151
|
O'Malley J, Kumar R, Inigo J, Yadava N, Chandra D. Mitochondrial Stress Response and Cancer. Trends Cancer 2020; 6:688-701. [PMID: 32451306 DOI: 10.1016/j.trecan.2020.04.009] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/16/2020] [Accepted: 04/22/2020] [Indexed: 12/20/2022]
Abstract
Cancer cells survive and adapt to many types of stress including hypoxia, nutrient deprivation, metabolic, and oxidative stress. These stresses are sensed by diverse cellular signaling processes, leading to either degradation of mitochondria or alleviation of mitochondrial stress. This review discusses signaling during sensing and mitigation of stress involving mitochondrial communication with the endoplasmic reticulum, and how retrograde signaling upregulates the mitochondrial stress response to maintain mitochondrial integrity. The importance of the mitochondrial unfolded protein response, an emerging pathway that alleviates cellular stress, will be elaborated with respect to cancer. Detailed understanding of cellular pathways will establish mitochondrial stress response as a key mechanism for cancer cell survival leading to cancer progression and resistance, and provide a potential therapeutic target in cancer.
Collapse
Affiliation(s)
- Jordan O'Malley
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Rahul Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Joseph Inigo
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Nagendra Yadava
- Department of Anesthesiology and Center for Shock, Trauma, and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
152
|
Bruzzone C, Loizaga-Iriarte A, Sánchez-Mosquera P, Gil-Redondo R, Astobiza I, Diercks T, Cortazar AR, Ugalde-Olano A, Schäfer H, Blanco FJ, Unda M, Cannet C, Spraul M, Mato JM, Embade N, Carracedo A, Millet O. 1H NMR-Based Urine Metabolomics Reveals Signs of Enhanced Carbon and Nitrogen Recycling in Prostate Cancer. J Proteome Res 2020; 19:2419-2428. [PMID: 32380831 DOI: 10.1021/acs.jproteome.0c00091] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Prostate cancer is the second most common tumor and the fifth cause of cancer-related death among men worldwide. PC cells exhibit profound signaling and metabolic reprogramming that account for the acquisition of aggressive features. Although the metabolic understanding of this disease has increased in recent years, the analysis of such alterations through noninvasive methodologies in biofluids remains limited. Here, we used NMR-based metabolomics on a large cohort of urine samples (more than 650) from PC and benign prostate hyperplasia (BPH) patients to investigate the molecular basis of this disease. Multivariate analysis failed to distinguish between the two classes, highlighting the modest impact of prostate alterations on urine composition and the multifactorial nature of PC. However, univariate analysis of urine metabolites unveiled significant changes, discriminating PC from BPH. Metabolites with altered abundance in urine from PC patients revealed changes in pathways related to cancer biology, including glycolysis and the urea cycle. We found out that metabolites from such pathways were diminished in the urine from PC individuals, strongly supporting the notion that PC reduces nitrogen and carbon waste in order to maximize their usage in anabolic processes that support cancer cell growth.
Collapse
Affiliation(s)
- Chiara Bruzzone
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Derio 48160, Spain
| | - Ana Loizaga-Iriarte
- CIBERONC, Madrid 28025, Spain.,Department of Urology, Basurto University Hospital, Bilbao 48013, Spain
| | | | - Rubén Gil-Redondo
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Derio 48160, Spain
| | - Ianire Astobiza
- CIBERONC, Madrid 28025, Spain.,Cancer Cell Signaling and Metabolism Lab, CIC bioGUNE, Derio 48160, Spain
| | - Tammo Diercks
- Structural Biology Unit, CIC bioGUNE, Derio 48160, Spain
| | - Ana R Cortazar
- CIBERONC, Madrid 28025, Spain.,Cancer Cell Signaling and Metabolism Lab, CIC bioGUNE, Derio 48160, Spain
| | - Aitziber Ugalde-Olano
- CIBERONC, Madrid 28025, Spain.,Department of Pathology, Basurto University Hospital, Bilbao 48013, Spain
| | - Hartmut Schäfer
- Bruker Biospin GmbH, Silberstreifen, 76287 Rheinstetten, Germany
| | - Francisco J Blanco
- Structural Biology of Cancer Lab, CIC bioGUNE, Derio 48160, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao 48011, Spain
| | - Miguel Unda
- CIBERONC, Madrid 28025, Spain.,Department of Urology, Basurto University Hospital, Bilbao 48013, Spain
| | - Claire Cannet
- Bruker Biospin GmbH, Silberstreifen, 76287 Rheinstetten, Germany
| | - Manfred Spraul
- Bruker Biospin GmbH, Silberstreifen, 76287 Rheinstetten, Germany
| | - José M Mato
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Derio 48160, Spain
| | - Nieves Embade
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Derio 48160, Spain
| | - Arkaitz Carracedo
- CIBERONC, Madrid 28025, Spain.,Cancer Cell Signaling and Metabolism Lab, CIC bioGUNE, Derio 48160, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao 48011, Spain.,Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Bilbao 20018, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Derio 48160, Spain
| |
Collapse
|
153
|
Ji JX, Cochrane DR, Tessier-Cloutier B, Chen SY, Ho G, Pathak KV, Alcazar IN, Farnell D, Leung S, Cheng A, Chow C, Colborne S, Negri GL, Kommoss F, Karnezis A, Morin GB, McAlpine JN, Gilks CB, Weissman BE, Trent JM, Hoang L, Pirrotte P, Wang Y, Huntsman DG. Arginine Depletion Therapy with ADI-PEG20 Limits Tumor Growth in Argininosuccinate Synthase-Deficient Ovarian Cancer, Including Small-Cell Carcinoma of the Ovary, Hypercalcemic Type. Clin Cancer Res 2020; 26:4402-4413. [PMID: 32409304 DOI: 10.1158/1078-0432.ccr-19-1905] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 01/02/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Many rare ovarian cancer subtypes, such as small-cell carcinoma of the ovary, hypercalcemic type (SCCOHT), have poor prognosis due to their aggressive nature and resistance to standard platinum- and taxane-based chemotherapy. The development of effective therapeutics has been hindered by the rarity of such tumors. We sought to identify targetable vulnerabilities in rare ovarian cancer subtypes. EXPERIMENTAL DESIGN We compared the global proteomic landscape of six cases each of endometrioid ovarian cancer (ENOC), clear cell ovarian cancer (CCOC), and SCCOHT to the most common subtype, high-grade serous ovarian cancer (HGSC), to identify potential therapeutic targets. IHC of tissue microarrays was used as validation of arginosuccinate synthase (ASS1) deficiency. The efficacy of arginine-depriving therapeutic ADI-PEG20 was assessed in vitro using cell lines and patient-derived xenograft mouse models representing SCCOHT. RESULTS Global proteomic analysis identified low ASS1 expression in ENOC, CCOC, and SCCOHT compared with HGSC. Low ASS1 levels were validated through IHC in large patient cohorts. The lowest levels of ASS1 were observed in SCCOHT, where ASS1 was absent in 12 of 31 cases, and expressed in less than 5% of the tumor cells in 9 of 31 cases. ASS1-deficient ovarian cancer cells were sensitive to ADI-PEG20 treatment regardless of subtype in vitro. Furthermore, in two cell line mouse xenograft models and one patient-derived mouse xenograft model of SCCOHT, once-a-week treatment with ADI-PEG20 (30 mg/kg and 15 mg/kg) inhibited tumor growth in vivo. CONCLUSIONS Preclinical in vitro and in vivo studies identified ADI-PEG20 as a potential therapy for patients with rare ovarian cancers, including SCCOHT.
