101
|
Wilde BR, Ye Z, Lim TY, Ayer DE. Cellular acidosis triggers human MondoA transcriptional activity by driving mitochondrial ATP production. eLife 2019; 8:40199. [PMID: 30717828 PMCID: PMC6363388 DOI: 10.7554/elife.40199] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/22/2019] [Indexed: 12/12/2022] Open
Abstract
Human MondoA requires glucose as well as other modulatory signals to function in transcription. One such signal is acidosis, which increases MondoA activity and also drives a protective gene signature in breast cancer. How low pH controls MondoA transcriptional activity is unknown. We found that low pH medium increases mitochondrial ATP (mtATP), which is subsequently exported from the mitochondrial matrix. Mitochondria-bound hexokinase transfers a phosphate from mtATP to cytoplasmic glucose to generate glucose-6-phosphate (G6P), which is an established MondoA activator. The outer mitochondrial membrane localization of MondoA suggests that it is positioned to coordinate the adaptive transcriptional response to a cell’s most abundant energy sources, cytoplasmic glucose and mtATP. In response to acidosis, MondoA shows preferential binding to just two targets, TXNIP and its paralog ARRDC4. Because these transcriptional targets are suppressors of glucose uptake, we propose that MondoA is critical for restoring metabolic homeostasis in response to high energy charge.
Collapse
Affiliation(s)
- Blake R Wilde
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, United States
| | - Zhizhou Ye
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, United States
| | - Tian-Yeh Lim
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, United States
| | - Donald E Ayer
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, United States
| |
Collapse
|
102
|
Baldini N, Avnet S. The Effects of Systemic and Local Acidosis on Insulin Resistance and Signaling. Int J Mol Sci 2018; 20:ijms20010126. [PMID: 30598026 PMCID: PMC6337415 DOI: 10.3390/ijms20010126] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 12/12/2018] [Accepted: 12/23/2018] [Indexed: 12/14/2022] Open
Abstract
Most pathological conditions that cause local or systemic acidosis by overcoming the buffering activities of body fluids overlap with those diseases that are characterized by glucose metabolic disorders, including diabetes mellitus, inflammation, and cancer. This simple observation suggests the existence of a strong relationship between acidosis and insulin metabolism or insulin receptor signaling. In this review, we summarized the current knowledge on the activity of insulin on the induction of acidosis and, vice versa, on the effects of changes of extracellular and intracellular pH on insulin resistance. Insulin influences acidosis by promoting glycolysis. Although with an unclear mechanism, the lowering of pH, in turn, inhibits insulin sensitivity or activity. In addition to ketoacidosis that is frequently associated with diabetes, other important and more complex factors are involved in this delicate feedback mechanism. Among these, in this review we discussed the acid-mediated inhibiting effects on insulin binding affinity to its receptor, on glycolysis, on the recycling of glucose transporters, and on insulin secretion via transforming growth factor β (TGF-β) activity by pancreatic β-cells. Finally, we revised current data available on the mutual interaction between insulin signaling and the activity of ion/proton transporters and pH sensors, and on how acidosis may enhance insulin resistance through the Nuclear Factor kappa B (NF-κB) inflammatory pathway.
Collapse
Affiliation(s)
- Nicola Baldini
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli IRCCS, 40136 Bologna, Italy.
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 401223 Bologna, Italy.
| | - Sofia Avnet
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli IRCCS, 40136 Bologna, Italy.
