDM2-mediated mechanism(s). Furthermore, the accumulation of ammonia causes a significant decline in mRNA translation of the polyamine biosynthetic rate-limiting enzyme ODC, thereby inhibiting the biosynthesis of polyamine and cell proliferation. Together, these findings link p53 to ureagenesis and ammonia metabolism, and further reveal a role for ammonia in controlling polyamine biosynthesis and cell proliferation.
Collapse
Affiliation(s)
- Le Li
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Youxiang Mao
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Lina Zhao
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Lijia Li
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Jinjun Wu
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Mengjia Zhao
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Wenjing Du
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Li Yu
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Peng Jiang
- School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
212
|
Pham-Danis C, Gehrke S, Danis E, Rozhok AI, Daniels MW, Gao D, Collins C, Paola JTD, D'Alessandro A, DeGregori J. Urea Cycle Sustains Cellular Energetics upon EGFR Inhibition in EGFR-Mutant NSCLC. Mol Cancer Res 2019; 17:1351-1364. [PMID: 30808730 DOI: 10.1158/1541-7786.mcr-18-1068] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 02/06/2019] [Accepted: 02/22/2019] [Indexed: 12/21/2022]
Abstract
Mutations in oncogenes and tumor suppressor genes engender unique metabolic phenotypes crucial to the survival of tumor cells. EGFR signaling has been linked to the rewiring of tumor metabolism in non-small cell lung cancer (NSCLC). We have integrated the use of a functional genomics screen and metabolomics to identify metabolic vulnerabilities induced by EGFR inhibition. These studies reveal that following EGFR inhibition, EGFR-driven NSCLC cells become dependent on the urea cycle and, in particular, the urea cycle enzyme CPS1. Combining knockdown of CPS1 with EGFR inhibition further reduces cell proliferation and impedes cell-cycle progression. Profiling of the metabolome demonstrates that suppression of CPS1 potentiates the effects of EGFR inhibition on central carbon metabolism, pyrimidine biosynthesis, and arginine metabolism, coinciding with reduced glycolysis and mitochondrial respiration. We show that EGFR inhibition and CPS1 knockdown lead to a decrease in arginine levels and pyrimidine derivatives, and the addition of exogenous pyrimidines partially rescues the impairment in cell growth. Finally, we show that high expression of CPS1 in lung adenocarcinomas correlated with worse patient prognosis in publicly available databases. These data collectively reveal that NSCLC cells have a greater dependency on the urea cycle to sustain central carbon metabolism, pyrimidine biosynthesis, and arginine metabolism to meet cellular energetics upon inhibition of EGFR. IMPLICATIONS: Our results reveal that the urea cycle may be a novel metabolic vulnerability in the context of EGFR inhibition, providing an opportunity to develop rational combination therapies with EGFR inhibitors for the treatment of EGFR-driven NSCLC.
Collapse
Affiliation(s)
- Catherine Pham-Danis
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sarah Gehrke
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Etienne Danis
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Andrii I Rozhok
- Department of Dermatology, Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michael W Daniels
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Dexiang Gao
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Christina Collins
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - José T Di Paola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado. .,Department of Dermatology, Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
213
|
Hall PE, Lewis R, Syed N, Shaffer R, Evanson J, Ellis S, Williams M, Feng X, Johnston A, Thomson JA, Harris FP, Jena R, Matys T, Jefferies S, Smith K, Wu BW, Bomalaski JS, Crook T, O'Neill K, Paraskevopoulos D, Khadeir RS, Sheaff M, Pacey S, Plowman PN, Szlosarek PW. A Phase I Study of Pegylated Arginine Deiminase (Pegargiminase), Cisplatin, and Pemetrexed in Argininosuccinate Synthetase 1-Deficient Recurrent High-grade Glioma. Clin Cancer Res 2019; 25:2708-2716. [PMID: 30796035 DOI: 10.1158/1078-0432.ccr-18-3729] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/30/2018] [Accepted: 02/12/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Patients with recurrent high-grade gliomas (HGG) are usually managed with alkylating chemotherapy ± bevacizumab. However, prognosis remains very poor. Preclinically, we showed that HGGs are a target for arginine depletion with pegargiminase (ADI-PEG20) due to epimutations of argininosuccinate synthetase (ASS1) and/or argininosuccinate lyase (ASL). Moreover, ADI-PEG20 disrupts pyrimidine pools in ASS1-deficient HGGs, thereby impacting sensitivity to the antifolate, pemetrexed. PATIENTS AND METHODS We expanded a phase I trial of ADI-PEG20 with pemetrexed and cisplatin (ADIPEMCIS) to patients with ASS1-deficient recurrent HGGs (NCT02029690). Patients were enrolled (01/16-06/17) to receive weekly ADI-PEG20 36 mg/m2 intramuscularly plus pemetrexed 500 mg/m2 and cisplatin 75 mg/m2 intravenously once every 3 weeks for up to 6 cycles. Patients with disease control were allowed ADI-PEG20 maintenance. The primary endpoints were safety, tolerability, and preliminary estimates of efficacy. RESULTS Ten ASS1-deficient heavily pretreated patients were treated with ADIPEMCIS therapy. Treatment was well tolerated with the majority of adverse events being Common Terminology Criteria for Adverse Events v4.03 grade 1-2. The best overall response was stable disease in 8 patients (80%). Plasma arginine was suppressed significantly below baseline with a reciprocal increase in citrulline during the sampling period. The anti-ADI-PEG20 antibody titer rose during the first 4 weeks of treatment before reaching a plateau. Median progression-free survival (PFS) was 5.2 months (95% confidence interval (CI), 2.5-20.8) and overall survival was 6.3 months (95% CI, 1.8-9.7). CONCLUSIONS In this recurrent HGG study, ADIPEMCIS was well tolerated and compares favorably to historical controls. Additional trials of ADI-PEG20 in HGG are planned.
Collapse
Affiliation(s)
- Peter E Hall
- Department of Oncology, Barts Health NHS Trust, London, United Kingdom
| | - Rachel Lewis
- Department of Oncology, Barts Health NHS Trust, London, United Kingdom
| | - Nelofer Syed
- Department of Medicine, Imperial College, London, United Kingdom
| | - Richard Shaffer
- St. Luke's Cancer Centre, Royal Surrey County Hospital, Guildford, United Kingdom
| | - Jane Evanson
- Department of Radiology, Barts Health NHS Trust, London, United Kingdom
| | - Stephen Ellis
- Department of Radiology, Barts Health NHS Trust, London, United Kingdom
| | - Matthew Williams
- Department of Oncology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | | | | | | | - Fiona P Harris
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Raj Jena
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Tomasz Matys
- Department of Radiology, University of Cambridge, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Sarah Jefferies
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Kate Smith
- Department of Oncology, Barts Health NHS Trust, London, United Kingdom
| | - Bor-Wen Wu
- Polaris Pharmaceuticals Inc., San Diego, California
| | | | - Timothy Crook
- St. Luke's Cancer Centre, Royal Surrey County Hospital, Guildford, United Kingdom
| | - Kevin O'Neill
- Department of Neurosurgery, Imperial College Healthcare NHS Trust, London, United Kingdom
| | | | - Ramsay S Khadeir
- Centre for Molecular Oncology, Queen Mary University of London, London, United Kingdom
| | - Michael Sheaff
- Department of Pathology, Barts Health NHS Trust, London, United Kingdom
| | - Simon Pacey
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Piers N Plowman
- Department of Oncology, Barts Health NHS Trust, London, United Kingdom
| | - Peter W Szlosarek
- Department of Oncology, Barts Health NHS Trust, London, United Kingdom.
- Centre for Molecular Oncology, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
214
|
Li T, Zhu Y, Cheng F, Lu C, Jung JU, Gao SJ. Oncogenic Kaposi's Sarcoma-Associated Herpesvirus Upregulates Argininosuccinate Synthase 1, a Rate-Limiting Enzyme of the Citrulline-Nitric Oxide Cycle, To Activate the STAT3 Pathway and Promote Growth Transformation. J Virol 2019; 93:JVI.01599-18. [PMID: 30463977 PMCID: PMC6364034 DOI: 10.1128/jvi.01599-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/13/2018] [Indexed: 02/05/2023] Open
Abstract
Cancer cells are required to rewire existing metabolic pathways to support their abnormal proliferation. We have previously shown that, unlike glucose-addicted cancers, Kaposi's sarcoma-associated herpesvirus (KSHV)-transformed cells depend on glutamine rather than glucose for energy production and amino acid and nucleotide syntheses. High-level consumption of glutamine is tightly regulated and often coupled with the citrulline-nitric oxide (NO) cycle. We have found that KSHV infection accelerates nitrogen efflux by upregulating the expression of argininosuccinate synthase 1 (ASS1), a key enzyme in the citrulline-NO cycle. KSHV utilizes multiple microRNAs to upregulate ASS1 expression. Depletion of either ASS1 or inducible nitric oxide synthase (iNOS) in KSHV-transformed cells suppresses growth proliferation, abolishes colony formation in soft agar, and decreases NO generation. Furthermore, by maintaining intracellular NO levels, ASS1 expression facilitates KSHV-mediated activation of the STAT3 pathway, which is critical for virus-induced transformation. These results illustrate a novel mechanism by which an oncogenic virus hijacks a key metabolic pathway to promote growth transformation and reveal a potential novel therapeutic target for KSHV-induced malignancies.IMPORTANCE We have previously shown that Kaposi's sarcoma-associated herpesvirus (KSHV)-transformed cells depend on glutamine rather than glucose for energy production and amino acid and nucleotide syntheses. In this study, we have further examined how the KSHV-reprogramed metabolic pathways are regulated and discovered that KSHV hijacks the citrulline-nitric oxide (NO) cycle to promote growth proliferation and transformation. Multiple KSHV-encoded microRNAs upregulate argininosuccinate synthase 1 (ASS1), a key enzyme in the citrulline-NO cycle. ASS1 is required for KSHV-induced proliferation, colony formation in soft agar, and NO generation of KSHV-transformed cells, which also depends on inducible nitric oxide synthase. By maintaining intracellular NO levels, ASS1 mediates KSHV activation of the STAT3 pathway, which is essential for KSHV-induced abnormal cell proliferation and transformation. These results illustrate a novel mechanism by which an oncogenic virus hijacks a key metabolic pathway to promote growth transformation and reveal a potential novel therapeutic target for KSHV-induced malignancies.