Collapse
Affiliation(s)
- Jennifer X Ji
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Dawn R Cochrane
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, Canada
| | - Basile Tessier-Cloutier
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Shary Yutin Chen
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, Canada
| | - Germain Ho
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, Canada
| | - Khyatiben V Pathak
- Collaborative Center for Translational Mass Spectrometry, The Translational Genomics Research Institute, Phoenix, Arizona
| | - Isabel N Alcazar
- Collaborative Center for Translational Mass Spectrometry, The Translational Genomics Research Institute, Phoenix, Arizona
| | - David Farnell
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Samuel Leung
- Genetic Pathology Evaluation Center, Vancouver, Canada
| | - Angela Cheng
- Genetic Pathology Evaluation Center, Vancouver, Canada
| | | | - Shane Colborne
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, Canada
| | - Gian Luca Negri
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, Canada
| | - Friedrich Kommoss
- Institute of Pathology, Medizin Campus Bodensee, Friedrichshafen, Germany
| | - Anthony Karnezis
- Department of Pathology and Laboratory Medicine, University of California, Davis, California
| | - Gregg B Morin
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Jessica N McAlpine
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
| | - C Blake Gilks
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Bernard E Weissman
- Department of Pathology and Laboratory Medicine, UNC-Chapel Hill, Chapel Hill, North Carolina
| | - Jeffrey M Trent
- Integrated Cancer Genomics, The Translational Genomics Research Institute, Phoenix, Arizona
| | - Lynn Hoang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Patrick Pirrotte
- Collaborative Center for Translational Mass Spectrometry, The Translational Genomics Research Institute, Phoenix, Arizona
| | - Yemin Wang
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, Canada
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada. .,Department of Molecular Oncology, BC Cancer Agency, Vancouver, Canada.,Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
154
|
Fan S, Shahid M, Jin P, Asher A, Kim J. Identification of Metabolic Alterations in Breast Cancer Using Mass Spectrometry-Based Metabolomic Analysis. Metabolites 2020; 10:E170. [PMID: 32344578 PMCID: PMC7241246 DOI: 10.3390/metabo10040170] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/12/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Breast cancer (BC) is a major global health issue and remains the second leading cause of cancer-related death in women, contributing to approximately 41,760 deaths annually. BC is caused by a combination of genetic and environmental factors. Although various molecular diagnostic tools have been developed to improve diagnosis of BC in the clinical setting, better detection tools for earlier diagnosis can improve survival rates. Given that altered metabolism is a characteristic feature of BC, we aimed to understand the comparative metabolic differences between BC and healthy controls. Metabolomics, the study of metabolism, can provide incredible insight and create useful tools for identifying potential BC biomarkers. In this study, we applied two analytical mass spectrometry (MS) platforms, including hydrophilic interaction chromatography (HILIC) and gas chromatography (GC), to generate BC-associated metabolic profiles using breast tissue from BC patients. These metabolites were further analyzed to identify differentially expressed metabolites in BC and their associated metabolic networks. Additionally, Chemical Similarity Enrichment Analysis (ChemRICH), MetaMapp, and Metabolite Set Enrichment Analysis (MSEA) identified significantly enriched clusters and networks in BC tissues. Since metabolomic signatures hold significant promise in the clinical setting, more effort should be placed on validating potential BC biomarkers based on identifying altered metabolomes.
Collapse
Affiliation(s)
- Sili Fan
- West Coast Metabolomics Center, University of California, Davis, CA 95616, USA;
| | - Muhammad Shahid
- Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (M.S.); (P.J.)
| | - Peng Jin
- Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (M.S.); (P.J.)
| | - Arash Asher
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Jayoung Kim
- Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (M.S.); (P.J.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Urology, Ga Cheon University College of Medicine, Incheon 461-701, Korea
| |
Collapse
|
155
|
López-Contreras F, Muñoz-Uribe M, Pérez-Laines J, Ascencio-Leal L, Rivera-Dictter A, Martin-Martin A, Burgos RA, Alarcon P, López-Muñoz R. Searching for Drug Synergy Against Cancer Through Polyamine Metabolism Impairment: Insight Into the Metabolic Effect of Indomethacin on Lung Cancer Cells. Front Pharmacol 2020; 10:1670. [PMID: 32256343 PMCID: PMC7093016 DOI: 10.3389/fphar.2019.01670] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most lethal and prevalent type of lung cancer. In almost all types of cancer, the levels of polyamines (putrescine, spermidine, and spermine) are increased, playing a pivotal role in tumor proliferation. Indomethacin, a non-steroidal anti-inflammatory drug, increases the abundance of an enzyme termed spermidine/spermine-N1-acetyltransferase (SSAT) encoded by the SAT1 gene. This enzyme is a key player in the export of polyamines from the cell. The aim of this study was to compare the effect of indomethacin on two NSCLC cell lines, and their combinatory potential with polyamine-inhibitor drugs in NSCLC cell lines. A549 and H1299 NSCLC cells were exposed to indomethacin and evaluations included SAT1 expression, SSAT levels, and the metabolic status of cells. Moreover, the difference in polyamine synthesis enzymes among these cell lines as well as the synergistic effect of indomethacin and chemical inhibitors of the polyamine pathway enzymes on cell viability were investigated. Indomethacin increased the expression of SAT1 and levels of SSAT in both cell lines. In A549 cells, it significantly reduced the levels of putrescine and spermidine. However, in H1299 cells, the impact of treatment on the polyamine pathway was insignificant. Also, the metabolic features upstream of the polyamine pathway (i.e., ornithine and methionine) were increased. In A549 cells, the increase of ornithine correlated with the increase of several metabolites involved in the urea cycle. Evaluation of the levels of the polyamine synthesis enzymes showed that ornithine decarboxylase is increased in A549 cells, whereas S-adenosylmethionine-decarboxylase and polyamine oxidase are increased in H1299 cells. This observation correlated with relative resistance to polyamine synthesis inhibitors eflornithine and SAM486 (inhibitors of ornithine decarboxylase and S-adenosyl-L-methionine decarboxylase, respectively), and MDL72527 (inhibitor of polyamine oxidase and spermine oxidase). Finally, indomethacin demonstrated a synergistic effect with MDL72527 in A549 cells and SAM486 in H1299 cells. Collectively, these results indicate that indomethacin alters polyamine metabolism in NSCLC cells and enhances the effect of polyamine synthesis inhibitors, such as MDL72527 or SAM486. However, this effect varies depending on the basal metabolic fingerprint of each type of cancer cell.
Collapse
Affiliation(s)
- Freddy López-Contreras
- Facultad de Ciencias Veterinarias, Instituto de Farmacología y Morfofisiología, Universidad Austral de Chile, Valdivia, Chile.,Facultad de Ciencias Veterinarias, Escuela de Graduados, Universidad Austral de Chile, Valdivia, Chile
| | - Matías Muñoz-Uribe
- Facultad de Ciencias Veterinarias, Instituto de Farmacología y Morfofisiología, Universidad Austral de Chile, Valdivia, Chile
| | - Jorge Pérez-Laines
- Facultad de Ciencias Veterinarias, Instituto de Farmacología y Morfofisiología, Universidad Austral de Chile, Valdivia, Chile
| | - Laura Ascencio-Leal
- Facultad de Ciencias Veterinarias, Instituto de Farmacología y Morfofisiología, Universidad Austral de Chile, Valdivia, Chile
| | - Andrés Rivera-Dictter
- Facultad de Ciencias Veterinarias, Instituto de Farmacología y Morfofisiología, Universidad Austral de Chile, Valdivia, Chile
| | - Antonia Martin-Martin
- Facultad de Ciencias Veterinarias, Instituto de Farmacología y Morfofisiología, Universidad Austral de Chile, Valdivia, Chile
| | - Rafael A Burgos
- Facultad de Ciencias Veterinarias, Instituto de Farmacología y Morfofisiología, Universidad Austral de Chile, Valdivia, Chile
| | - Pablo Alarcon
- Facultad de Ciencias Veterinarias, Instituto de Farmacología y Morfofisiología, Universidad Austral de Chile, Valdivia, Chile
| | - Rodrigo López-Muñoz
- Facultad de Ciencias Veterinarias, Instituto de Farmacología y Morfofisiología, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
156
|
Poirier JT, George J, Owonikoko TK, Berns A, Brambilla E, Byers LA, Carbone D, Chen HJ, Christensen CL, Dive C, Farago AF, Govindan R, Hann C, Hellmann MD, Horn L, Johnson JE, Ju YS, Kang S, Krasnow M, Lee J, Lee SH, Lehman J, Lok B, Lovly C, MacPherson D, McFadden D, Minna J, Oser M, Park K, Park KS, Pommier Y, Quaranta V, Ready N, Sage J, Scagliotti G, Sos ML, Sutherland KD, Travis WD, Vakoc CR, Wait SJ, Wistuba I, Wong KK, Zhang H, Daigneault J, Wiens J, Rudin CM, Oliver TG. New Approaches to SCLC Therapy: From the Laboratory to the Clinic. J Thorac Oncol 2020; 15:520-540. [PMID: 32018053 PMCID: PMC7263769 DOI: 10.1016/j.jtho.2020.01.016] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/12/2022]
Abstract
The outcomes of patients with SCLC have not yet been substantially impacted by the revolution in precision oncology, primarily owing to a paucity of genetic alterations in actionable driver oncogenes. Nevertheless, systemic therapies that include immunotherapy are beginning to show promise in the clinic. Although, these results are encouraging, many patients do not respond to, or rapidly recur after, current regimens, necessitating alternative or complementary therapeutic strategies. In this review, we discuss ongoing investigations into the pathobiology of this recalcitrant cancer and the therapeutic vulnerabilities that are exposed by the disease state. Included within this discussion, is a snapshot of the current biomarker and clinical trial landscapes for SCLC. Finally, we identify key knowledge gaps that should be addressed to advance the field in pursuit of reduced SCLC mortality. This review largely summarizes work presented at the Third Biennial International Association for the Study of Lung Cancer SCLC Meeting.