| |
Collapse
|
103
|
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
|
104
|
Rabiee S, Tavakol S, Barati M, Joghataei MT. Autophagic, apoptotic, and necrotic cancer cell fates triggered by acidic pH microenvironment. J Cell Physiol 2018; 234:12061-12069. [DOI: 10.1002/jcp.27876] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 11/13/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Shadi Rabiee
- Department of Biology Rasht Branch, Islamic Azad University Rasht Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences Tehran Iran
| | - Mahmoud Barati
- Department of Medical Biotechnology Iran University of Medical Sciences Tehran Iran
| | | |
Collapse
|
105
|
Andreucci E, Pietrobono S, Peppicelli S, Ruzzolini J, Bianchini F, Biagioni A, Stecca B, Calorini L. SOX2 as a novel contributor of oxidative metabolism in melanoma cells. Cell Commun Signal 2018; 16:87. [PMID: 30466459 PMCID: PMC6249961 DOI: 10.1186/s12964-018-0297-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/08/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Deregulated metabolism is a hallmark of cancer and recent evidence underlines that targeting tumor energetics may improve therapy response and patient outcome. Despite the general attitude of cancer cells to exploit the glycolytic pathway even in the presence of oxygen (aerobic glycolysis or "Warburg effect"), tumor metabolism is extremely plastic, and such ability to switch from glycolysis to oxidative phosphorylation (OxPhos) allows cancer cells to survive under hostile microenvironments. Recently, OxPhos has been related with malignant progression, chemo-resistance and metastasis. OxPhos is induced under extracellular acidosis, a well-known characteristic of most solid tumors, included melanoma. METHODS To evaluate whether SOX2 modulation is correlated with metabolic changes under standard or acidic conditions, SOX2 was silenced and overexpressed in several melanoma cell lines. To demonstrate that SOX2 directly represses HIF1A expression we used chromatin immunoprecipitation (ChIP) and luciferase assay. RESULTS In A375-M6 melanoma cells, extracellular acidosis increases SOX2 expression, that sustains the oxidative cancer metabolism exploited under acidic conditions. By studying non-acidic SSM2c and 501-Mel melanoma cells (high- and very low-SOX2 expressing cells, respectively), we confirmed the metabolic role of SOX2, attributing SOX2-driven OxPhos reprogramming to HIF1α pathway disruption. CONCLUSIONS SOX2 contributes to the acquisition of an aggressive oxidative tumor phenotype, endowed with enhanced drug resistance and metastatic ability.
Collapse
Affiliation(s)
- Elena Andreucci
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Viale G.B. Morgagni, 50, 50134, Florence, Italy
| | - Silvia Pietrobono
- Core Research Laboratory, Institute for Cancer Research and Prevention (ISPRO), Florence, Italy
| | - Silvia Peppicelli
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Viale G.B. Morgagni, 50, 50134, Florence, Italy
| | - Jessica Ruzzolini
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Viale G.B. Morgagni, 50, 50134, Florence, Italy
| | - Francesca Bianchini
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Viale G.B. Morgagni, 50, 50134, Florence, Italy
| | - Alessio Biagioni
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Viale G.B. Morgagni, 50, 50134, Florence, Italy
| | - Barbara Stecca
- Core Research Laboratory, Institute for Cancer Research and Prevention (ISPRO), Florence, Italy
| | - Lido Calorini
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, Viale G.B. Morgagni, 50, 50134, Florence, Italy. .,Center of Excellence for Research, Transfer and High Education DenoTHE University of Florence, Florence, Italy.