Collapse
Affiliation(s)
- Tingting Li
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ying Zhu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fan Cheng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Chun Lu
- Department of Microbiology, Nanjing Medical University, Nanjing, Jiansu, China
| | - Jae U Jung
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Shou-Jiang Gao
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Microbiology, Nanjing Medical University, Nanjing, Jiansu, China
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
215
|
Abstract
Kidney cancer, or renal cell carcinoma (RCC), is a disease of increasing incidence that commonly is seen in the general practice of nephrology. Despite this state of affairs, this fascinating and highly morbid disease frequently is under-represented, or even absent, from the curriculum of nephrologists in training and generally is underemphasized in national nephrology meetings, both scientific as well as clinical. Although classic concepts in cancer research in general had led to the concept that cancer is a disease resulting from mutations in the control of growth-regulating pathways, reinforced by the discovery of oncogenes, more contemporary research, particularly in kidney cancer, has uncovered changes in metabolic pathways mediated by those same genes that control tumor energetics and biosynthesis. This adaptation of classic biochemical pathways to the tumor's advantage has been labeled metabolic reprogramming. For example, in the case of kidney cancer there exists a near-universal presence of von Hippel-Lindau tumor suppressor (pVHL) inactivation in the most common form, clear cell RCC (ccRCC), leading to activation of hypoxia-relevant and other metabolic pathways. Studies of this and other pathways in clear cell RCC (ccRCC) have been particularly revealing, leading to the concept that ccRCC can itself be considered a metabolic disease. For this reason, the relatively new method of metabolomics has become a useful technique in the study of ccRCC to tease out those pathways that have been reprogrammed by the tumor to its maximum survival advantage. Furthermore, identification of the nodes of such pathways can lead to novel areas for drug intervention in a disease for which such targets are seriously lacking. Further research and dissemination of these concepts, likely using omics techniques, will lead to clinical trials of therapeutics specifically targeted to tumor metabolism, rather than those generally toxic to all proliferating cells. Such novel agents are highly likely to be more effective than existing drugs and to have far fewer adverse effects. This review provides a general overview of the technique of metabolomics and then discusses how it and other omics techniques have been used to further our understanding of the basic biology of kidney cancer as well as to identify new therapeutic approaches.
Collapse
Affiliation(s)
- Robert H Weiss
- Division of Nephrology, University of California, Davis, CA and Medical Service, VA Northern California Health Care System, Sacramento, CA.
| |
Collapse
|
216
|
Du S, Wang Y, Alatrash N, Weatherly CA, Roy D, MacDonnell FM, Armstrong DW. Altered profiles and metabolism of l- and d-amino acids in cultured human breast cancer cells vs. non-tumorigenic human breast epithelial cells. J Pharm Biomed Anal 2019; 164:421-429. [DOI: 10.1016/j.jpba.2018.10.047] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 12/01/2022]
|
217
|
Epigenetic upregulation and functional role of the mitochondrial aspartate/glutamate carrier isoform 1 in hepatocellular carcinoma. Biochim Biophys Acta Mol Basis Dis 2019; 1865:38-47. [PMID: 30321589 DOI: 10.1016/j.bbadis.2018.10.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 10/05/2018] [Accepted: 10/11/2018] [Indexed: 12/12/2022]
|
218
|
Chen Y, Li G, Nielsen J. Genome-Scale Metabolic Modeling from Yeast to Human Cell Models of Complex Diseases: Latest Advances and Challenges. Methods Mol Biol 2019; 2049:329-345. [PMID: 31602620 DOI: 10.1007/978-1-4939-9736-7_19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Genome-scale metabolic models (GEMs) are mathematical models that enable systematic analysis of metabolism. This modeling concept has been applied to study the metabolism of many organisms including the eukaryal model organism, the yeast Saccharomyces cerevisiae, that also serves as an important cell factory for production of fuels and chemicals. With the application of yeast GEMs, our knowledge of metabolism is increasing. Therefore, GEMs have also been used for modeling human cells to study metabolic diseases. Here we introduce the concept of GEMs and provide a protocol for reconstructing GEMs. Besides, we show the historic development of yeast GEMs and their applications. Also, we review human GEMs as well as their uses in the studies of complex diseases.
Collapse
Affiliation(s)
- Yu Chen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, Gothenburg, Sweden
| | - Gang Li
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, Gothenburg, Sweden
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, Gothenburg, Sweden.
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
219
|
Silberman A, Goldman O, Boukobza Assayag O, Jacob A, Rabinovich S, Adler L, Lee JS, Keshet R, Sarver A, Frug J, Stettner N, Galai S, Persi E, Halpern KB, Zaltsman-Amir Y, Pode-Shakked B, Eilam R, Anikster Y, Nagamani SCS, Ulitsky I, Ruppin E, Erez A. Acid-Induced Downregulation of ASS1 Contributes to the Maintenance of Intracellular pH in Cancer. Cancer Res 2018; 79:518-533. [PMID: 30573518 DOI: 10.1158/0008-5472.can-18-1062] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 08/30/2018] [Accepted: 12/17/2018] [Indexed: 12/23/2022]
Abstract
Downregulation of the urea cycle enzyme argininosuccinate synthase (ASS1) by either promoter methylation or by HIF1α is associated with increased metastasis and poor prognosis in multiple cancers. We have previously shown that in normoxic conditions, ASS1 downregulation facilitates cancer cell proliferation by increasing aspartate availability for pyrimidine synthesis by the enzyme complex CAD. Here we report that in hypoxia, ASS1 expression in cancerous cells is downregulated further by HIF1α-mediated induction of miR-224-5p, making the cells more invasive and dependent on upstream substrates of ASS1 for survival. ASS1 was downregulated under acidic conditions, and ASS1-depleted cancer cells maintained a higher intracellular pH (pHi), depended less on extracellular glutamine, and displayed higher glutathione levels. Depletion of substrates of urea cycle enzymes in ASS1-deficient cancers decreased cancer cell survival. Thus, ASS1 levels in cancer are differentially regulated in various environmental conditions to metabolically benefit cancer progression. Understanding these alterations may help uncover specific context-dependent cancer vulnerabilities that may be targeted for therapeutic purposes. SIGNIFICANCE: Cancer cells in an acidic or hypoxic environment downregulate the expression of the urea cycle enzyme ASS1, which provides them with a redox and pH advantage, resulting in better survival.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/3/518/F1.large.jpg.
Collapse
Affiliation(s)
- Alon Silberman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Omer Goldman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | | - Adi Jacob
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Shiran Rabinovich
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Lital Adler
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Joo Sang Lee
- Center for Bioinformatics and Computational Biology and Dept. of Computer Science, University of Maryland, College Park, Maryland.,Cancer Data Science Lab, National Cancer Institute, NIH, Bethesda, Maryland
| | - Rom Keshet
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Alona Sarver
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Julia Frug
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Stettner
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.,Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Sivan Galai
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Erez Persi
- Computational Biology and Bioinformatics Branch (CBB), National Library of Medicine, National Center for Biotechnology Information (NCBI), NIH, Bethesda, Maryland
| | - Keren Bahar Halpern
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Ben Pode-Shakked
- Metabolic Disease Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel.,The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Raya Eilam
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Yair Anikster
- Metabolic Disease Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel.,The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Sandesh C S Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Igor Ulitsky
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Eytan Ruppin
- Center for Bioinformatics and Computational Biology and Dept. of Computer Science, University of Maryland, College Park, Maryland.,Cancer Data Science Lab, National Cancer Institute, NIH, Bethesda, Maryland
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
220
|
Bahat A, Goldman A, Zaltsman Y, Khan DH, Halperin C, Amzallag E, Krupalnik V, Mullokandov M, Silberman A, Erez A, Schimmer AD, Hanna JH, Gross A. MTCH2-mediated mitochondrial fusion drives exit from naïve pluripotency in embryonic stem cells. Nat Commun 2018; 9:5132. [PMID: 30510213 PMCID: PMC6277412 DOI: 10.1038/s41467-018-07519-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 11/07/2018] [Indexed: 01/16/2023] Open
Abstract
The role of mitochondria dynamics and its molecular regulators remains largely unknown during naïve-to-primed pluripotent cell interconversion. Here we report that mitochondrial MTCH2 is a regulator of mitochondrial fusion, essential for the naïve-to-primed interconversion of murine embryonic stem cells (ESCs). During this interconversion, wild-type ESCs elongate their mitochondria and slightly alter their glutamine utilization. In contrast, MTCH2-/- ESCs fail to elongate their mitochondria and to alter their metabolism, maintaining high levels of histone acetylation and expression of naïve pluripotency markers. Importantly, enforced mitochondria elongation by the pro-fusion protein Mitofusin (MFN) 2 or by a dominant negative form of the pro-fission protein dynamin-related protein (DRP) 1 is sufficient to drive the exit from naïve pluripotency of both MTCH2-/- and wild-type ESCs. Taken together, our data indicate that mitochondria elongation, governed by MTCH2, plays a critical role and constitutes an early driving force in the naïve-to-primed pluripotency interconversion of murine ESCs.
Collapse
Affiliation(s)
- Amir Bahat
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Andres Goldman
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Yehudit Zaltsman
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Dilshad H Khan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Coral Halperin
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Emmanuel Amzallag
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Vladislav Krupalnik
- Department of Molecular Genetics, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Michael Mullokandov
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Alon Silberman
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jacob H Hanna
- Department of Molecular Genetics, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Atan Gross
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel.
| |
Collapse
|
221
|
Abstract
Oncogenic signalling and metabolic alterations are interrelated in cancer cells. mTOR, which is frequently activated in cancer, controls cell growth and metabolism. mTOR signalling regulates amino acid, glucose, nucleotide, fatty acid and lipid metabolism. Conversely, metabolic inputs, such as amino acids, activate mTOR. In this Review, we discuss how mTOR signalling rewires cancer cell metabolism and delineate how changes in metabolism, in turn, sustain mTOR signalling and tumorigenicity. Several drugs are being developed to perturb cancer cell metabolism. However, their efficacy as stand-alone therapies, similar to mTOR inhibitors, is limited. Here, we discuss how the interdependence of mTOR signalling and metabolism can be exploited for cancer therapy.