Collapse
Affiliation(s)
- John T Poirier
- Perlmutter Cancer Center, New York University Langone Health, New York, New York
| | - Julie George
- Department of Translational Genomics, Center of Integrated Oncology Cologne-Bonn, Medical Faculty, University of Cologne, Cologne Germany
| | | | - Anton Berns
- The Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | | | | | | | | | - Caroline Dive
- Cancer Research United Kingdom, Manchester Institute, Manchester, United Kingdom
| | - Anna F Farago
- Massachusetts General Hospital, Boston, Massachusetts
| | | | - Christine Hann
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Leora Horn
- Vanderbilt University, Nashville, Tennessee
| | | | | | - Sumin Kang
- Emory University, Winship Cancer Institute, Atlanta, Georgia
| | - Mark Krasnow
- Stanford University School of Medicine, Stanford, California
| | - James Lee
- The Ohio State University, Columbus, Ohio
| | - Se-Hoon Lee
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - Benjamin Lok
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | | | | | - John Minna
- UT Southwestern Medical Center, Dallas, Texas
| | - Matthew Oser
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - Yves Pommier
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | - Julien Sage
- Stanford University School of Medicine, Stanford, California
| | | | - Martin L Sos
- Department of Translational Genomics, Center of Integrated Oncology Cologne-Bonn, Medical Faculty, University of Cologne, Cologne Germany; Molecular Pathology, Institute of Pathology, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Kate D Sutherland
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | | | | | - Sarah J Wait
- Huntsman Cancer Institute and University of Utah, Salt Lake City, Utah
| | | | - Kwok Kin Wong
- Perlmutter Cancer Center, New York University Langone Health, New York, New York
| | - Hua Zhang
- Perlmutter Cancer Center, New York University Langone Health, New York, New York
| | - Jillian Daigneault
- International Association for the Study of Lung Cancer, Aurora, Colorado
| | - Jacinta Wiens
- International Association for the Study of Lung Cancer, Aurora, Colorado
| | | | - Trudy G Oliver
- Huntsman Cancer Institute and University of Utah, Salt Lake City, Utah.
| |
Collapse
|
157
|
Nitrogen Metabolism in Cancer and Immunity. Trends Cell Biol 2020; 30:408-424. [PMID: 32302552 DOI: 10.1016/j.tcb.2020.02.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/03/2020] [Accepted: 02/11/2020] [Indexed: 12/16/2022]
Abstract
As one of the fundamental requirements for cell growth and proliferation, nitrogen acquisition and utilization must be tightly regulated. Nitrogen can be generated from amino acids (AAs) and utilized for biosynthetic processes through transamination and deamination reactions. Importantly, limitations of nitrogen availability in cells can disrupt the synthesis of proteins, nucleic acids, and other important nitrogen-containing compounds. Rewiring cellular metabolism to support anabolic processes is a feature common to both cancer and proliferating immune cells. In this review, we discuss how nitrogen is utilized in biosynthetic pathways and highlight different metabolic and oncogenic programs that alter the flow of nitrogen to sustain biomass production and growth, an important emerging feature of cancer and immune cell proliferation.
Collapse
|
158
|
Yao S, Nguyen TV, Rolfe A, Agrawal AA, Ke J, Peng S, Colombo F, Yu S, Bouchard P, Wu J, Huang KC, Bao X, Omoto K, Selvaraj A, Yu L, Ioannidis S, Vaillancourt FH, Zhu P, Larsen NA, Bolduc DM. Small Molecule Inhibition of CPS1 Activity through an Allosteric Pocket. Cell Chem Biol 2020; 27:259-268.e5. [PMID: 32017919 DOI: 10.1016/j.chembiol.2020.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/09/2019] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Carbamoyl phosphate synthetase 1 (CPS1) catalyzes the first step in the ammonia-detoxifying urea cycle, converting ammonia to carbamoyl phosphate under physiologic conditions. In cancer, CPS1 overexpression supports pyrimidine synthesis to promote tumor growth in some cancer types, while in others CPS1 activity prevents the buildup of toxic levels of intratumoral ammonia to allow for sustained tumor growth. Targeted CPS1 inhibitors may, therefore, provide a therapeutic benefit for cancer patients with tumors overexpressing CPS1. Herein, we describe the discovery of small-molecule CPS1 inhibitors that bind to a previously unknown allosteric pocket to block ATP hydrolysis in the first step of carbamoyl phosphate synthesis. CPS1 inhibitors are active in cellular assays, blocking both urea synthesis and CPS1 support of the pyrimidine biosynthetic pathway, while having no activity against CPS2. These newly discovered CPS1 inhibitors are a first step toward providing researchers with valuable tools for probing CPS1 cancer biology.
Collapse
Affiliation(s)
- Shihua Yao
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Tuong-Vi Nguyen
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Alan Rolfe
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Anant A Agrawal
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Jiyuan Ke
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Shouyong Peng
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Federico Colombo
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Sean Yu
- RMI Laboratories LLC, 418 Industrial Drive, North Wales, PA 19454, USA
| | - Patricia Bouchard
- NMX Research and Solutions, Inc., 500 Cartier Boulevard W., Laval, Quebec H7V 5B7, Canada
| | - Jiayi Wu
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Kuan-Chun Huang
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Xingfeng Bao
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Kiyoyuki Omoto
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Anand Selvaraj
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Lihua Yu
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | | | | | - Ping Zhu
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Nicholas A Larsen
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - David M Bolduc
- H3 Biomedicine Inc., 300 Technology Square, Cambridge, MA 02139, USA.
| |
Collapse
|
159
|
Hu X, Li S, Cirillo PM, Krigbaum NY, Tran V, Jones DP, Cohn BA. Reprint of "Metabolome Wide Association Study of Serum Poly and Perfluoroalkyl Substances (PFASs) in Pregnancy and Early Postpartum". Reprod Toxicol 2020; 92:120-128. [PMID: 31923462 DOI: 10.1016/j.reprotox.2020.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 12/29/2022]
Abstract
High-resolution metabolomics (HRM) profiling of metabolic fingerprints can improve understanding of how poly and perfluoroalkyl substances (PFASs) induce metabolic alterations of in utero environment and impact fetal health. HRM profiling and quantification of PFASs were performed for 397 maternal perinatal serum samples collected from 1959-1967 in the Child Health and Development Studies (CHDS). We used Metabolome-Wide Association Studies (MWAS) and pathway enrichment analysis for metabolic associations with PFOS, its precursor EtFOSAA, and EtFOSAA-to-PFOS ratio. Distinct metabolic profiles were found with EtFOSAA and PFOS. Urea cycle metabolites such as arginine, lysine and creatine had opposite associations with EtFOSAA (negative) and PFOS (positive); whereas, carnitine shuttle metabolites were found to be exclusively and positively associated with PFOS indicating perturbation in fatty acid metabolism. These differential metabolic associations for precursor and end-product represent an important first step in identifying how PFASs alter the in utero environment and potentially leads to disease risk.