| |
Collapse
|
106
|
Wang G, Wang JJ, Guan R, Sun Y, Shi F, Gao J, Fu XL. Targeting Strategies for Glucose Metabolic Pathways and T Cells in Colorectal Cancer. Curr Cancer Drug Targets 2018; 19:534-550. [PMID: 30360743 DOI: 10.2174/1568009618666181015150138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 11/23/2017] [Accepted: 12/24/2017] [Indexed: 11/22/2022]
Abstract
Colorectal cancer is a heterogeneous group of diseases that result from the accumulation of different sets of genomic alterations, together with epigenomic alterations, and it is influenced by tumor-host interactions, leading to tumor cell growth and glycolytic imbalances. This review summarizes recent findings that involve multiple signaling molecules and downstream genes in the dysregulated glycolytic pathway. This paper further discusses the role of the dysregulated glycolytic pathway in the tumor initiation, progression and the concomitant systemic immunosuppression commonly observed in colorectal cancer patients. Moreover, the relationship between colorectal cancer cells and T cells, especially CD8+ T cells, is discussed, while different aspects of metabolic pathway regulation in cancer cell proliferation are comprehensively defined. Furthermore, this study elaborates on metabolism in colorectal cancer, specifically key metabolic modulators together with regulators, glycolytic enzymes, and glucose deprivation induced by tumor cells and how they inhibit T-cell glycolysis and immunogenic functions. Moreover, metabolic pathways that are integral to T cell function, differentiation, and activation are described. Selective metabolic inhibitors or immunemodulation agents targeting these pathways may be clinically useful to increase effector T cell responses for colorectal cancer treatment. However, there is a need to identify specific antigens using a cancer patient-personalized approach and combination strategies with other therapeutic agents to effectively target tumor metabolic pathways.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, 200235, Shanghai, China
| | - Jun-Jie Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, 200235, Shanghai, China
| | - Rui Guan
- Hubei University of Medicine, NO. 30 People South Road, Shiyan City, Hubei Province 442000, China
| | - Yan Sun
- Hubei University of Medicine, NO. 30 People South Road, Shiyan City, Hubei Province 442000, China
| | - Feng Shi
- Department of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province 212001, China
| | - Jing Gao
- Department of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province 212001, China
| | - Xing-Li Fu
- Department of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province 212001, China
| |
Collapse
|
107
|
McGarry T, Biniecka M, Veale DJ, Fearon U. Hypoxia, oxidative stress and inflammation. Free Radic Biol Med 2018; 125:15-24. [PMID: 29601945 DOI: 10.1016/j.freeradbiomed.2018.03.042] [Citation(s) in RCA: 376] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/20/2018] [Accepted: 03/24/2018] [Indexed: 12/20/2022]
Abstract
Inflammatory Arthritis is characterized by synovial proliferation, neovascularization and leukocyte extravasation leading to joint destruction and functional disability. Efficiency of oxygen supply to the synovium is poor due to the highly dysregulated synovial microvasculature. This along with the increased energy demands of activated infiltrating immune cells and inflamed resident cells leads to an hypoxic microenvironment and mitochondrial dysfunction. This favors an increase of reactive oxygen species, leading to oxidative damage which further promotes inflammation. In this adverse microenvironment synovial cells adapt to generate energy and switch their cell metabolism from a resting regulatory state to a highly metabolically active state which allows them to produce essential building blocks to support their proliferation. This metabolic shift results in the accumulation of metabolic intermediates which act as signaling molecules that further dictate the inflammatory response. Understanding the complex interplay between hypoxia-induced signaling pathways, oxidative stress and mitochondrial function will provide better insight into the underlying mechanisms of disease pathogenesis.
Collapse
Affiliation(s)
- Trudy McGarry
- The Department of Molecular Rheumatology, Trinity College Dublin, Ireland
| | - Monika Biniecka
- The Centre for Arthritis and Rheumatic Disease, Dublin Academic Medical Centre, St. Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Douglas J Veale
- The Centre for Arthritis and Rheumatic Disease, Dublin Academic Medical Centre, St. Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Ursula Fearon
- The Department of Molecular Rheumatology, Trinity College Dublin, Ireland.
| |
Collapse
|
108
|
Sun H, Huang Z, Sheng W, Xu MD. Emerging roles of long non-coding RNAs in tumor metabolism. J Hematol Oncol 2018; 11:106. [PMID: 30134946 PMCID: PMC6104013 DOI: 10.1186/s13045-018-0648-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 08/08/2018] [Indexed: 01/17/2023] Open
Abstract
Compared with normal cells, tumor cells display distinct metabolic characteristics. Long non-coding RNAs (lncRNAs), a large class of regulatory RNA molecules with limited or no protein-coding capacity, play key roles in tumorigenesis and progression. Recent advances have revealed that lncRNAs play a vital role in cell metabolism by regulating the reprogramming of the metabolic pathways in cancer cells. LncRNAs could regulate various metabolic enzymes that integrate cell malignant transformation and metabolic reprogramming. In addition to the known functions of lncRNAs in regulating glycolysis and glucose homeostasis, recent studies also implicate lncRNAs in amino acid and lipid metabolism. These observations reveal the high complexity of the malignant metabolism. Elucidating the metabolic-related functions of lncRNAs will provide a better understanding of the regulatory mechanisms of metabolism and thus may provide insights for the clinical development of cancer diagnostics, prognostics and therapeutics.