Collapse
Affiliation(s)
| | - Sujin Park
- Biozentrum, University of Basel, Basel, Switzerland
| | | |
Collapse
|
222
|
Poillet-Perez L, Xie X, Zhan L, Yang Y, Sharp DW, Hu ZS, Su X, Maganti A, Jiang C, Lu W, Zheng H, Bosenberg MW, Mehnert JM, Guo JY, Lattime E, Rabinowitz JD, White E. Autophagy maintains tumour growth through circulating arginine. Nature 2018; 563:569-573. [PMID: 30429607 PMCID: PMC6287937 DOI: 10.1038/s41586-018-0697-7] [Citation(s) in RCA: 260] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 09/17/2018] [Indexed: 11/10/2022]
Abstract
Autophagy captures intracellular components and delivers them to lysosomes, where they are degraded and recycled to sustain metabolism and to enable survival during starvation1-5. Acute, whole-body deletion of the essential autophagy gene Atg7 in adult mice causes a systemic metabolic defect that manifests as starvation intolerance and gradual loss of white adipose tissue, liver glycogen and muscle mass1. Cancer cells also benefit from autophagy. Deletion of essential autophagy genes impairs the metabolism, proliferation, survival and malignancy of spontaneous tumours in models of autochthonous cancer6,7. Acute, systemic deletion of Atg7 or acute, systemic expression of a dominant-negative ATG4b in mice induces greater regression of KRAS-driven cancers than does tumour-specific autophagy deletion, which suggests that host autophagy promotes tumour growth1,8. Here we show that host-specific deletion of Atg7 impairs the growth of multiple allografted tumours, although not all tumour lines were sensitive to host autophagy status. Loss of autophagy in the host was associated with a reduction in circulating arginine, and the sensitive tumour cell lines were arginine auxotrophs owing to the lack of expression of the enzyme argininosuccinate synthase 1. Serum proteomic analysis identified the arginine-degrading enzyme arginase I (ARG1) in the circulation of Atg7-deficient hosts, and in vivo arginine metabolic tracing demonstrated that serum arginine was degraded to ornithine. ARG1 is predominantly expressed in the liver and can be released from hepatocytes into the circulation. Liver-specific deletion of Atg7 produced circulating ARG1, and reduced both serum arginine and tumour growth. Deletion of Atg5 in the host similarly regulated [corrected] circulating arginine and suppressed tumorigenesis, which demonstrates that this phenotype is specific to autophagy function rather than to deletion of Atg7. Dietary supplementation of Atg7-deficient hosts with arginine partially restored levels of circulating arginine and tumour growth. Thus, defective autophagy in the host leads to the release of ARG1 from the liver and the degradation of circulating arginine, which is essential for tumour growth; this identifies a metabolic vulnerability of cancer.
Collapse
Affiliation(s)
| | - Xiaoqi Xie
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Le Zhan
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Yang Yang
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Daniel W Sharp
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | | | - Xiaoyang Su
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Anurag Maganti
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Cherry Jiang
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Wenyun Lu
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Haiyan Zheng
- Biological Mass Spectrometry Facility, Rutgers University, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Marcus W Bosenberg
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Janice M Mehnert
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Medicine, Division of Medical Oncology, Developmental Therapeutics Unit, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Jessie Yanxiang Guo
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA.,Department of Chemical Biology, Rutgers Ernest Mario School of Pharmacy, Piscataway, NJ, USA
| | - Edmund Lattime
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Surgery, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Joshua D Rabinowitz
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA. .,Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
223
|
Cheng CT, Qi Y, Wang YC, Chi KK, Chung Y, Ouyang C, Chen YR, Oh ME, Sheng X, Tang Y, Liu YR, Lin HH, Kuo CY, Schones D, Vidal CM, Chu JCY, Wang HJ, Chen YH, Miller KM, Chu P, Yen Y, Jiang L, Kung HJ, Ann DK. Arginine starvation kills tumor cells through aspartate exhaustion and mitochondrial dysfunction. Commun Biol 2018; 1:178. [PMID: 30393775 PMCID: PMC6203837 DOI: 10.1038/s42003-018-0178-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/24/2018] [Indexed: 12/22/2022] Open
Abstract
Defective arginine synthesis, due to the silencing of argininosuccinate synthase 1 (ASS1), is a common metabolic vulnerability in cancer, known as arginine auxotrophy. Understanding how arginine depletion kills arginine-auxotrophic cancer cells will facilitate the development of anti-cancer therapeutic strategies. Here we show that depletion of extracellular arginine in arginine-auxotrophic cancer cells causes mitochondrial distress and transcriptional reprogramming. Mechanistically, arginine starvation induces asparagine synthetase (ASNS), depleting these cancer cells of aspartate, and disrupting their malate-aspartate shuttle. Supplementation of aspartate, depletion of mitochondria, and knockdown of ASNS all protect the arginine-starved cells, establishing the causal effects of aspartate depletion and mitochondrial dysfunction on the arginine starvation-induced cell death. Furthermore, dietary arginine restriction reduced tumor growth in a xenograft model of ASS1-deficient breast cancer. Our data challenge the view that ASNS promotes homeostasis, arguing instead that ASNS-induced aspartate depletion promotes cytotoxicity, which can be exploited for anti-cancer therapies.
Collapse
Affiliation(s)
- Chun-Ting Cheng
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Yue Qi
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Yi-Chang Wang
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Kevin K Chi
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, 91010, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA, 91010, USA
| | - Yiyin Chung
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Ching Ouyang
- Department of Information Sciences, City of Hope, Duarte, CA, 91010, USA
| | - Yun-Ru Chen
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Myung Eun Oh
- Department of Molecular and Cellular Endocrinology, City of Hope, Duarte, CA, 91010, USA
| | - Xiangpeng Sheng
- Department of Molecular and Cellular Endocrinology, City of Hope, Duarte, CA, 91010, USA
| | - Yulong Tang
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Yun-Ru Liu
- Office of Human Research, Center for Cancer Research, Taipei Medical University, Taipei City, Taiwan
| | - H Helen Lin
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Ching-Ying Kuo
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Dustin Schones
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, 91010, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA, 91010, USA
| | - Christina M Vidal
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, 91010, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA, 91010, USA
| | - Jenny C-Y Chu
- Institute for Translational Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Hung-Jung Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Yu-Han Chen
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA, 92697, USA
| | - Kyle M Miller
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Peiguo Chu
- Shared Pathology Core, City of Hope, Duarte, CA, 91010, USA
| | - Yun Yen
- Office of Human Research, Center for Cancer Research, Taipei Medical University, Taipei City, Taiwan
| | - Lei Jiang
- Department of Molecular and Cellular Endocrinology, City of Hope, Duarte, CA, 91010, USA
| | - Hsing-Jien Kung
- Institute for Translational Medicine, Taipei Medical University, Taipei City, Taiwan.
- Department of Biochemistry and Molecular Medicine, University of California at Davis Comprehensive Cancer Center, Sacramento, CA, 95817, USA.
| | - David K Ann
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, 91010, USA.
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
224
|
Trott JF, Hwang VJ, Ishimaru T, Chmiel KJ, Zhou JX, Shim K, Stewart BJ, Mahjoub MR, Jen KY, Barupal DK, Li X, Weiss RH. Arginine reprogramming in ADPKD results in arginine-dependent cystogenesis. Am J Physiol Renal Physiol 2018; 315:F1855-F1868. [PMID: 30280600 DOI: 10.1152/ajprenal.00025.2018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Research into metabolic reprogramming in cancer has become commonplace, yet this area of research has only recently come of age in nephrology. In light of the parallels between cancer and autosomal dominant polycystic kidney disease (ADPKD), the latter is currently being studied as a metabolic disease. In clear cell renal cell carcinoma (RCC), which is now considered a metabolic disease, we and others have shown derangements in the enzyme arginosuccinate synthase 1 (ASS1), resulting in RCC cells becoming auxotrophic for arginine and leading to a new therapeutic paradigm involving reducing extracellular arginine. Based on our earlier finding that glutamine pathways are reprogrammed in ARPKD, and given the connection between arginine and glutamine synthetic pathways via citrulline, we investigated the possibility of arginine reprogramming in ADPKD. We now show that, in a remarkable parallel to RCC, ASS1 expression is reduced in murine and human ADPKD, and arginine depletion results in a dose-dependent compensatory increase in ASS1 levels as well as decreased cystogenesis in vitro and ex vivo with minimal toxicity to normal cells. Nontargeted metabolomics analysis of mouse kidney cell lines grown in arginine-deficient versus arginine-replete media suggests arginine-dependent alterations in the glutamine and proline pathways. Thus, depletion of this conditionally essential amino acid by dietary or pharmacological means, such as with arginine-degrading enzymes, may be a novel treatment for this disease.
Collapse
Affiliation(s)
- Josephine F Trott
- Division of Nephrology, Department of Internal Medicine, University of California , Davis, California
| | - Vicki J Hwang
- Division of Nephrology, Department of Internal Medicine, University of California , Davis, California
| | - Tatsuto Ishimaru
- Division of Nephrology, Department of Internal Medicine, University of California , Davis, California
| | - Kenneth J Chmiel
- Division of Nephrology, Department of Internal Medicine, University of California , Davis, California
| | - Julie X Zhou
- Kidney Institute, Department of Internal Medicine, University of Kansas Medical Center , Kansas City, Kansas
| | - Kyuhwan Shim
- Division of Nephrology, Department of Medicine, Washington University , St. Louis, Missouri
| | | | - Moe R Mahjoub
- Division of Nephrology, Department of Medicine, Washington University , St. Louis, Missouri
| | - Kuang-Yu Jen
- Department of Pathology, University of California , Davis, California
| | - Dinesh K Barupal
- West Coast Metabolomics Center, University of California , Davis, California
| | - Xiaogang Li
- Kidney Institute, Department of Internal Medicine, University of Kansas Medical Center , Kansas City, Kansas
| | - Robert H Weiss
- Division of Nephrology, Department of Internal Medicine, University of California , Davis, California.,Cancer Center, University of California , Davis, California.,Medical Service, VA Northern California Health Care System, Sacramento, California
| |
Collapse
|
225
|
Abstract
Cancer cells reprogramme metabolism to maximize the use of nitrogen and carbon for the anabolic synthesis of macromolecules that are required during tumour proliferation and growth. To achieve this aim, one strategy is to reduce catabolism and nitrogen disposal. The urea cycle (UC) in the liver is the main metabolic pathway to convert excess nitrogen into disposable urea. Outside the liver, UC enzymes are differentially expressed, enabling the use of nitrogen for the synthesis of UC intermediates that are required to accommodate cellular needs. Interestingly, the expression of UC enzymes is altered in cancer, revealing a revolutionary mechanism to maximize nitrogen incorporation into biomass. In this Review, we discuss the metabolic benefits underlying UC deregulation in cancer and the relevance of these alterations for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Rom Keshet
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Peter Szlosarek
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, UK
- Barts Health NHS Trust, St Bartholomew's Hospital, London, UK
| | - Arkaitz Carracedo
- CIC bioGUNE, Bizkaia, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Biochemistry and Molecular Biology Department, University of the Basque Country, Bilbao, Spain
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
226
|
Mishra R, Haldar S, Placencio V, Madhav A, Rohena-Rivera K, Agarwal P, Duong F, Angara B, Tripathi M, Liu Z, Gottlieb RA, Wagner S, Posadas EM, Bhowmick NA. Stromal epigenetic alterations drive metabolic and neuroendocrine prostate cancer reprogramming. J Clin Invest 2018; 128:4472-4484. [PMID: 30047926 PMCID: PMC6159981 DOI: 10.1172/jci99397] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 07/18/2018] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer is an androgen-dependent disease subject to interactions between the tumor epithelium and its microenvironment. Here, we found that epigenetic changes in prostatic cancer-associated fibroblasts (CAF) initiated a cascade of stromal-epithelial interactions. This facilitated lethal prostate cancer growth and development of resistance to androgen signaling deprivation therapy (ADT). We identified a Ras inhibitor, RASAL3, as epigenetically silenced in human prostatic CAF, leading to oncogenic Ras activity driving macropinocytosis-mediated glutamine synthesis. Interestingly, ADT further promoted RASAL3 epigenetic silencing and glutamine secretion by prostatic fibroblasts. In an orthotopic xenograft model, subsequent inhibition of macropinocytosis and glutamine transport resulted in antitumor effects. Stromal glutamine served as a source of energy through anaplerosis and as a mediator of neuroendocrine differentiation for prostate adenocarcinoma. Antagonizing the uptake of glutamine restored sensitivity to ADT in a castration-resistant xenograft model. In validating these findings, we found that prostate cancer patients on ADT with therapeutic resistance had elevated blood glutamine levels compared with those with therapeutically responsive disease (odds ratio = 7.451, P = 0.02). Identification of epigenetic regulation of Ras activity in prostatic CAF revealed RASAL3 as a sensor for metabolic and neuroendocrine reprogramming in prostate cancer patients failing ADT.