Collapse
Affiliation(s)
- Xin Hu
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30303, USA
| | - Shuzhao Li
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30303, USA
| | - Piera M Cirillo
- The Center for Research on Women and Children's Health, Child Health and Development Studies, Public Health Institute, 1683 Shattuck Avenue, Suite B, Berkeley, CA 94709, USA
| | - Nickilou Y Krigbaum
- The Center for Research on Women and Children's Health, Child Health and Development Studies, Public Health Institute, 1683 Shattuck Avenue, Suite B, Berkeley, CA 94709, USA
| | - ViLinh Tran
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30303, USA
| | - Dean P Jones
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30303, USA.
| | - Barbara A Cohn
- The Center for Research on Women and Children's Health, Child Health and Development Studies, Public Health Institute, 1683 Shattuck Avenue, Suite B, Berkeley, CA 94709, USA.
| |
Collapse
|
160
|
Dumenci OE, U AM, Khan SA, Holmes E, Taylor-Robinson SD. Exploring Metabolic Consequences of CPS1 and CAD Dysregulation in Hepatocellular Carcinoma by Network Reconstruction. J Hepatocell Carcinoma 2020; 7:1-9. [PMID: 32021853 PMCID: PMC6955626 DOI: 10.2147/jhc.s239039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 12/19/2019] [Indexed: 01/05/2023] Open
Abstract
Purpose Hepatocellular carcinoma (HCC) is the fourth commonest cause of cancer-related mortality; it is associated with various genetic alterations, some involved in metabolic reprogramming. This study aimed to explore the potential metabolic impact of Carbamoyl Phosphate Synthase I (CPS1) and carbamoyl phosphate synthetase/aspartate transcarbamoylase/dihydroorotase (CAD) dysregulation through the reconstruction of a network that integrates information from the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, Human Metabolome Database (HMDB) and Human Protein Atlas (HPA). Methods and results Existing literature was used to determine the roles of CPS1 and CAD in HCC. CPS1 downregulation is thought to play a role in hepatocarcinogenesis through an increased glutamine availability for de novo pyrimidine biosynthesis, which CAD catalyzes the first three steps for. KEGG, HMDB and HPA were used to reconstruct a network of relevant pathways, demonstrating the relationships between genes and metabolites using the MetaboSignal package in R. The network was filtered to exclude any duplicates, and those greater than three steps away from CPS1 or CAD. Consequently, a network of 18 metabolites, 28 metabolic genes and 1 signaling gene was obtained, which indicated expression profiles and prognostic information of each gene in the network. Conclusion Information from different databases was collated to form an informative network that integrated different “-omics” approaches, demonstrating the relationships between genetic and metabolic components of urea cycle and the de novo pyrimidine biosynthesis pathway. This study paves the way for further research by acting as a template to investigate the relationships between genes and metabolites, explore their potential roles in various diseases and aid the development of new screening and treatment methods through network reconstruction.
Collapse
|
161
|
Genetically modified C3A cells with restored urea cycle for improved bioartificial liver. Biocybern Biomed Eng 2020. [DOI: 10.1016/j.bbe.2019.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
162
|
Miao Y, Li Q, Wang J, Quan W, Li C, Yang Y, Mi D. Prognostic implications of metabolism-associated gene signatures in colorectal cancer. PeerJ 2020; 8:e9847. [PMID: 32953273 PMCID: PMC7474523 DOI: 10.7717/peerj.9847] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common and deadly malignancies. Novel biomarkers for the diagnosis and prognosis of this disease must be identified. Besides, metabolism plays an essential role in the occurrence and development of CRC. This article aims to identify some critical prognosis-related metabolic genes (PRMGs) and construct a prognosis model of CRC patients for clinical use. We obtained the expression profiles of CRC from The Cancer Genome Atlas database (TCGA), then identified differentially expressed PRMGs by R and Perl software. Hub genes were filtered out by univariate Cox analysis and least absolute shrinkage and selection operator Cox analysis. We used functional enrichment analysis methods, such as Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and Gene Set Enrichment Analysis, to identify involved signaling pathways of PRMGs. The nomogram predicted overall survival (OS). Calibration traces were used to evaluate the consistency between the actual and the predicted survival rate. Finally, a prognostic model was constructed based on six metabolic genes (NAT2, XDH, GPX3, AKR1C4, SPHK1, and ADCY5), and the risk score was an independent prognostic prognosticator. Genetic expression and risk score were significantly correlated with clinicopathologic characteristics of CRC. A nomogram based on the clinicopathological feature of CRC and risk score accurately predicted the OS of individual CRC cancer patients. We also validated the results in the independent colorectal cancer cohorts GSE39582 and GSE87211. Our study demonstrates that the risk score is an independent prognostic biomarker and is closely correlated with the malignant clinicopathological characteristics of CRC patients. We also determined some metabolic genes associated with the survival and clinical stage of CRC as potential biomarkers for CRC diagnosis and treatment.
Collapse
Affiliation(s)
- Yandong Miao
- The First Clinical Medical College, Lanzhou University, Lanzhou City, Gansu Province, PR China
| | - Qiutian Li
- Department of Oncology, The 920th Hospital of the Chinese People’s Liberation Army Joint Logistic Support Force, Kunming City, Yunnan Province, PR China
| | - Jiangtao Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou City, Gansu Province, PR China
| | - Wuxia Quan
- Qingyang People’s Hospital, Qingyang City, Gansu Province, PR China
| | - Chen Li
- The 3rd Affiliated Hospital, Kunming Medical College, Tumor Hospital of Yunnan Province, Kunming City, Yunnan Province, PR China
| | - Yuan Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou City, Gansu Province, PR China
| | - Denghai Mi
- The First Clinical Medical College, Lanzhou University, Lanzhou City, Gansu Province, PR China
- Gansu Academy of Traditional Chinese Medicine, Lanzhou City, Gansu Province, PR China
| |
Collapse
|
163
|
Church BV, Williams HT, Mar JC. Investigating skewness to understand gene expression heterogeneity in large patient cohorts. BMC Bioinformatics 2019; 20:668. [PMID: 31861976 PMCID: PMC6923883 DOI: 10.1186/s12859-019-3252-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Skewness is an under-utilized statistical measure that captures the degree of asymmetry in the distribution of any dataset. This study applied a new metric based on skewness to identify regulators or genes that have outlier expression in large patient cohorts. RESULTS We investigated whether specific patterns of skewed expression were related to the enrichment of biological pathways or genomic properties like DNA methylation status. Our study used publicly available datasets that were generated using both RNA-sequencing and microarray technology platforms. For comparison, the datasets selected for this study also included different samples derived from control donors and cancer patients. When comparing the shift in expression skewness between cancer and control datasets, we observed an enrichment of pathways related to the immune function that reflects an increase towards positive skewness in the cancer relative to control datasets. A significant correlation was also detected between expression skewness and the top 500 genes corresponding to the most significant differential DNA methylation occurring in the promotor regions for four Cancer Genome Atlas cancer cohorts. CONCLUSIONS Our results indicate that expression skewness can reveal new insights into transcription based on outlier and asymmetrical behaviour in large patient cohorts.
Collapse
Affiliation(s)
- Benjamin V. Church
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, 10461 NY USA
- Department of Mathematics, Columbia University, 2990 Broadway, New York, 10027 NY USA
| | - Henry T. Williams
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, 10461 NY USA
- Department of Mathematics, Columbia University, 2990 Broadway, New York, 10027 NY USA
| | - Jessica C. Mar
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, 10461 NY USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, 10461 NY USA
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, 4072 QLD Australia
| |
Collapse
|
164
|
Cable J, Finley L, Tu BP, Patti GJ, Oliver TG, Vardhana S, Mana M, Ericksen R, Khare S, DeBerardinis R, Stockwell BR, Edinger A, Haigis M, Kaelin W. Leveraging insights into cancer metabolism-a symposium report. Ann N Y Acad Sci 2019; 1462:5-13. [PMID: 31792987 DOI: 10.1111/nyas.14274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022]
Abstract
Tumor cells have devised unique metabolic strategies to garner enough nutrients to sustain continuous growth and cell division. Oncogenic mutations may alter metabolic pathways to unlock new sources of energy, and cells take the advantage of various scavenging pathways to ingest material from their environment. These changes in metabolism result in a metabolic profile that, in addition to providing the building blocks for macromolecules, can also influence cell signaling pathways to promote tumor initiation and progression. Understanding what pathways tumor cells use to synthesize the materials necessary to support metabolic growth can pave the way for new cancer therapeutics. Potential strategies include depriving tumors of the materials needed to grow or targeting pathways involved in dependencies that arise by virtue of their altered metabolis.