Collapse
Affiliation(s)
- Hui Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
| | - Mi-die Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Pathology, Tissue bank, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
| |
Collapse
|
109
|
Muir A, Danai LV, Vander Heiden MG. Microenvironmental regulation of cancer cell metabolism: implications for experimental design and translational studies. Dis Model Mech 2018; 11:dmm035758. [PMID: 30104199 PMCID: PMC6124553 DOI: 10.1242/dmm.035758] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cancers have an altered metabolism, and there is interest in understanding precisely how oncogenic transformation alters cellular metabolism and how these metabolic alterations can translate into therapeutic opportunities. Researchers are developing increasingly powerful experimental techniques to study cellular metabolism, and these techniques have allowed for the analysis of cancer cell metabolism, both in tumors and in ex vivo cancer models. These analyses show that, while factors intrinsic to cancer cells such as oncogenic mutations, alter cellular metabolism, cell-extrinsic microenvironmental factors also substantially contribute to the metabolic phenotype of cancer cells. These findings highlight that microenvironmental factors within the tumor, such as nutrient availability, physical properties of the extracellular matrix, and interactions with stromal cells, can influence the metabolic phenotype of cancer cells and might ultimately dictate the response to metabolically targeted therapies. In an effort to better understand and target cancer metabolism, this Review focuses on the experimental evidence that microenvironmental factors regulate tumor metabolism, and on the implications of these findings for choosing appropriate model systems and experimental approaches.
Collapse
Affiliation(s)
- Alexander Muir
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Laura V Danai
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Dana-Farber Cancer Institute, Boston, MA 02115, USA
| |
Collapse
|
110
|
Conrad M, Kagan VE, Bayir H, Pagnussat GC, Head B, Traber MG, Stockwell BR. Regulation of lipid peroxidation and ferroptosis in diverse species. Genes Dev 2018; 32:602-619. [PMID: 29802123 PMCID: PMC6004068 DOI: 10.1101/gad.314674.118] [Citation(s) in RCA: 351] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review by Conrad et al. reviews the functions and regulation of lipid peroxidation, ferroptosis, and the antioxidant network in diverse species, including humans, other mammals and vertebrates, plants, invertebrates, yeast, bacteria, and archaea, and discusses the potential evolutionary roles of lipid peroxidation and ferroptosis. Lipid peroxidation is the process by which oxygen combines with lipids to generate lipid hydroperoxides via intermediate formation of peroxyl radicals. Vitamin E and coenzyme Q10 react with peroxyl radicals to yield peroxides, and then these oxidized lipid species can be detoxified by glutathione and glutathione peroxidase 4 (GPX4) and other components of the cellular antioxidant defense network. Ferroptosis is a form of regulated nonapoptotic cell death involving overwhelming iron-dependent lipid peroxidation. Here, we review the functions and regulation of lipid peroxidation, ferroptosis, and the antioxidant network in diverse species, including humans, other mammals and vertebrates, plants, invertebrates, yeast, bacteria, and archaea. We also discuss the potential evolutionary roles of lipid peroxidation and ferroptosis.