Collapse
Affiliation(s)
| | | | | | - Anisha Madhav
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | | | | | | | | | | | | - Roberta A. Gottlieb
- Department of Medicine, and
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shawn Wagner
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | | - Neil A. Bhowmick
- Department of Medicine, and
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Research, Greater Los Angeles Veterans Administration, Los Angeles, California, USA
| |
Collapse
|
227
|
Lee JS, Adler L, Karathia H, Carmel N, Rabinovich S, Auslander N, Keshet R, Stettner N, Silberman A, Agemy L, Helbling D, Eilam R, Sun Q, Brandis A, Malitsky S, Itkin M, Weiss H, Pinto S, Kalaora S, Levy R, Barnea E, Admon A, Dimmock D, Stern-Ginossar N, Scherz A, Nagamani SCS, Unda M, Wilson DM, Elhasid R, Carracedo A, Samuels Y, Hannenhalli S, Ruppin E, Erez A. Urea Cycle Dysregulation Generates Clinically Relevant Genomic and Biochemical Signatures. Cell 2018; 174:1559-1570.e22. [PMID: 30100185 PMCID: PMC6225773 DOI: 10.1016/j.cell.2018.07.019] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/21/2018] [Accepted: 07/12/2018] [Indexed: 01/02/2023]
Abstract
The urea cycle (UC) is the main pathway by which mammals dispose of waste nitrogen. We find that specific alterations in the expression of most UC enzymes occur in many tumors, leading to a general metabolic hallmark termed "UC dysregulation" (UCD). UCD elicits nitrogen diversion toward carbamoyl-phosphate synthetase2, aspartate transcarbamylase, and dihydrooratase (CAD) activation and enhances pyrimidine synthesis, resulting in detectable changes in nitrogen metabolites in both patient tumors and their bio-fluids. The accompanying excess of pyrimidine versus purine nucleotides results in a genomic signature consisting of transversion mutations at the DNA, RNA, and protein levels. This mutational bias is associated with increased numbers of hydrophobic tumor antigens and a better response to immune checkpoint inhibitors independent of mutational load. Taken together, our findings demonstrate that UCD is a common feature of tumors that profoundly affects carcinogenesis, mutagenesis, and immunotherapy response.
Collapse
Affiliation(s)
- Joo Sang Lee
- Cancer Data Science Lab, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Center for Bioinformatics and Computational Biology, University of Maryland Institute for Advanced Computer Studies, Department of Computer Science, University of Maryland, College Park, MD 20742, USA
| | - Lital Adler
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Hiren Karathia
- Center for Bioinformatics and Computational Biology, University of Maryland Institute for Advanced Computer Studies, Department of Computer Science, University of Maryland, College Park, MD 20742, USA
| | - Narin Carmel
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Shiran Rabinovich
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Noam Auslander
- Cancer Data Science Lab, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Center for Bioinformatics and Computational Biology, University of Maryland Institute for Advanced Computer Studies, Department of Computer Science, University of Maryland, College Park, MD 20742, USA
| | - Rom Keshet
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Noa Stettner
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel; Department of Veterinary Resources, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Alon Silberman
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Lilach Agemy
- Department of Plant and Environmental Science, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | | | - Raya Eilam
- Department of Veterinary Resources, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Qin Sun
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander Brandis
- Life Sciences Core Facilities, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Sergey Malitsky
- Life Sciences Core Facilities, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Maxim Itkin
- Life Sciences Core Facilities, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Hila Weiss
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Sivan Pinto
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Shelly Kalaora
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Ronen Levy
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Eilon Barnea
- Faculty of Biology, Technion - Israel Institute of Technology, 3200003 Haifa, Israel
| | - Arie Admon
- Faculty of Biology, Technion - Israel Institute of Technology, 3200003 Haifa, Israel
| | - David Dimmock
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | - Noam Stern-Ginossar
- Department of Molecular Genetics, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Avigdor Scherz
- Department of Veterinary Resources, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Sandesh C S Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital, Houston, TX 77030, USA
| | - Miguel Unda
- Department of Urology, Basurto University Hospital, 48013 Bilbao, Spain; CIBERONC, Madrid, Spain
| | - David M Wilson
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, NIH, 251 Bayview Blvd., Baltimore, MD 21224, USA
| | - Ronit Elhasid
- Sackler Faculty of Medicine, Department of Pediatric Hemato Oncology, Sourasky Medical Center, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Arkaitz Carracedo
- CIBERONC, Madrid, Spain; CIC bioGUNE, Bizkaia Technology Park, 801 Building, 48160 Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain; Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Sridhar Hannenhalli
- Center for Bioinformatics and Computational Biology, University of Maryland Institute for Advanced Computer Studies, Department of Computer Science, University of Maryland, College Park, MD 20742, USA
| | - Eytan Ruppin
- Cancer Data Science Lab, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Center for Bioinformatics and Computational Biology, University of Maryland Institute for Advanced Computer Studies, Department of Computer Science, University of Maryland, College Park, MD 20742, USA; Schools of Medicine and Computer Science, Tel Aviv University, 6997801 Tel Aviv, Israel.
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel.
| |
Collapse
|
228
|
Schulze A, Yuneva M. The big picture: exploring the metabolic cross-talk in cancer. Dis Model Mech 2018; 11:11/8/dmm036673. [PMID: 30154190 PMCID: PMC6124556 DOI: 10.1242/dmm.036673] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Metabolic reprogramming is now well established as one of the hallmarks of cancer. The renewed interest in this topic has spurred a remarkable advance in our understanding of the metabolic alterations in cancer cells and in the tumour microenvironment. Initially, this research focussed on identifying the metabolic processes that provided cancer cells with building blocks for growth or to prevent oxidative damage and death. In addition to providing detailed insight into the mechanisms by which oncogenic signalling pathways modulate metabolic processes, this research also revealed multiple nodes within the metabolic network that can be targeted for the selective elimination of cancer cells. However, recent years have seen a paradigm shift in the field of cancer metabolism; while early studies focussed mainly on the metabolic processes within a cancer cell, recent approaches also consider the impact of metabolic cross-talk between different cell types within the tumour or study cancer within the organismal metabolic context. The Review articles presented in this themed Special Collection of Disease Models & Mechanisms aim to provide an overview of the recent advances in the field. The Collection also contains research articles that describe how metabolic inhibition can improve the efficacy of targeted therapy and introduce a new zebrafish model to study metabolic tumour-host interactions. We also present 'A model for life' interviews: a new interview with Karen Vousden and a previously published one with Lewis Cantley that provide insight into these two leaders' personal scientific journeys that resulted in seminal discoveries in the field of cancer metabolism. In this Editorial, we summarise some of the key insights obtained from studying cancer metabolism. We also describe some of the many exciting developments in the field and discuss its future challenges.
Collapse
Affiliation(s)
- Almut Schulze
- Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074 Würzburg, Germany .,Comprehensive Cancer Center Mainfranken, Josef-Schneider-Str.6, 97080 Würzburg, Germany
| | - Mariia Yuneva
- Oncogenes and Tumour Metabolism Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
229
|
Alexandrou C, Al-Aqbi SS, Higgins JA, Boyle W, Karmokar A, Andreadi C, Luo JL, Moore DA, Viskaduraki M, Blades M, Murray GI, Howells LM, Thomas A, Brown K, Cheng PN, Rufini A. Sensitivity of Colorectal Cancer to Arginine Deprivation Therapy is Shaped by Differential Expression of Urea Cycle Enzymes. Sci Rep 2018; 8:12096. [PMID: 30108309 PMCID: PMC6092409 DOI: 10.1038/s41598-018-30591-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/02/2018] [Indexed: 02/08/2023] Open
Abstract
Tumors deficient in the urea cycle enzymes argininosuccinate synthase-1 (ASS1) and ornithine transcarbamylase (OTC) are unable to synthesize arginine and can be targeted using arginine-deprivation therapy. Here, we show that colorectal cancers (CRCs) display negligible expression of OTC and, in subset of cases, ASS1 proteins. CRC cells fail to grow in arginine-free medium and dietary arginine deprivation slows growth of cancer cells implanted into immunocompromised mice. Moreover, we report that clinically-formulated arginine-degrading enzymes are effective anticancer drugs in CRC. Pegylated arginine deiminase (ADI-PEG20), which degrades arginine to citrulline and ammonia, affects growth of ASS1-negative cells, whereas recombinant human arginase-1 (rhArg1peg5000), which degrades arginine into urea and ornithine, is effective against a broad spectrum of OTC-negative CRC cell lines. This reflects the inability of CRC cells to recycle citrulline and ornithine into the urea cycle. Finally, we show that arginase antagonizes chemotherapeutic drugs oxaliplatin and 5-fluorouracil (5-FU), whereas ADI-PEG20 synergizes with oxaliplatin in ASS1-negative cell lines and appears to interact with 5-fluorouracil independently of ASS1 status. Overall, we conclude that CRC is amenable to arginine-deprivation therapy, but we warrant caution when combining arginine deprivation with standard chemotherapy.