Collapse
Affiliation(s)
| | - Lydia Finley
- Center for Epigenetics Research, Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Benjamin P Tu
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas
| | - Gary J Patti
- Departments of Chemistry and Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Trudy G Oliver
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Santosha Vardhana
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Miyeko Mana
- The David H. Koch Institute for Integrative Cancer Research at the Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Russell Ericksen
- Singapore Bioimaging Consortium, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Sanika Khare
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ralph DeBerardinis
- Howard Hughes Medical Institute and Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, Texas
| | - Brent R Stockwell
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York
| | - Aimee Edinger
- Department of Developmental and Cell Biology, University of California, Irvine, California
| | - Marcia Haigis
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - William Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Howard Hughes Medical Institute, Chevy Chase, Maryland
| |
Collapse
|
165
|
Dervisevic E, Tuck KL, Voelcker NH, Cadarso VJ. Recent Progress in Lab-On-a-Chip Systems for the Monitoring of Metabolites for Mammalian and Microbial Cell Research. SENSORS (BASEL, SWITZERLAND) 2019; 19:E5027. [PMID: 31752167 PMCID: PMC6891382 DOI: 10.3390/s19225027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 12/11/2022]
Abstract
Lab-on-a-chip sensing technologies have changed how cell biology research is conducted. This review summarises the progress in the lab-on-a-chip devices implemented for the detection of cellular metabolites. The review is divided into two subsections according to the methods used for the metabolite detection. Each section includes a table which summarises the relevant literature and also elaborates the advantages of, and the challenges faced with that particular method. The review continues with a section discussing the achievements attained due to using lab-on-a-chip devices within the specific context. Finally, a concluding section summarises what is to be resolved and discusses the future perspectives.
Collapse
Affiliation(s)
- Esma Dervisevic
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, VIC 3800, Australia;
| | - Kellie L. Tuck
- School of Chemistry, Monash University, Clayton, VIC 3800, Australia;
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical Sciences (MIPS), Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia;
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, VIC 3168, Australia
- The Melbourne Centre for Nanofabrication, Australian National Fabrication Facility-Victorian Node, Clayton, VIC 3800, Australia
- Department of Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Victor J. Cadarso
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, VIC 3800, Australia;
- The Melbourne Centre for Nanofabrication, Australian National Fabrication Facility-Victorian Node, Clayton, VIC 3800, Australia
| |
Collapse
|
166
|
Kim DJ, Cho EJ, Yu KS, Jang IJ, Yoon JH, Park T, Cho JY. Comprehensive Metabolomic Search for Biomarkers to Differentiate Early Stage Hepatocellular Carcinoma from Cirrhosis. Cancers (Basel) 2019; 11:E1497. [PMID: 31590436 PMCID: PMC6826937 DOI: 10.3390/cancers11101497] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/02/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023] Open
Abstract
The established biomarker for hepatocellular carcinoma (HCC), serum α-fetoprotein (AFP), has suboptimal performance in early disease stages. This study aimed to develop a metabolite panel to differentiate early-stage HCC from cirrhosis. Cross-sectional metabolomic analyses of serum samples were performed for 53 and 47 patients with early HCC and cirrhosis, respectively, and 50 matched healthy controls. Results were validated in 82 and 80 patients with early HCC and cirrhosis, respectively. To retain a broad spectrum of metabolites, technically distinct analyses (global metabolomic profiling using gas chromatography time-of-flight mass spectrometry and targeted analyses using liquid chromatography with tandem mass spectrometry) were employed. Multivariate analyses classified distinct metabolites; logistic regression was employed to construct a prediction model for HCC diagnosis. Five metabolites (methionine, proline, ornithine, pimelylcarnitine, and octanoylcarnitine) were selected in a panel. The panel distinguished HCC from cirrhosis and normal controls, with an area under the receiver operating curve (AUC) of 0.82; this was significantly better than that of AFP (AUC: 0.75). During validation, the panel demonstrated significantly better predictability (AUC: 0.94) than did AFP (AUC: 0.78). Defects in ammonia recycling, the urea cycle, and amino acid metabolism, demonstrated on enrichment pathway analysis, may reliably distinguish HCC from cirrhosis. Compared with AFP alone, the metabolite panel substantially improved early-stage HCC detection.
Collapse
Affiliation(s)
- Da Jung Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea.
| | - Eun Ju Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea.
| | - In-Jin Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea.
| | - Jung-Hwan Yoon
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.
| | - Taesung Park
- Department of Statistics, Seoul National University, Seoul 08826, Korea.
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea.
| |
Collapse
|
167
|
Liu X, Zhang X, Bi J, Li Z, Zhang Z, Kong C. Caspase recruitment domain family member 10 regulates carbamoyl phosphate synthase 1 and promotes cancer growth in bladder cancer cells. J Cell Mol Med 2019; 23:8128-8138. [PMID: 31565867 PMCID: PMC6850932 DOI: 10.1111/jcmm.14683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 08/21/2019] [Accepted: 09/01/2019] [Indexed: 12/22/2022] Open
Abstract
Bladder cancer, which can be divided into non‐muscle‐invasive and muscle‐invasive bladder cancer, is the most common urinary cancer in the United States. Caspase recruitment domain family member 10 (CARD10), also named CARD‐containing MAGUK protein 3 (CARMA3), is a member of the CARMA family and may activate the nuclear factor kappa B (NF‐κB) pathway. We utilized RNA sequencing and metabolic mass spectrometry to identify the molecular and metabolic feature of CARD10. The signalling pathway of CARD10 was verified by Western blotting analysis and functional assays. RNA sequencing and metabolic mass spectrometry of CARD10 knockdown identified the metabolic enzyme carbamoyl phosphate synthase 1 (CPS1) in the urea cycle as the downstream gene regulated by CARD10. We confirmed that CARD10 affected cell proliferation and nucleotide metabolism through regulating CPS1. We indicated that CARD10 promote bladder cancer growth via CPS1 and maybe a potential therapeutic target in bladder cancer.
Collapse
Affiliation(s)
- Xi Liu
- Department of Urology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaotong Zhang
- Department of Urology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jianbin Bi
- Department of Urology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Zhenhua Li
- Department of Urology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Zhe Zhang
- Department of Urology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Chuize Kong
- Department of Urology, The First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
168
|
Ahmad F, Wang X, Jiang Z, Yu X, Liu X, Mao R, Chen X, Li W. Codoping Enhanced Radioluminescence of Nanoscintillators for X-ray-Activated Synergistic Cancer Therapy and Prognosis Using Metabolomics. ACS NANO 2019; 13:10419-10433. [PMID: 31430127 DOI: 10.1021/acsnano.9b04213] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Radio- and photodynamic therapies are the first line of cancer treatments but suffer from poor light penetration and less radiation accumulation in soft tissues with high radiation toxicity. Therefore, a multifunctional nanoplatform with diagnosis-assisted synergistic radio- and photodynamic therapy and tools facilitating early prognosis are urgently needed to fight the war against cancer. Further, integrating cancer therapy with untargeted metabolomic analysis would collectively offer clinical pertinence through facilitating early diagnosis and prognosis. Here, we enriched scintillation of CeF3 nanoparticles (NPs) through codoping Tb3+ and Gd3+ (CeF3:Gd3+,Tb3+) for viable clinical approach in the treatment of deep-seated tumors. The codoped CeF3:Gd3+,Tb3+ scintillating theranostic NPs were then coated with mesoporous silica, followed by loading with rose bengal (CGTS-RB) for later computed tomography (CT)- and magnetic resonance image (MRI)-guided X-ray stimulated synergistic radio- and photodynamic therapy (RT+XPDT) using low-dose, one-time X-ray irradiation. The results corroborated an efficient tumor regression with synergistic RT+XPDT relative to single RT. Global untargeted metabolome shifts highlighted the mechanism behind this efficient tumor regression using RT, and synergistic RT+XPDT treatment is due to the starvation of nonessential amino acids involved in protein and DNA synthesis and energy regulation pathways necessary for growth and progression. Our study also concluded that tumor and serum metabolites shift during disease progression and regression and serve as robust biomarkers for early assessment of disease state and prognosis. From our results, we propose that codoping is an effective and extendable technique to other materials for gaining high optical yield and multifunctionality and for use in diagnostic and therapeutic applications. Critically, the integration of multifunctional theranostic nanomedicines with metabolomics has excellent potential for the discovery of early metabolic biomarkers to aid in better clinical disease diagnosis and prognosis.