Collapse
Affiliation(s)
- Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764 Neuherberg, Germany
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.,Department of Environmental Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.,Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.,Laboratory of Navigational Lipidomics of Cell Death and Regeneration, I.M. Sechenov First Moscow State Medical University, Moscow 119992, Russia
| | - Hülya Bayir
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.,Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Gabriela C Pagnussat
- Instituto de Investigaciones Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Mar del Plata, 7600 Mar del Plata, Argentina
| | - Brian Head
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon 97330.,Molecular and Cell Biology Graduate Program, Oregon State University, Corvallis, Oregon 97330, USA
| | - Maret G Traber
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon 97330.,College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon 97330, USA
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA.,Department of Chemistry, Columbia University, New York, New York 10027, USA
| |
Collapse
|
111
|
Machida K. Pluripotency Transcription Factors and Metabolic Reprogramming of Mitochondria in Tumor-Initiating Stem-like Cells. Antioxid Redox Signal 2018; 28:1080-1089. [PMID: 29256636 PMCID: PMC5865250 DOI: 10.1089/ars.2017.7241] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/15/2017] [Accepted: 12/16/2017] [Indexed: 12/26/2022]
Abstract
Significance: Neoplasms contain tumor-initiating stem-like cells (TICs) that drive malignant progression and tumor growth with drug resistance. TICs proliferate through a self-renewal process in which the two daughter cells differ in their proliferative potential, with one retaining the self-renewing phenotype and another displaying the differentiated phenotype. Recent Advances: Cancer traits (hepatocellular carcinoma) are triggered by alcoholism, obesity, and hepatitis B or C virus (HBV and HCV), including genetic changes, angiogenesis, defective tumor immunity, immortalization, metabolic reprogramming, excessive and prolonged inflammation, migration/invasion/metastasis, evasion of cell cycle arrest, anticell death, and compensatory regeneration/proliferation. Critical Issues: This review describes how metabolic reprogramming in mitochondria promotes self-renewal and oncogenicity of TICs. Pluripotency transcription factors (TFs), NANOG, OCT4, MYC, and SOX2, contribute to cancer progression by mitochondrial reprogramming, leading to the genesis of TICs and cancer. For example, oxidative phosphorylation (OXPHOS) and fatty acid metabolism are identified as major pathways contributing to pluripotency TF-mediated oncogenesis. Future Directions: Identification of novel metabolic pathways provides potential drug targets for neutralizing the activity of highly malignant TICs found in cancer patients. Antioxid. Redox Signal. 28, 1080-1089.
Collapse
Affiliation(s)
- Keigo Machida
- Department of Molecular Microbiology and Immunology, Southern California Research Center for ALPD and Cirrhosis, University of Southern California Keck School of Medicine, Los Angeles, California
| |
Collapse
|
112
|
Lister A, Bourgeois S, Imenez Silva PH, Rubio-Aliaga I, Marbet P, Walsh J, Shelton LM, Keller B, Verrey F, Devuyst O, Giesbertz P, Daniel H, Goldring CE, Copple IM, Wagner CA, Odermatt A. NRF2 regulates the glutamine transporter Slc38a3 (SNAT3) in kidney in response to metabolic acidosis. Sci Rep 2018; 8:5629. [PMID: 29618784 PMCID: PMC5884861 DOI: 10.1038/s41598-018-24000-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 03/19/2018] [Indexed: 12/28/2022] Open
Abstract
Expression of the glutamine transporter SNAT3 increases in kidney during metabolic acidosis, suggesting a role during ammoniagenesis. Microarray analysis of Nrf2 knock-out (KO) mouse kidney identified Snat3 as the most significantly down-regulated transcript compared to wild-type (WT). We hypothesized that in the absence of NRF2 the kidney would be unable to induce SNAT3 under conditions of metabolic acidosis and therefore reduce the availability of glutamine for ammoniagenesis. Metabolic acidosis was induced for 7 days in WT and Nrf2 KO mice. Nrf2 KO mice failed to induce Snat3 mRNA and protein expression during metabolic acidosis. However, there were no differences in blood pH, bicarbonate, pCO2, chloride and calcium or urinary pH, ammonium and phosphate levels. Normal induction of ammoniagenic enzymes was observed whereas several amino acid transporters showed differential regulation. Moreover, Nrf2 KO mice during acidosis showed increased expression of renal markers of oxidative stress and injury and NRF2 activity was increased during metabolic acidosis in WT kidney. We conclude that NRF2 is required to adapt the levels of SNAT3 in response to metabolic acidosis. In the absence of NRF2 and SNAT3, the kidney does not have any major acid handling defect; however, increased oxidative stress and renal injury may occur.