Collapse
Affiliation(s)
- Constantinos Alexandrou
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK
| | - Saif Sattar Al-Aqbi
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK.,Department of Pathology and Poultry Diseases, Faculty of Veterinary Medicine, University of Kufa, Kufa, Iraq
| | - Jennifer A Higgins
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK
| | - William Boyle
- Birmingham Women's Hospital, Birmingham, B15 2TG, UK
| | - Ankur Karmokar
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK
| | - Catherine Andreadi
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK
| | - Jin-Li Luo
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK
| | - David A Moore
- Department of Pathology, UCL Cancer Centre, UCL, London, UK
| | - Maria Viskaduraki
- Bioinformatics and Biostatistics Support Hub, University of Leicester, Leicester, LE1 7RH, UK
| | - Matthew Blades
- Bioinformatics and Biostatistics Support Hub, University of Leicester, Leicester, LE1 7RH, UK
| | - Graeme I Murray
- Department of Pathology, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25, 2ZD, UK
| | - Lynne M Howells
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK
| | - Anne Thomas
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK
| | - Karen Brown
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK
| | - Paul N Cheng
- Bio-Cancer Treatment International Limited, Hong Kong, Hong Kong
| | - Alessandro Rufini
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK.
| |
Collapse
|
230
|
Keshet R, Erez A. Arginine and the metabolic regulation of nitric oxide synthesis in cancer. Dis Model Mech 2018; 11:11/8/dmm033332. [PMID: 30082427 PMCID: PMC6124554 DOI: 10.1242/dmm.033332] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Nitric oxide (NO) is a signaling molecule that plays important roles in diverse biological processes and thus its dysregulation is involved in the pathogenesis of various disorders. In cancer, NO has broad and sometimes dichotomous roles; it is involved in cancer initiation and progression, but also restricts cancer proliferation and invasion, and contributes to the anti-tumor immune response. The importance of NO in a range of cellular processes is exemplified by its tight spatial and dosage control at multiple levels, including via its transcriptional, post-translational and metabolic regulation. In this Review, we focus on the regulation of NO via the synthesis and availability of its precursor, arginine, and discuss the implications of this metabolic regulation for cancer biology and therapy. Despite the established contribution of NO to cancer pathogenesis, the implementation of NO-related cancer therapeutics remains limited, likely due to the challenge of targeting and inducing its protective functions in a cell- and dosage-specific manner. A better understanding of how arginine regulates the production of NO in cancer might thus support the development of anti-cancer drugs that target this key metabolic pathway, and other metabolic pathways involved in NO production.
Collapse
Affiliation(s)
- Rom Keshet
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
231
|
Fujigaki S, Nishiumi S, Kobayashi T, Suzuki M, Iemoto T, Kojima T, Ito Y, Daiko H, Kato K, Shouji H, Honda K, Azuma T, Yoshida M. Identification of serum biomarkers of chemoradiosensitivity in esophageal cancer via the targeted metabolomics approach. Biomark Med 2018; 12:827-840. [PMID: 30043633 DOI: 10.2217/bmm-2017-0449] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIM To identify the serum metabolomics signature that is correlated with the chemoradiosensitivity of esophageal squamous cell carcinoma (ESCC). MATERIALS & METHODS Untargeted and targeted metabolomics analysis of serum samples from 26 ESCC patients, which were collected before the neoadjuvant chemoradiotherapy, was performed. RESULTS On receiving the results of untargeted metabolomics analysis, we performed the targeted metabolomics analysis of the six metabolites (arabitol, betaine, glycine, L-serine, L-arginine and L-aspartate). The serum levels of the four metabolites (arabitol, glycine, L-serine and L-arginine) were significantly lower in the patients who achieved pathological complete response with neoadjuvant chemoradiotherapy compared with the patients who did not achieve pathological complete response (p = 0.0086, 0.0345, 0.0106 and 0.0373, respectively). CONCLUSION The serum levels of metabolites might be useful for predicting the chemoradiosensitivity of ESCC patients.
Collapse
Affiliation(s)
- Seiji Fujigaki
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Shin Nishiumi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Takashi Kobayashi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Makoto Suzuki
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Takao Iemoto
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Takashi Kojima
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Yoshinori Ito
- Department of Radiation Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroyuki Daiko
- Department of Esophageal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Ken Kato
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hirokazu Shouji
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kazufumi Honda
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, Japan
| | - Takeshi Azuma
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Masaru Yoshida
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan.,Division of Metabolomics Research, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, Japan.,AMED-CREST, AMED, Kobe, Japan
| |
Collapse
|
232
|
Givechian KB, Garner C, Garban H, Rabizadeh S, Soon-Shiong P. CAD/POLD2 gene expression is associated with poor overall survival and chemoresistance in bladder urothelial carcinoma. Oncotarget 2018; 9:29743-29752. [PMID: 30038717 PMCID: PMC6049856 DOI: 10.18632/oncotarget.25701] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/13/2018] [Indexed: 12/13/2022] Open
Abstract
Somatic mutations in DNA repair genes have been clinically associated with chemosensitivity, although few studies have interrogated the nucleotide synthesis pathways that supply DNA repair processes. Previous work suggests that bladder urothelial carcinoma is uniquely enriched for mutations in nucleotide excision repair genes, and that these mutations are associated with response to platinum-based therapy and favorable survival. Conversely, the de novo pyrimidine synthesis pathway has recently emerged as a putative clinical target. This anabolic process is thought to supply DNA repair processes such as nucleotide excision repair; that is, DNA repair enzymes may require a sufficient nucleotide supply available to reverse the intended genotoxic damage of systemic chemotherapy in rapidly proliferating cancer cells. Therefore, we explored the prognostic complementarity between de novo pyrimidine synthesis and nucleotide excision repair expression in a total of 570 bladder urothelial carcinoma patients. Ultimately, we show that the de novo pyrimidine synthesis gene CAD is associated with poor survival (P = 0.008) and is co-altered with the nucleotide excision repair gene POLD2. High expression of POLD2 was also associated with poor overall survival (P = 0.019) and was significantly correlated with CAD expression in pre-treatment patient tumor samples (P = 2.44e-4). Expression of each gene was associated with cisplatin-based therapy resistance, and accordingly, CADhighPOLD2high patients were associated with worse survival than CADhighPOLD2low and CADlowPOLD2high patients. Together, these biomarkers could help elucidate mechanisms of chemoresistance to further personalize therapeutic strategies in bladder urothelial carcinoma.
Collapse
Affiliation(s)
| | | | - Hermes Garban
- NantBioscience, Inc. | NantWorks, Culver City, CA 90232, USA
| | - Shahrooz Rabizadeh
- NantOmics LLC, Culver City, CA 90232, USA
- NantBioscience, Inc. | NantWorks, Culver City, CA 90232, USA
| | - Patrick Soon-Shiong
- NantOmics LLC, Culver City, CA 90232, USA
- NantBioscience, Inc. | NantWorks, Culver City, CA 90232, USA
| |
Collapse
|
233
|
A phase 1 study of ADI-PEG 20 and modified FOLFOX6 in patients with advanced hepatocellular carcinoma and other gastrointestinal malignancies. Cancer Chemother Pharmacol 2018; 82:429-440. [PMID: 29971467 DOI: 10.1007/s00280-018-3635-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/27/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE Arginine depletion interferes with pyrimidine metabolism as well as DNA damage repair pathways. Preclinical data indicates that pairing pegylated arginine deiminase (ADI-PEG 20) with fluoropyrimidines or platinum enhances cytotoxicity in vitro and in vivo in arginine auxotrophs. METHODS This is a single-center, open-label, phase 1 trial of ADI-PEG 20 and modified FOLFOX6 (mFOLFOX6) in treatment-refractory hepatocellular carcinoma (HCC) and other advanced gastrointestinal tumors. A 3 + 3 dose escalation design was employed to assess safety, tolerability, and determine the recommended phase 2 dose (RP2D) of ADI-PEG 20. A RP2D expansion cohort for patients with HCC was employed to define the objective response rate (ORR). Secondary objectives were to estimate progression-free survival (PFS), overall survival (OS), and to explore pharmacodynamics and immunogenicity. Eligible patients were treated with mFOLFOX6 intravenously biweekly at standard doses and ADI-PEG-20 intramuscularly weekly at 18 (Cohort 1) or 36 mg/m2 (Cohort 2 and RP2D expansion). RESULTS Twenty-seven patients enrolled-23 with advanced HCC and 4 with other gastrointestinal tumors. No dose-limiting toxicities were observed in cohort 1 or 2. The RP2D for ADI-PEG 20 was 36 mg/m2 weekly with mFOLFOX6. The most common any grade adverse events (AEs) were thrombocytopenia, neutropenia, leukopenia, anemia, and fatigue. Among the 23 HCC patients, the most frequent treatment-related Grade ≥ 3 AEs were neutropenia (47.8%), thrombocytopenia (34.7%), leukopenia (21.7%), anemia (21.7%), and lymphopenia (17.4%). The ORR for this group was 21% (95% CI 7.5-43.7). Median PFS and OS were 7.3 and 14.5 months, respectively. Arginine levels were depleted with therapy despite the emergence of low levels of anti-ADI-PEG 20 antibodies. Arginine depletion at 4 and 8 weeks and archival tumoral argininosuccinate synthetase-1 levels did not correlate with response. CONCLUSIONS Concurrent mFOLFOX6 plus ADI-PEG-20 intramuscularly at 36 mg/m2 weekly shows an acceptable safety profile and favorable efficacy compared to historic controls. Further evaluation of this combination is warranted in advanced HCC patients.