Collapse
Affiliation(s)
- Farooq Ahmad
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering , Shanghai Jiao Tong University , Shanghai 200240 , P.R. China
| | - Xiaoyan Wang
- Shanghai Center for Systems Biomedicine , Shanghai Jiao Tong University , Shanghai 200240 , P.R. China
| | - Zhao Jiang
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering , Shanghai Jiao Tong University , Shanghai 200240 , P.R. China
| | - Xujiang Yu
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering , Shanghai Jiao Tong University , Shanghai 200240 , P.R. China
| | - Xinyi Liu
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering , Shanghai Jiao Tong University , Shanghai 200240 , P.R. China
| | - Rihua Mao
- Laboratory for Advanced Scintillation Materials & Performance , Shanghai Institute of Ceramics, Chinese Academy of Sciences , Shanghai , 201800 , P.R. China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Wanwan Li
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering , Shanghai Jiao Tong University , Shanghai 200240 , P.R. China
| |
Collapse
|
169
|
Abstract
Macroautophagy (referred to here as autophagy) degrades and recycles cytoplasmic constituents to sustain cellular and mammalian metabolism and survival during starvation. Deregulation of autophagy is involved in numerous diseases, such as cancer. Cancers up-regulate autophagy and depend on it for survival, growth, and malignancy in a tumor cell-autonomous fashion. Recently, it has become apparent that autophagy in host tissues as well as the tumor cells themselves contribute to tumor growth. Understanding how autophagy regulates metabolism and tumor growth has revealed new essential tumor nutrients, where they come from, and how they are supplied and used, which can now be targeted for cancer therapy.
Collapse
Affiliation(s)
- Laura Poillet-Perez
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
170
|
Hu X, Li S, Cirillo PM, Krigbaum NY, Tran V, Jones DP, Cohn BA. Metabolome Wide Association Study of Serum Poly and Perfluoroalkyl Substances (PFASs) in Pregnancy and Early Postpartum. Reprod Toxicol 2019; 87:70-78. [PMID: 31121237 DOI: 10.1016/j.reprotox.2019.05.058] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 12/16/2022]
Abstract
High-resolution metabolomics (HRM) profiling of metabolic fingerprints can improve understanding of how poly and perfluoroalkyl substances (PFASs) induce metabolic alterations of in utero environment and impact fetal health. HRM profiling and quantification of PFASs were performed for 397 maternal perinatal serum samples collected from 1959-1967 in the Child Health and Development Studies (CHDS). We used Metabolome-Wide Association Studies (MWAS) and pathway enrichment analysis for metabolic associations with PFOS, its precursor EtFOSAA, and EtFOSAA-to-PFOS ratio. Distinct metabolic profiles were found with EtFOSAA and PFOS. Urea cycle metabolites such as arginine, lysine and creatine had opposite associations with EtFOSAA (negative) and PFOS (positive); whereas, carnitine shuttle metabolites were found to be exclusively and positively associated with PFOS indicating perturbation in fatty acid metabolism. These differential metabolic associations for precursor and end-product represent an important first step in identifying how PFASs alter the in utero environment and potentially leads to disease risk.
Collapse
Affiliation(s)
- Xin Hu
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30303, USA
| | - Shuzhao Li
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30303, USA
| | - Piera M Cirillo
- The Center for Research on Women and Children's Health, Child Health and Development Studies, Public Health Institute, 1683 Shattuck Avenue, Suite B, Berkeley, CA 94709, USA
| | - Nickilou Y Krigbaum
- The Center for Research on Women and Children's Health, Child Health and Development Studies, Public Health Institute, 1683 Shattuck Avenue, Suite B, Berkeley, CA 94709, USA
| | - ViLinh Tran
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30303, USA
| | - Dean P Jones
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30303, USA.
| | - Barbara A Cohn
- The Center for Research on Women and Children's Health, Child Health and Development Studies, Public Health Institute, 1683 Shattuck Avenue, Suite B, Berkeley, CA 94709, USA.
| |
Collapse
|
171
|
An essential role for GLUT5-mediated fructose utilization in exacerbating the malignancy of clear cell renal cell carcinoma. Cell Biol Toxicol 2019; 35:471-483. [PMID: 31102011 DOI: 10.1007/s10565-019-09478-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/06/2019] [Indexed: 10/26/2022]
Abstract
Fructose is an important alternative carbon source for several tumors, and GLUT5 is the major fructose transporter which mediates most of fructose uptake in cells. So far, it is unclear whether GLUT5-mediated fructose utilization is important for clear cell renal cell carcinoma (ccRCC). Here, we demonstrated that GLUT5 was highly expressed in a panel of ccRCC cell lines. High GLUT5 expression exacerbated the neoplastic phenotypes of ccRCC cells, including cell proliferation and colony formation. On the other hand, deletion of the GLUT5-encoding gene SLC2A5 dramatically attenuated cellular malignancy via activating the apoptotic pathway. Moreover, administration of 2,5-anhydro-D-mannitol (2,5-AM), a competitive inhibitor of fructose uptake, could markedly suppress ccRCC cell growth. Together, we provide a new mechanistic insight for GLUT5-mediated fructose utilization in ccRCC cells and highlight the therapeutic potential for targeting this metabolic pathway against ccRCC.
Collapse
|
172
|
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: 105] [Impact Index Per Article: 21.0] [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
|
173
|
The Fate of Glutamine in Human Metabolism. The Interplay with Glucose in Proliferating Cells. Metabolites 2019; 9:metabo9050081. [PMID: 31027329 PMCID: PMC6571637 DOI: 10.3390/metabo9050081] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/23/2019] [Indexed: 01/13/2023] Open
Abstract
Genome-scale models of metabolism (GEM) are used to study how metabolism varies in different physiological conditions. However, the great number of reactions involved in GEM makes it difficult to understand these variations. In order to have a more understandable tool, we developed a reduced metabolic model of central carbon and nitrogen metabolism, C2M2N with 77 reactions, 54 internal metabolites, and 3 compartments, taking into account the actual stoichiometry of the reactions, including the stoichiometric role of the cofactors and the irreversibility of some reactions. In order to model oxidative phosphorylation (OXPHOS) functioning, the proton gradient through the inner mitochondrial membrane is represented by two pseudometabolites DPH (∆pH) and DPSI (∆ψ). To illustrate the interest of such a reduced and quantitative model of metabolism in mammalian cells, we used flux balance analysis (FBA) to study all the possible fates of glutamine in metabolism. Our analysis shows that glutamine can supply carbon sources for cell energy production and can be used as carbon and nitrogen sources to synthesize essential metabolites. Finally, we studied the interplay between glucose and glutamine for the formation of cell biomass according to ammonia microenvironment. We then propose a quantitative analysis of the Warburg effect.