Collapse
Affiliation(s)
- Adam Lister
- Department of Pharmaceutical Sciences, Division of Molecular and Systems Toxicology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Soline Bourgeois
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Pedro H Imenez Silva
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Isabel Rubio-Aliaga
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Philippe Marbet
- Department of Pharmaceutical Sciences, Division of Molecular and Systems Toxicology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Joanne Walsh
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, L69 3GE, UK
| | - Luke M Shelton
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, L69 3GE, UK
| | - Bettina Keller
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Francois Verrey
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Olivier Devuyst
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Pieter Giesbertz
- Department of Biochemistry, ZIEL Research Center of Nutrition and Food Sciences, Technische Universität München, Freising, Germany
| | - Hannelore Daniel
- Department of Biochemistry, ZIEL Research Center of Nutrition and Food Sciences, Technische Universität München, Freising, Germany
| | - Christopher E Goldring
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, L69 3GE, UK
| | - Ian M Copple
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, L69 3GE, UK
| | - Carsten A Wagner
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland. .,National Center for Competence in Research Kidney.CH, Zürich, Switzerland.
| | - Alex Odermatt
- Department of Pharmaceutical Sciences, Division of Molecular and Systems Toxicology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland. .,National Center for Competence in Research Kidney.CH, Zürich, Switzerland.
| |
Collapse
|
113
|
Wright Muelas M, Ortega F, Breitling R, Bendtsen C, Westerhoff HV. Rational cell culture optimization enhances experimental reproducibility in cancer cells. Sci Rep 2018; 8:3029. [PMID: 29445172 PMCID: PMC5813001 DOI: 10.1038/s41598-018-21050-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/25/2018] [Indexed: 01/14/2023] Open
Abstract
Optimization of experimental conditions is critical in ensuring robust experimental reproducibility. Through detailed metabolomic analysis we found that cell culture conditions significantly impacted on glutaminase (GLS1) sensitivity resulting in variable sensitivity and irreproducibility in data. Baseline metabolite profiling highlighted that untreated cells underwent significant changes in metabolic status. Both the extracellular levels of glutamine and lactate and the intracellular levels of multiple metabolites changed drastically during the assay. We show that these changes compromise the robustness of the assay and make it difficult to reproduce. We discuss the implications of the cells' metabolic environment when studying the effects of perturbations to cell function by any type of inhibitor. We then devised 'metabolically rationalized standard' assay conditions, in which glutaminase-1 inhibition reduced glutamine metabolism differently in both cell lines assayed, and decreased the proliferation of one of them. The adoption of optimized conditions such as the ones described here should lead to an improvement in reproducibility and help eliminate false negatives as well as false positives in these assays.
Collapse
Affiliation(s)
- Marina Wright Muelas
- Manchester Centre for Integrative Systems Biology and Doctoral Training Centre, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK.
- Quantitative Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge, UK.
- Manchester Institute of Biotechnology, School of Chemistry, Faculty of Science and Engineering, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK.
| | - Fernando Ortega
- Manchester Pharmacy School, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - Rainer Breitling
- Manchester Institute of Biotechnology, School of Chemistry, Faculty of Science and Engineering, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Claus Bendtsen
- Quantitative Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Hans V Westerhoff
- Manchester Centre for Integrative Systems Biology and Doctoral Training Centre, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
- Netherlands Institute for Systems Biology, VU University Amsterdam and University of Amsterdam, Amsterdam, The Netherlands
- Manchester Institute of Biotechnology, School of Chemistry, Faculty of Science and Engineering, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| |
Collapse
|
114
|
Targeting of stress response pathways in the prevention and treatment of cancer. Biotechnol Adv 2018; 36:583-602. [PMID: 29339119 DOI: 10.1016/j.biotechadv.2018.01.007] [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: 08/30/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/12/2022]
Abstract
The hallmarks of tumor tissue are not only genetic aberrations but also the presence of metabolic and oxidative stress as a result of hypoxia and lactic acidosis. The stress activates several prosurvival pathways including metabolic remodeling, autophagy, antioxidant response, mitohormesis, and glutaminolysis, whose upregulation in tumors is associated with a poor survival of patients, while their activation in healthy tissue with statins, metformin, physical activity, and natural compounds prevents carcinogenesis. This review emphasizes the dual role of stress response pathways in cancer and suggests the integrative understanding as a basis for the development of rational therapy targeting the stress response.