Collapse
|
234
|
Sullivan LB, Luengo A, Danai LV, Bush LN, Diehl FF, Hosios AM, Lau AN, Elmiligy S, Malstrom S, Lewis CA, Vander Heiden MG. Aspartate is an endogenous metabolic limitation for tumour growth. Nat Cell Biol 2018; 20:782-788. [PMID: 29941931 PMCID: PMC6051729 DOI: 10.1038/s41556-018-0125-0] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/18/2018] [Indexed: 12/12/2022]
Abstract
Defining the metabolic limitations of tumour growth will help to develop cancer therapies1. Cancer cells proliferate slower in tumours than in standard culture conditions, indicating that a metabolic limitation may restrict cell proliferation in vivo. Aspartate synthesis can limit cancer cell proliferation when respiration is impaired2-4; however, whether acquiring aspartate is endogenously limiting for tumour growth is unknown. We confirm that aspartate has poor cell permeability, which prevents environmental acquisition, whereas the related amino acid asparagine is available to cells in tumours, but cancer cells lack asparaginase activity to convert asparagine to aspartate. Heterologous expression of guinea pig asparaginase 1 (gpASNase1), an enzyme that produces aspartate from asparagine5, confers the ability to use asparagine to supply intracellular aspartate to cancer cells in vivo. Tumours expressing gpASNase1 grow at a faster rate, indicating that aspartate acquisition is an endogenous metabolic limitation for the growth of some tumours. Tumours expressing gpASNase1 are also refractory to the growth suppressive effects of metformin, suggesting that metformin inhibits tumour growth by depleting aspartate. These findings suggest that therapeutic aspartate suppression could be effective to treat cancer.
Collapse
Affiliation(s)
- Lucas B Sullivan
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| | - Alba Luengo
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Laura V Danai
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lauren N Bush
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Frances F Diehl
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Aaron M Hosios
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Allison N Lau
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sarah Elmiligy
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Scott Malstrom
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Matthew G Vander Heiden
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
235
|
Kesarwani P, Kant S, Prabhu A, Chinnaiyan P. The interplay between metabolic remodeling and immune regulation in glioblastoma. Neuro Oncol 2018; 19:1308-1315. [PMID: 28541512 DOI: 10.1093/neuonc/nox079] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The fields of tumor metabolism and immune oncology have both independently received considerable attention over the last several years. The majority of research in tumor metabolism has largely focused on the Warburg effect and its resulting biologic consequences, including energy and macromolecule production. However, recent investigations have identified elegant, multifaceted strategies by which alterations in tumor metabolism can also contribute to a potent tolerogenic immune environment. One of the most notable is increased tryptophan metabolism through activation of indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO). However, this pathway represents one of numerous metabolic pathways that may modulate the immune system. For example, metabolites associated with aerobic glycolysis, adenosine, arginine, and prostaglandin metabolism have all been implicated in cancer-mediated immune tolerance and represent attractive therapeutic targets. In this review, we will provide an overview of the emerging interface between these 2 timely areas of cancer research and provide an overview of strategies currently being tested to target these next-generation metabolic immune checkpoints.
Collapse
Affiliation(s)
| | - Shiva Kant
- Radiation Oncology, Beaumont Health, Royal Oak, Michigan
| | - Antony Prabhu
- Radiation Oncology, Beaumont Health, Royal Oak, Michigan
| | | |
Collapse
|
236
|
Alternative assembly of respiratory complex II connects energy stress to metabolic checkpoints. Nat Commun 2018; 9:2221. [PMID: 29880867 PMCID: PMC5992162 DOI: 10.1038/s41467-018-04603-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 05/07/2018] [Indexed: 01/07/2023] Open
Abstract
Cell growth and survival depend on a delicate balance between energy production and synthesis of metabolites. Here, we provide evidence that an alternative mitochondrial complex II (CII) assembly, designated as CIIlow, serves as a checkpoint for metabolite biosynthesis under bioenergetic stress, with cells suppressing their energy utilization by modulating DNA synthesis and cell cycle progression. Depletion of CIIlow leads to an imbalance in energy utilization and metabolite synthesis, as evidenced by recovery of the de novo pyrimidine pathway and unlocking cell cycle arrest from the S-phase. In vitro experiments are further corroborated by analysis of paraganglioma tissues from patients with sporadic, SDHA and SDHB mutations. These findings suggest that CIIlow is a core complex inside mitochondria that provides homeostatic control of cellular metabolism depending on the availability of energy. Mitochondrial complex II is normally composed of four subunits. Here the authors show that bioenergetic stress conditions give rise to a partially assembled variant of complex II, which shifts the anabolic pathways to less energy demanding processes.
Collapse
|
237
|
Ochocki JD, Khare S, Hess M, Ackerman D, Qiu B, Daisak JI, Worth AJ, Lin N, Lee P, Xie H, Li B, Wubbenhorst B, Maguire TG, Nathanson KL, Alwine JC, Blair IA, Nissim I, Keith B, Simon MC. Arginase 2 Suppresses Renal Carcinoma Progression via Biosynthetic Cofactor Pyridoxal Phosphate Depletion and Increased Polyamine Toxicity. Cell Metab 2018; 27:1263-1280.e6. [PMID: 29754953 PMCID: PMC5990482 DOI: 10.1016/j.cmet.2018.04.009] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 02/14/2018] [Accepted: 04/11/2018] [Indexed: 01/02/2023]
Abstract
Kidney cancer, one of the ten most prevalent malignancies in the world, has exhibited increased incidence over the last decade. The most common subtype is "clear cell" renal cell carcinoma (ccRCC), which features consistent metabolic abnormalities, such as highly elevated glycogen and lipid deposition. By integrating metabolomics, genomic, and transcriptomic data, we determined that enzymes in multiple metabolic pathways are universally depleted in human ccRCC tumors, which are otherwise genetically heterogeneous. Notably, the expression of key urea cycle enzymes, including arginase 2 (ARG2) and argininosuccinate synthase 1 (ASS1), is strongly repressed in ccRCC. Reduced ARG2 activity promotes ccRCC tumor growth through at least two distinct mechanisms: conserving the critical biosynthetic cofactor pyridoxal phosphate and avoiding toxic polyamine accumulation. Pharmacological approaches to restore urea cycle enzyme expression would greatly expand treatment strategies for ccRCC patients, where current therapies only benefit a subset of those afflicted with renal cancer.
Collapse
Affiliation(s)
- Joshua D Ochocki
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sanika Khare
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Markus Hess
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel Ackerman
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bo Qiu
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jennie I Daisak
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew J Worth
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nan Lin
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pearl Lee
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hong Xie
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bo Li
- Program in Cancer Biology, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Bradley Wubbenhorst
- Department of Medicine, Division of Translational Medicine and Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tobi G Maguire
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katherine L Nathanson
- Department of Medicine, Division of Translational Medicine and Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James C Alwine
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ian A Blair
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Itzhak Nissim
- Division of Genetics and Metabolism, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Biochemistry, and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brian Keith
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
238
|
Jahani M, Noroznezhad F, Mansouri K. Arginine: Challenges and opportunities of this two-faced molecule in cancer therapy. Biomed Pharmacother 2018; 102:594-601. [DOI: 10.1016/j.biopha.2018.02.109] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/21/2018] [Accepted: 02/23/2018] [Indexed: 12/15/2022] Open
|
239
|
McCambridge AJ, Napolitano A, Mansfield AS, Fennell DA, Sekido Y, Nowak AK, Reungwetwattana T, Mao W, Pass HI, Carbone M, Yang H, Peikert T. Progress in the Management of Malignant Pleural Mesothelioma in 2017. J Thorac Oncol 2018; 13:606-623. [PMID: 29524617 PMCID: PMC6544834 DOI: 10.1016/j.jtho.2018.02.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 02/07/2023]
Abstract
Malignant pleural mesothelioma (MPM) is an uncommon, almost universally fatal, asbestos-induced malignancy. New and effective strategies for diagnosis, prognostication, and treatment are urgently needed. Herein we review the advances in MPM achieved in 2017. Whereas recent epidemiological data demonstrated that the incidence of MPM-related death continued to increase in United States between 2009 and 2015, new insight into the molecular pathogenesis and the immunological tumor microenvironment of MPM, for example, regarding the role of BRCA1 associated protein 1 and the expression programmed death receptor ligand 1, are highlighting new potential therapeutic strategies. Furthermore, there continues to be an ever-expanding number of clinical studies investigating systemic therapies for MPM. These trials are primarily focused on immunotherapy using immune checkpoint inhibitors alone or in combination with other immunotherapies and nonimmunotherapies. In addition, other promising targeted therapies, including pegylated adenosine deiminase (ADI-PEG20), which focuses on argininosuccinate synthase 1-deficient tumors, and tazemetostat, an enhancer of zeste 2 polycomb repressive complex 2 subunit inhibitor of BRCA1 associated protein 1 gene (BAP1)-deficient tumors, are currently being explored.
Collapse
Affiliation(s)
| | - Andrea Napolitano
- University of Hawaii Cancer Center, Honolulu, HI, USA
- Medical Oncology Department, Campus Bio-Medico, University of Rome,
Rome, Italy
| | | | - Dean A. Fennell
- Department of Genetics and Genome Biology, University of Leicester
& University Hospitals of Leicester, UK
| | - Yoshitaka Sekido
- Division of Molecular Oncology, Aichi Cancer Center Research
Institute, Chikusa-ku, Nagoya, Japan
| | - Anna K. Nowak
- Division of Medical Oncology, School of Medicine, Faculty of Health
and Medical Sciences; National Center for Asbestos Related Diseases, University of
Western Australia, Perth, Australia
| | - Thanyanan Reungwetwattana
- Division of Medical Oncology, Department of Medicine, Faculty of
Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Weimin Mao
- Department of Thoracic Surgery, Zhejiang Cancer Hospital; Key
Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zehjiang
Province, Hangzhou, China
| | - Harvey I. Pass
- Department of Cardiothoracic Surgery, New York University, Langone
Medical Center, New York, NY, USA
| | | | - Haining Yang
- University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Tobias Peikert
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic,
Rochester, MN, USA
| |
Collapse
|
240
|
De Santo C, Booth S, Vardon A, Cousins A, Tubb V, Perry T, Noyvert B, Beggs A, Ng M, Halsey C, Kearns P, Cheng P, Mussai F. The arginine metabolome in acute lymphoblastic leukemia can be targeted by the pegylated-recombinant arginase I BCT-100. Int J Cancer 2018; 142:1490-1502. [PMID: 29168171 PMCID: PMC5849425 DOI: 10.1002/ijc.31170] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 10/11/2017] [Accepted: 11/06/2017] [Indexed: 01/13/2023]
Abstract
Arginine is a semi-essential amino acid that plays a key role in cell survival and proliferation in normal and malignant cells. BCT-100, a pegylated (PEG) recombinant human arginase, can deplete arginine and starve malignant cells of the amino acid. Acute lymphoblastic leukemia (ALL) is the most common cancer of childhood, yet for patients with high risk or relapsed disease prognosis remains poor. We show that BCT-100 is cytotoxic to ALL blasts from patients in vitro by necrosis, and is synergistic in combination with dexamethasone. Against ALL xenografts, BCT-100 leads to a reduction in ALL engraftment and a prolongation of survival. ALL blasts express the arginine transporter CAT-1, yet the majority of blasts are arginine auxotrophic due to deficiency in either argininosuccinate synthase (ASS) or ornithine transcarbamylase (OTC). Although endogenous upregulation or retroviral transduced increases in ASS or OTC may promote ALL survival under moderately low arginine conditions, expression of these enzymes cannot prevent BCT-100 cytotoxicity at arginine depleting doses. RNA-sequencing of ALL blasts and supporting stromal cells treated with BCT-100 identifies a number of candidate pathways which are altered in the presence of arginine depletion. Therefore, BCT-100 provides a new clinically relevant therapeutic approach to target arginine metabolism in ALL.