Collapse
|
174
|
Mazat JP, Ransac S. The Fate of Glutamine in Human Metabolism. The Interplay with Glucose in Proliferating Cells. Metabolites 2019. [PMID: 31027329 DOI: 10.1101/477224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Genome-scale models of metabolism (GEM) are used to study how metabolism varies in different physiological conditions. However, the great number of reactions involved in GEM makes it difficult to understand these variations. In order to have a more understandable tool, we developed a reduced metabolic model of central carbon and nitrogen metabolism, C2M2N with 77 reactions, 54 internal metabolites, and 3 compartments, taking into account the actual stoichiometry of the reactions, including the stoichiometric role of the cofactors and the irreversibility of some reactions. In order to model oxidative phosphorylation (OXPHOS) functioning, the proton gradient through the inner mitochondrial membrane is represented by two pseudometabolites DPH (∆pH) and DPSI (∆ψ). To illustrate the interest of such a reduced and quantitative model of metabolism in mammalian cells, we used flux balance analysis (FBA) to study all the possible fates of glutamine in metabolism. Our analysis shows that glutamine can supply carbon sources for cell energy production and can be used as carbon and nitrogen sources to synthesize essential metabolites. Finally, we studied the interplay between glucose and glutamine for the formation of cell biomass according to ammonia microenvironment. We then propose a quantitative analysis of the Warburg effect.
Collapse
Affiliation(s)
- Jean-Pierre Mazat
- IBGC CNRS UMR 5095 & Université de Bordeaux, 1, rue Camille Saint-Saëns, 33077 Bordeaux-CEDEX, France.
| | - Stéphane Ransac
- IBGC CNRS UMR 5095 & Université de Bordeaux, 1, rue Camille Saint-Saëns, 33077 Bordeaux-CEDEX, France.
| |
Collapse
|
175
|
Cho A, Eskandari R, Granlund KL, Keshari KR. Hyperpolarized [6- 13C, 15N 3]-Arginine as a Probe for in Vivo Arginase Activity. ACS Chem Biol 2019; 14:665-673. [PMID: 30893552 DOI: 10.1021/acschembio.8b01044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Alterations in arginase enzyme expression are linked with various diseases and have been shown to support disease progression, thus motivating the development of an imaging probe for this enzymatic target. 13C-enriched arginine can be used as a hyperpolarized (HP) magnetic resonance (MR) probe for arginase flux since the arginine carbon-6 resonance (157 ppm) is converted to urea (163 ppm) following arginase-catalyzed hydrolysis. However, scalar relaxation from adjacent 14N-nuclei shortens cabon-6 T 1 and T 2 times, yielding poor spectral properties. To address these limitations, we report the synthesis of [6-13C,15N3]-arginine and demonstrate that 15N-enrichment increases carbon-6 relaxation times, thereby improving signal-to-noise ratio and spectral resolution. By overcoming these limitations with this novel isotope-labeling scheme, we were able to perform in vitro and in vivo arginase activity measurements with HP MR. We present HP [6-13C,15N3]-arginine as a noninvasive arginase imaging agent for preclinical studies, with the potential for future clinical diagnostic use.
Collapse
Affiliation(s)
- Andrew Cho
- Department of Biochemistry and Structural Biology, Weill Cornell Graduate School, New York, New York 10065, United States
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, New York 10065, United States
| | - Roozbeh Eskandari
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Kristin L. Granlund
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Kayvan R. Keshari
- Department of Biochemistry and Structural Biology, Weill Cornell Graduate School, New York, New York 10065, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| |
Collapse
|
176
|
Cao Y, Ding W, Zhang J, Gao Q, Yang H, Cao W, Wang Z, Fang L, Du R. Significant Down-Regulation of Urea Cycle Generates Clinically Relevant Proteomic Signature in Hepatocellular Carcinoma Patients with Macrovascular Invasion. J Proteome Res 2019; 18:2032-2044. [DOI: 10.1021/acs.jproteome.8b00921] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yin Cao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Gulou District, Nanjing 210093, China
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Gulou District, Nanjing 210008, China
| | - WenWen Ding
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Gulou District, Nanjing 210093, China
| | - JingZi Zhang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Gulou District, Nanjing 210093, China
| | - Qi Gao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Gulou District, Nanjing 210093, China
| | - HaoXiang Yang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Gulou District, Nanjing 210093, China
| | - WangSen Cao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Gulou District, Nanjing 210093, China
| | - ZhongXia Wang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Gulou District, Nanjing 210008, China
| | - Lei Fang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Gulou District, Nanjing 210093, China
| | - RongHui Du
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Gulou District, Nanjing 210093, China
| |
Collapse
|
177
|
Defining Metabolic Rewiring in Lung Squamous Cell Carcinoma. Metabolites 2019; 9:metabo9030047. [PMID: 30866469 PMCID: PMC6468359 DOI: 10.3390/metabo9030047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 02/26/2019] [Accepted: 03/02/2019] [Indexed: 01/19/2023] Open
Abstract
Metabolomics based on untargeted flow infusion electrospray ionization high-resolution mass spectrometry (FIE-HRMS) can provide a snap-shot of metabolism in living cells. Lung Squamous Cell Carcinoma (SCC) is one of the predominant subtypes of Non-Small Cell Lung Cancers (NSCLCs), which usually shows a poor prognosis. We analysed lung SCC samples and matched histologically normal lung tissues from eight patients. Metabolites were profiled by FIE-HRMS and assessed using t-test and principal component analysis (PCA). Differentially accumulating metabolites were mapped to pathways using the mummichog algorithm in R, and biologically meaningful patterns were indicated by Metabolite Set Enrichment Analysis (MSEA). We identified metabolic rewiring networks, including the suppression of the oxidative pentose pathway and found that the normal tricarboxylic acid (TCA) cycle were decoupled from increases in glycolysis and glutamine reductive carboxylation. Well-established associated effects on nucleotide, amino acid and thiol metabolism were also seen. Novel aspects in SCC tissue were increased in Vitamin B complex cofactors, serotonin and a reduction of γ-aminobutyric acid (GABA). Our results show the value of FIE-HRMS as a high throughput screening method that could be exploited in clinical contexts.
Collapse
|
178
|
Qi Y, Qi H, Liu Z, He P, Li B. Bioinformatics Analysis of Key Genes and Pathways in Colorectal Cancer. J Comput Biol 2019; 26:364-375. [PMID: 30810359 DOI: 10.1089/cmb.2018.0237] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent cancer in the world. Although great progress has been made, the specific molecular mechanism remains unclear. This study aimed to explore the differentially expressed genes (DEGs) and underlying mechanisms of CRC using bioinformatics analysis. In this study, we identified a total of 1353 DEGs in the database of GSE113513, including 715 up- and 638 downregulated genes. Gene ontology analysis results showed that upregulated DEGs were significantly enriched in cell division, cell proliferation, and DNA replication. The downregulated DEGs were enriched in immune response, relation of cell growth and inflammatory response. The Kyoto Encyclopedia of Genes and Genomes pathway analysis showed that upregulated DEGs were enriched in cell cycle and p53 signaling pathway, whereas the downregulated DEGs were enriched in drug metabolism, metabolism of xenobiotics by cytochrome P450, and nitrogen metabolism. A total of 124 up-key genes and 35 down-key genes were identified from the protein-protein interaction networks. Furthermore, we identified five up-modules (up-A, up-B, up-C, up-D, and up-E) and three down-modules (d-A, d-B, and d-C) by module analysis. The module up-A was enriched in sister chromatid cohesion, cell division, and mitotic nuclear division. Pathways associated with cell cycle, progesterone-mediated oocyte maturation, oocyte meiosis, and p53 signaling pathway. Whereas the d-A was mainly enriched in G-protein coupled receptor signaling pathway, cell chemotaxis, and chemokine-mediated signaling pathway. The pathways enriched in chemokine signaling pathway, cytokine-cytokine receptor interaction, and alcoholism. These key genes and pathways might be used as molecular targets and diagnostic biomarkers for the treatment of CRC.
Collapse
Affiliation(s)
- Yuewen Qi
- 1 Department of Gastroenterology, Affiliated Hospital of Chengde Medical College, Chengde, P.R. China
| | - Haowen Qi
- 2 Department of Acupuncture and Massage, Chengde Hospital of Traditional Chinese Medicine, Chengde, P.R. China
| | - Zeyuan Liu
- 3 Department of Special Medicine, Qingdao University Medical College, Qingdao, P.R. China
| | - Peiyuan He
- 1 Department of Gastroenterology, Affiliated Hospital of Chengde Medical College, Chengde, P.R. China
| | - Bingqing Li
- 1 Department of Gastroenterology, Affiliated Hospital of Chengde Medical College, Chengde, P.R. China
| |
Collapse
|
179
|
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: 10.2] [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
|
180
|
Marx D, Metzger J, Olagne J, Belczacka I, Faguer S, Colombat M, Husi H, Mullen W, Gwinner W, Caillard S. Proteomics in Kidney Allograft Transplantation—Application of Molecular Pathway Analysis for Kidney Allograft Disease Phenotypic Biomarker Selection. Proteomics Clin Appl 2019; 13:e1800091. [DOI: 10.1002/prca.201800091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/10/2019] [Indexed: 02/06/2023]
Affiliation(s)
- David Marx
- Nephrology – Transplantation DepartmentUMR_S. INSERM UMR_S 1109ImmunoRhumatologie MoléculaireFédération Hospitalo‐Universitaire OMICAREFédération de Médecine Translationnelle de StrasbourgInstitut d'Immunologie et d'Hématologie 67085 Strasbourg France
- Laboratoire de Spectrométrie de Masse BioOrganiqueUniversity of StrasbourgCentre National de la Recherche ScientifiqueInstitut Pluridisciplinaire Hubert Curien UMR 7178 67037 Strasbourg France
| | | | - Jérôme Olagne
- Nephrology – Transplantation DepartmentUMR_S. INSERM UMR_S 1109ImmunoRhumatologie MoléculaireFédération Hospitalo‐Universitaire OMICAREFédération de Médecine Translationnelle de StrasbourgInstitut d'Immunologie et d'Hématologie 67085 Strasbourg France
- Department of PathologyUniversity Hospital of Strasbourg 67091 Strasbourg France
| | | | - Stanislas Faguer
- Department of Nephrology and Organ TransplantationUniversity Hospital of Toulouse 31059 Toulouse France
- Institut National de la Santé et de la Recherche Médicale (INSERM)Institut of Cardiovascular and Metabolic Disease U1048 31432 Toulouse France
- Université Toulouse III Paul‐Sabatier 31330 Toulouse France
| | - Magali Colombat
- Department of PathologyCancer University Institute of Toulouse 31100 Toulouse France
| | - Holger Husi
- Division of Biomedical SciencesCentre for Health ScienceUniversity of the Highlands and Islands Inverness IV2 3JH UK
| | - William Mullen
- Institute of Cardiovascular and Medical SciencesUniversity of Glasgow Glasgow G12 8TA United Kingdom
| | - Wilfried Gwinner
- Department of NephrologyHannover Medical School 30625 Hannover Germany
| | - Sophie Caillard
- Nephrology – Transplantation DepartmentUMR_S. INSERM UMR_S 1109ImmunoRhumatologie MoléculaireFédération Hospitalo‐Universitaire OMICAREFédération de Médecine Translationnelle de StrasbourgInstitut d'Immunologie et d'Hématologie 67085 Strasbourg France
| |
Collapse
|
181
|
Lin C, Salzillo TC, Bader DA, Wilkenfeld SR, Awad D, Pulliam TL, Dutta P, Pudakalakatti S, Titus M, McGuire SE, Bhattacharya PK, Frigo DE. Prostate Cancer Energetics and Biosynthesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:185-237. [PMID: 31900911 PMCID: PMC8096614 DOI: 10.1007/978-3-030-32656-2_10] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancers must alter their metabolism to satisfy the increased demand for energy and to produce building blocks that are required to create a rapidly growing tumor. Further, for cancer cells to thrive, they must also adapt to an often changing tumor microenvironment, which can present new metabolic challenges (ex. hypoxia) that are unfavorable for most other cells. As such, altered metabolism is now considered an emerging hallmark of cancer. Like many other malignancies, the metabolism of prostate cancer is considerably different compared to matched benign tissue. However, prostate cancers exhibit distinct metabolic characteristics that set them apart from many other tumor types. In this chapter, we will describe the known alterations in prostate cancer metabolism that occur during initial tumorigenesis and throughout disease progression. In addition, we will highlight upstream regulators that control these metabolic changes. Finally, we will discuss how this new knowledge is being leveraged to improve patient care through the development of novel biomarkers and metabolically targeted therapies.
Collapse
Affiliation(s)
- Chenchu Lin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Travis C Salzillo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - David A Bader
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Sandi R Wilkenfeld
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Dominik Awad
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Thomas L Pulliam
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Prasanta Dutta
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shivanand Pudakalakatti
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark Titus
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sean E McGuire
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pratip K Bhattacharya
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA.
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Molecular Medicine Program, The Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
182
|
Wang S, Li F, Qiao R, Hu X, Liao H, Chen L, Wu J, Wu H, Zhao M, Liu J, Chen R, Ma X, Kim D, Sun J, Davis TP, Chen C, Tian J, Hyeon T, Ling D. Arginine-Rich Manganese Silicate Nanobubbles as a Ferroptosis-Inducing Agent for Tumor-Targeted Theranostics. ACS NANO 2018; 12:12380-12392. [PMID: 30495919 DOI: 10.1021/acsnano.8b06399] [Citation(s) in RCA: 257] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Ferroptosis, an iron-based cell-death pathway, has recently attracted great attention owing to its effectiveness in killing cancer cells. Previous investigations focused on the development of iron-based nanomaterials to induce ferroptosis in cancer cells by the up-regulation of reactive oxygen species (ROS) generated by the well-known Fenton reaction. Herein, we report a ferroptosis-inducing agent based on arginine-rich manganese silicate nanobubbles (AMSNs) that possess highly efficient glutathione (GSH) depletion ability and thereby induce ferroptosis by the inactivation of glutathione-dependent peroxidases 4 (GPX4). The AMSNs were synthesized via a one-pot reaction with arginine (Arg) as the surface ligand for tumor homing. Subsequently, a significant tumor suppression effect can be achieved by GSH depletion-induced ferroptosis. Moreover, the degradation of AMSNs during the GSH depletion contributed to T1-weighted magnetic resonance imaging (MRI) enhancement as well as on-demand chemotherapeutic drug release for synergistic cancer therapy. We anticipate that the GSH-depletion-induced ferroptosis strategy by using manganese-based nanomaterials would provide insights in designing nanomedicines for tumor-targeted theranostics.
Collapse
Affiliation(s)
| | | | - Ruirui Qiao
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , Monash Institute of Pharmaceutical Sciences, Monash University , Parkville , Victoria 3052 , Australia
| | | | | | - Lumin Chen
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine , Hangzhou 310058 , China
| | | | | | | | - Jianan Liu
- Center for Nanoparticle Research , Institute for Basic Science (IBS) , Seoul , 08826 , Korea
| | - Rui Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , National Center for Nanoscience & Technology of China , Beijing , 100190 , China
| | - Xibo Ma
- Chinese Academy of Sciences Key Laboratory of Molecular Imaging , Institute of Automation, Chinese Academy of Sciences , Beijing 100190 , China
| | - Dokyoon Kim
- Center for Nanoparticle Research , Institute for Basic Science (IBS) , Seoul , 08826 , Korea
| | - Jihong Sun
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine , Hangzhou 310058 , China
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , Monash Institute of Pharmaceutical Sciences, Monash University , Parkville , Victoria 3052 , Australia
- Department of Chemistry , University of Warwick , Coventry , West Midlands CV4 7AL , United Kingdom
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , National Center for Nanoscience & Technology of China , Beijing , 100190 , China
| | - Jie Tian
- Chinese Academy of Sciences Key Laboratory of Molecular Imaging , Institute of Automation, Chinese Academy of Sciences , Beijing 100190 , China
| | - Taeghwan Hyeon
- Center for Nanoparticle Research , Institute for Basic Science (IBS) , Seoul , 08826 , Korea
- School of Chemical and Biological Engineering , Seoul National University , Seoul , 08826 , Korea
| | - Daishun Ling
- Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science , Zhejiang University , Hangzhou 310058 , China
| |
Collapse
|
183
|
Seton-Rogers S. Changing mutational patterns. Nat Rev Cancer 2018; 18:597. [PMID: 30209308 DOI: 10.1038/s41568-018-0058-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|