Collapse
Affiliation(s)
- Carmela De Santo
- Institute of Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| | - Sarah Booth
- Institute of Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| | - Ashley Vardon
- Institute of Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| | - Antony Cousins
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary and Life Sciences, University of GlasgowUnited Kingdom
| | - Vanessa Tubb
- Institute of Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| | - Tracey Perry
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| | - Boris Noyvert
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| | - Andrew Beggs
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| | - Margaret Ng
- Department of Anatomic PathologyThe Chinese University of Hong KongHong Kong
| | - Christina Halsey
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary and Life Sciences, University of GlasgowUnited Kingdom
| | - Pamela Kearns
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| | - Paul Cheng
- Bio‐Cancer Treatment International LtdHong Kong
| | - Francis Mussai
- Institute of Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| |
Collapse
|
241
|
Turgeon MO, Perry NJS, Poulogiannis G. DNA Damage, Repair, and Cancer Metabolism. Front Oncol 2018; 8:15. [PMID: 29459886 PMCID: PMC5807667 DOI: 10.3389/fonc.2018.00015] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 01/17/2018] [Indexed: 12/12/2022] Open
Abstract
Although there has been a renewed interest in the field of cancer metabolism in the last decade, the link between metabolism and DNA damage/DNA repair in cancer has yet to be appreciably explored. In this review, we examine the evidence connecting DNA damage and repair mechanisms with cell metabolism through three principal links. (1) Regulation of methyl- and acetyl-group donors through different metabolic pathways can impact DNA folding and remodeling, an essential part of accurate double strand break repair. (2) Glutamine, aspartate, and other nutrients are essential for de novo nucleotide synthesis, which dictates the availability of the nucleotide pool, and thereby influences DNA repair and replication. (3) Reactive oxygen species, which can increase oxidative DNA damage and hence the load of the DNA-repair machinery, are regulated through different metabolic pathways. Interestingly, while metabolism affects DNA repair, DNA damage can also induce metabolic rewiring. Activation of the DNA damage response (DDR) triggers an increase in nucleotide synthesis and anabolic glucose metabolism, while also reducing glutamine anaplerosis. Furthermore, mutations in genes involved in the DDR and DNA repair also lead to metabolic rewiring. Links between cancer metabolism and DNA damage/DNA repair are increasingly apparent, yielding opportunities to investigate the mechanistic basis behind potential metabolic vulnerabilities of a substantial fraction of tumors.
Collapse
Affiliation(s)
- Marc-Olivier Turgeon
- Department of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - Nicholas J S Perry
- Department of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - George Poulogiannis
- Department of Cancer Biology, Institute of Cancer Research, London, United Kingdom.,Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| |
Collapse
|
242
|
Fischer GM, Gopal YV, McQuade JL, Peng W, DeBerardinis RJ, Davies MA. Metabolic strategies of melanoma cells: Mechanisms, interactions with the tumor microenvironment, and therapeutic implications. Pigment Cell Melanoma Res 2018; 31:11-30. [PMID: 29049843 PMCID: PMC5742019 DOI: 10.1111/pcmr.12661] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 10/09/2017] [Indexed: 12/15/2022]
Abstract
Melanomas are metabolically heterogeneous, and they are able to adapt in order to utilize a variety of fuels that facilitate tumor progression and metastasis. The significance of metabolism in melanoma is supported by growing evidence of impact on the efficacy of contemporary therapies for this disease. There are also data to support that the metabolic phenotypes of melanoma cells depend upon contributions from both intrinsic oncogenic pathways and extrinsic factors in the tumor microenvironment. This review summarizes current understanding of the metabolic processes that promote cutaneous melanoma tumorigenesis and progression, the regulation of cancer cell metabolism by the tumor microenvironment, and the impact of metabolic pathways on targeted and immune therapies.
Collapse
Affiliation(s)
- Grant M. Fischer
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
- Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
| | - Y.N. Vashisht Gopal
- Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
| | - Jennifer L. McQuade
- Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
| | - Weiyi Peng
- Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
| | - Ralph J. DeBerardinis
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd. Dallas, TX 75390
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd. Dallas, TX 75390
| | - Michael A. Davies
- Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
- Systems Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030
| |
Collapse
|
243
|
Khadeir R, Szyszko T, Szlosarek PW. Optimizing arginine deprivation for hard-to-treat cancers. Oncotarget 2017; 8:96468-96469. [PMID: 29228540 PMCID: PMC5722492 DOI: 10.18632/oncotarget.22099] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 10/24/2017] [Indexed: 12/05/2022] Open
Affiliation(s)
- Ramsay Khadeir
- Peter W. Szlosarek: Center for Molecular Oncology, Barts Cancer Institute - a Cancer Research UK Center of Excellence, Queen Mary University of London, John Vane Science Center, London, UK and Barts Health NHS Trust, St Bartholomew's Hospital, London, UK
| | - Teresa Szyszko
- Peter W. Szlosarek: Center for Molecular Oncology, Barts Cancer Institute - a Cancer Research UK Center of Excellence, Queen Mary University of London, John Vane Science Center, London, UK and Barts Health NHS Trust, St Bartholomew's Hospital, London, UK
| | - Peter W Szlosarek
- Peter W. Szlosarek: Center for Molecular Oncology, Barts Cancer Institute - a Cancer Research UK Center of Excellence, Queen Mary University of London, John Vane Science Center, London, UK and Barts Health NHS Trust, St Bartholomew's Hospital, London, UK
| |
Collapse
|
244
|
Luengo A, Gui DY, Vander Heiden MG. Targeting Metabolism for Cancer Therapy. Cell Chem Biol 2017; 24:1161-1180. [PMID: 28938091 PMCID: PMC5744685 DOI: 10.1016/j.chembiol.2017.08.028] [Citation(s) in RCA: 657] [Impact Index Per Article: 82.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/06/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Abstract
Metabolic reprogramming contributes to tumor development and introduces metabolic liabilities that can be exploited to treat cancer. Chemotherapies targeting metabolism have been effective cancer treatments for decades, and the success of these therapies demonstrates that a therapeutic window exists to target malignant metabolism. New insights into the differential metabolic dependencies of tumors have provided novel therapeutic strategies to exploit altered metabolism, some of which are being evaluated in preclinical models or clinical trials. Here, we review our current understanding of cancer metabolism and discuss how this might guide treatments targeting the metabolic requirements of tumor cells.
Collapse
Affiliation(s)
- Alba Luengo
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dan Y Gui
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Matthew G Vander Heiden
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| |
Collapse
|
245
|
PRMT7 Interacts with ASS1 and Citrullinemia Mutations Disrupt the Interaction. J Mol Biol 2017; 429:2278-2289. [DOI: 10.1016/j.jmb.2017.05.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 05/25/2017] [Accepted: 05/31/2017] [Indexed: 11/23/2022]
|
246
|
Mayevska O, Chen O, Karatsai O, Bobak Y, Barska M, Lyniv L, Pavlyk I, Rzhepetskyy Y, Igumentseva N, Redowicz MJ, Stasyk O. Nitric oxide donor augments antineoplastic effects of arginine deprivation in human melanoma cells. Exp Cell Res 2017; 355:162-171. [DOI: 10.1016/j.yexcr.2017.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/25/2017] [Accepted: 04/04/2017] [Indexed: 01/08/2023]
|
247
|
Thongkum A, Wu C, Li YY, Wangpaichitr M, Navasumrit P, Parnlob V, Sricharunrat T, Bhudhisawasdi V, Ruchirawat M, Savaraj N. The Combination of Arginine Deprivation and 5-Fluorouracil Improves Therapeutic Efficacy in Argininosuccinate Synthetase Negative Hepatocellular Carcinoma. Int J Mol Sci 2017; 18:ijms18061175. [PMID: 28587170 PMCID: PMC5485998 DOI: 10.3390/ijms18061175] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/18/2017] [Accepted: 05/26/2017] [Indexed: 12/12/2022] Open
Abstract
Argininosuccinate synthetase (ASS), a key enzyme to synthesize arginine is down regulated in many tumors including hepatocellular carcinoma (HCC). Similar to previous reports, we have found the decrease in ASS expression in poorly differentiated HCC. These ASS(-) tumors are auxotrophic for arginine. Pegylated arginine deiminase (ADI-PEG20), which degrades arginine, has shown activity in these tumors, but the antitumor effect is not robust and hence combination treatment is needed. Herein, we have elucidated the effectiveness of ADI-PEG20 combined with 5-Fluorouracil (5-FU) in ASS(-)HCC by targeting urea cycle and pyrimidine metabolism using four HCC cell lines as model. SNU398 and SNU387 express very low levels of ASS or ASS(-) while Huh-1, and HepG2 express high ASS similar to normal cells. Our results showed that the augmented cytotoxic effect of combination treatment only occurs in SNU398 and SNU387, and not in HepG2 and Huh-1 (ASS(+)) cells, and is partly due to reduced anti-apoptotic proteins X-linked inhibitor of apoptosis protein (XIAP), myeloid leukemia cell differentiation protein (Mcl-1) and B-cell lymphoma-2 (Bcl-2). Importantly, lack of ASS also influences essential enzymes in pyrimidine synthesis (carbamoyl-phosphate synthetase2, aspartate transcarbamylase and dihydrooratase (CAD) and thymidylate synthase (TS)) and malate dehydrogenase-1 (MDH-1) in TCA cycle. ADI-PEG20 treatment decreased these enzymes and made them more vulnerable to 5-FU. Transfection of ASS restored these enzymes and abolished the sensitivity to ADI-PEG20 and combination treatment. Overall, our data suggest that ASS influences multiple enzymes involved in 5-FU sensitivity. Combining ADI-PEG20 and 5-FU may be effective to treat ASS(-)hepatoma and warrants further clinical investigation.
Collapse
Affiliation(s)
- Angkana Thongkum
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Laksi, Bangkok 10210, Thailand.
- Chulabhorn Graduate Institute, Laksi, Bangkok 10210, Thailand.
| | - Chunjing Wu
- Division of Hematology/Oncology, Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA.
| | - Ying-Ying Li
- Division of Hematology/Oncology, Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Medhi Wangpaichitr
- Division of Hematology/Oncology, Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33125, USA.
| | - Panida Navasumrit
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Laksi, Bangkok 10210, Thailand.
- Chulabhorn Graduate Institute, Laksi, Bangkok 10210, Thailand.
- Center of Excellence on Environmental Health, Toxicology (EHT), Ministry of Education, Bangkok 10300, Thailand.
| | - Varabhorn Parnlob
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Laksi, Bangkok 10210, Thailand.
| | - Thaniya Sricharunrat
- Laboratory Unit of Pathology, Chulabhorn Hospital, Laksi, Bangkok 10210, Thailand.
| | - Vajarabhongsa Bhudhisawasdi
- Department of Surgery, Faculty of Medicine, Khonkaen University, Khonkaen 40000, Thailand.
- Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Laksi, Bangkok 10210, Thailand.
| | - Mathuros Ruchirawat
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Laksi, Bangkok 10210, Thailand.
- Center of Excellence on Environmental Health, Toxicology (EHT), Ministry of Education, Bangkok 10300, Thailand.
| | - Niramol Savaraj
- Division of Hematology/Oncology, Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
248
|
Beddowes E, Spicer J, Chan PY, Khadeir R, Corbacho JG, Repana D, Steele JP, Schmid P, Szyszko T, Cook G, Diaz M, Feng X, Johnston A, Thomson J, Sheaff M, Wu BW, Bomalaski J, Pacey S, Szlosarek PW. Phase 1 Dose-Escalation Study of Pegylated Arginine Deiminase, Cisplatin, and Pemetrexed in Patients With Argininosuccinate Synthetase 1-Deficient Thoracic Cancers. J Clin Oncol 2017; 35:1778-1785. [PMID: 28388291 PMCID: PMC6141244 DOI: 10.1200/jco.2016.71.3230] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Purpose Pegylated arginine deiminase (ADI-PEG 20) depletes essential amino acid levels in argininosuccinate synthetase 1 (ASS1) -negative tumors by converting arginine to citrulline and ammonia. The main aim of this study was to determine the recommended dose, safety, and tolerability of ADI-PEG 20, cisplatin, and pemetrexed in patients with ASS1-deficient malignant pleural mesothelioma (MPM) or non-small-cell lung cancer (NSCLC). Patients and Methods Using a 3 + 3 + 3 dose-escalation study, nine chemotherapy-naïve patients (five MPM, four NSCLC) received weekly ADI-PEG 20 doses of 18 mg/m2, 27 mg/m2, or 36 mg/m2, together with pemetrexed 500 mg/m2 and cisplatin 75 mg/m2 which were given every three weeks (maximum of six cycles). Patients achieving stable disease or better could continue ADI-PEG 20 monotherapy until disease progression or withdrawal. Adverse events were assessed by Common Terminology Criteria for Adverse Events version 4.03, and pharmacodynamics and immunogenicity were also evaluated. Tumor response was assessed by Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 for NSCLC and by modified RECIST criteria for MPM. Results No dose-limiting toxicities were reported; nine of 38 reported adverse events (all grade 1 or 2) were related to ADI-PEG 20. Circulating arginine concentrations declined rapidly, and citrulline levels increased; both changes persisted at 18 weeks. Partial responses were observed in seven of nine patients (78%), including three with either sarcomatoid or biphasic MPM. Conclusion Target engagement with depletion of arginine was maintained throughout treatment with no dose-limiting toxicities. In this biomarker-selected group of patients with ASS1-deficient cancers, clinical activity was observed in patients with poor-prognosis tumors. Therefore, we recommend a dose for future studies of weekly ADI-PEG 20 36 mg/m2 plus three-weekly cisplatin 75 mg/m2 and pemetrexed 500 mg/m2.
Collapse
Affiliation(s)
- Emma Beddowes
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - James Spicer
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Pui Ying Chan
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Ramsay Khadeir
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Javier Garcia Corbacho
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Dimitra Repana
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Jeremy P. Steele
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Peter Schmid
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Teresa Szyszko
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Gary Cook
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Monica Diaz
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Xiaoxing Feng
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Amanda Johnston
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Jim Thomson
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Michael Sheaff
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Bor-Wen Wu
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - John Bomalaski
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Simon Pacey
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| | - Peter W. Szlosarek
- Emma Beddowes, Javier Garcia Corbacho, and Simon Pacey, University of Cambridge, Cambridge; James Spicer, Dimitra Repana, Teresa Szyszko, and Gary Cook, King's College London; Pui Ying Chan, Jeremy P. Steele, Peter Schmid, Michael Sheaff, and Peter W. Szlosarek, St Bartholomew’s Hospital; Ramsay Khadeir, Peter Schmid, and Peter W. Szlosarek, Queen Mary University of London, London, United Kindgom; and Monica Diaz, Xiaoxing Feng, Amanda Johnston, Jim Thomson, Bor-Wen Wu, and John Bomalaski, Polaris Pharmaceuticals, San Diego, CA
| |
Collapse
|
249
|
Kim J, Hu Z, Cai L, Li K, Choi E, Faubert B, Bezwada D, Rodriguez-Canales J, Villalobos P, Lin YF, Ni M, Huffman KE, Girard L, Byers LA, Unsal-Kacmaz K, Peña CG, Heymach JV, Wauters E, Vansteenkiste J, Castrillon DH, Chen BPC, Wistuba I, Lambrechts D, Xu J, Minna JD, DeBerardinis RJ. CPS1 maintains pyrimidine pools and DNA synthesis in KRAS/LKB1-mutant lung cancer cells. Nature 2017; 546:168-172. [PMID: 28538732 PMCID: PMC5472349 DOI: 10.1038/nature22359] [Citation(s) in RCA: 225] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 04/10/2017] [Indexed: 01/05/2023]
Abstract
Metabolic reprogramming by oncogenic signals promotes cancer initiation and progression. The oncogene KRAS and tumour suppressor STK11, which encodes the kinase LKB1, regulate metabolism and are frequently mutated in non-small-cell lung cancer (NSCLC). Concurrent occurrence of oncogenic KRAS and loss of LKB1 (KL) in cells specifies aggressive oncological behaviour. Here we show that human KL cells and tumours share metabolomic signatures of perturbed nitrogen handling. KL cells express the urea cycle enzyme carbamoyl phosphate synthetase-1 (CPS1), which produces carbamoyl phosphate in the mitochondria from ammonia and bicarbonate, initiating nitrogen disposal. Transcription of CPS1 is suppressed by LKB1 through AMPK, and CPS1 expression correlates inversely with LKB1 in human NSCLC. Silencing CPS1 in KL cells induces cell death and reduces tumour growth. Notably, cell death results from pyrimidine depletion rather than ammonia toxicity, as CPS1 enables an unconventional pathway of nitrogen flow from ammonia into pyrimidines. CPS1 loss reduces the pyrimidine to purine ratio, compromises S-phase progression and induces DNA-polymerase stalling and DNA damage. Exogenous pyrimidines reverse DNA damage and rescue growth. The data indicate that the KL oncological genotype imposes a metabolic vulnerability related to a dependence on a cross-compartmental pathway of pyrimidine metabolism in an aggressive subset of NSCLC.
Collapse
Affiliation(s)
- Jiyeon Kim
- Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Zeping Hu
- Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Ling Cai
- Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Kailong Li
- Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Eunhee Choi
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Brandon Faubert
- Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Divya Bezwada
- Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, 2130 West Holcombe Boulevard, Houston, Texas 77030, USA
| | - Pamela Villalobos
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, 2130 West Holcombe Boulevard, Houston, Texas 77030, USA
| | - Yu-Fen Lin
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Min Ni
- Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Kenneth E Huffman
- Hamon Center for Therapeutic Oncology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Luc Girard
- Hamon Center for Therapeutic Oncology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Lauren A Byers
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, 2130 West Holcombe Boulevard, Houston, Texas 77030, USA
| | - Keziban Unsal-Kacmaz
- Oncology Research Unit, Pfizer, 401 North Middletown Road, Pearl River, New York 10965, USA
| | - Christopher G Peña
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, 2130 West Holcombe Boulevard, Houston, Texas 77030, USA
| | - Els Wauters
- Respiratory Division, University of Gasthuisberg, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Johan Vansteenkiste
- Respiratory Division, University of Gasthuisberg, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Diego H Castrillon
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Benjamin P C Chen
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, 2130 West Holcombe Boulevard, Houston, Texas 77030, USA
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, O&N 4 Herestraat 49 - box 912, 3000 Leuven, Belgium.,VIB Center for Cancer Biology, KU Leuven, O&N 4 Herestraat 49 - box 912, 3000 Leuven, Belgium
| | - Jian Xu
- Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - John D Minna
- Hamon Center for Therapeutic Oncology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas 75390, USA.,McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
250
|
Vander Heiden MG, DeBerardinis RJ. Understanding the Intersections between Metabolism and Cancer Biology. Cell 2017; 168:657-669. [PMID: 28187287 DOI: 10.1016/j.cell.2016.12.039] [Citation(s) in RCA: 1526] [Impact Index Per Article: 190.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/23/2016] [Accepted: 12/27/2016] [Indexed: 12/12/2022]
Abstract
Transformed cells adapt metabolism to support tumor initiation and progression. Specific metabolic activities can participate directly in the process of transformation or support the biological processes that enable tumor growth. Exploiting cancer metabolism for clinical benefit requires defining the pathways that are limiting for cancer progression and understanding the context specificity of metabolic preferences and liabilities in malignant cells. Progress toward answering these questions is providing new insight into cancer biology and can guide the more effective targeting of metabolism to help patients.
Collapse
Affiliation(s)
- Matthew G Vander Heiden
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, Department of Pediatrics, and Eugene McDermott Center for Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX 75390.
| |
Collapse
|