Collapse
|
115
|
Pellegrini P, Dyczynski M, Sbrana FV, Karlgren M, Buoncervello M, Hägg-Olofsson M, Ma R, Hartman J, Bajalica-Lagercrantz S, Grander D, Kharaziha P, De Milito A. Tumor acidosis enhances cytotoxic effects and autophagy inhibition by salinomycin on cancer cell lines and cancer stem cells. Oncotarget 2018; 7:35703-35723. [PMID: 27248168 PMCID: PMC5094956 DOI: 10.18632/oncotarget.9601] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/20/2016] [Indexed: 01/07/2023] Open
Abstract
Sustained autophagy contributes to the metabolic adaptation of cancer cells to hypoxic and acidic microenvironments. Since cells in such environments are resistant to conventional cytotoxic drugs, inhibition of autophagy represents a promising therapeutic strategy in clinical oncology. We previously reported that the efficacy of hydroxychloroquine (HCQ), an autophagy inhibitor under clinical investigation is strongly impaired in acidic tumor environments, due to poor uptake of the drug, a phenomenon widely associated with drug resistance towards many weak bases. In this study we identified salinomycin (SAL) as a potent inhibitor of autophagy and cytotoxic agent effective on several cancer cell lines under conditions of transient and chronic acidosis. Since SAL has been reported to specifically target cancer-stem cells (CSC), we used an established model of breast CSC and CSC derived from breast cancer patients to examine whether this specificity may be associated with autophagy inhibition. We indeed found that CSC-like cells are more sensitive to autophagy inhibition compared to cells not expressing CSC markers. We also report that the ability of SAL to inhibit mammosphere formation from CSC-like cells was dramatically enhanced in acidic conditions. We propose that the development and use of clinically suitable SAL derivatives may result in improved autophagy inhibition in cancer cells and CSC in the acidic tumor microenvironment and lead to clinical benefits.
Collapse
Affiliation(s)
- Paola Pellegrini
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Matheus Dyczynski
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | | | - Maria Karlgren
- Department of Pharmacy and Uppsala University Drug Optimization and Pharmaceutical Profiling Platform (UDOPP) - Science for Life Laboratory, Department of Pharmacy, Uppsala Biomedical Center, Uppsala University, Sweden
| | | | - Maria Hägg-Olofsson
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Ran Ma
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Johan Hartman
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | | | - Dan Grander
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Pedram Kharaziha
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Angelo De Milito
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
116
|
Extracellular acidification induces ROS- and mPTP-mediated death in HEK293 cells. Redox Biol 2017; 15:394-404. [PMID: 29331741 PMCID: PMC5767902 DOI: 10.1016/j.redox.2017.12.018] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/27/2017] [Accepted: 12/28/2017] [Indexed: 12/20/2022] Open
Abstract
The extracellular pH (pHe) is a key determinant of the cellular (micro)environment and needs to be maintained within strict boundaries to allow normal cell function. Here we used HEK293 cells to study the effects of pHe acidification (24 h), induced by mitochondrial inhibitors (rotenone, antimycin A) and/or extracellular HCl addition. Lowering pHe from 7.2 to 5.8 reduced cell viability by 70% and was paralleled by a decrease in cytosolic pH (pHc), hyperpolarization of the mitochondrial membrane potential (Δψ), increased levels of hydroethidine-oxidizing ROS and stimulation of protein carbonylation. Co-treatment with the antioxidant α-tocopherol, the mitochondrial permeability transition pore (mPTP) desensitizer cyclosporin A and Necrostatin-1, a combined inhibitor of Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and Indoleamine 2,3-dioxygenase (IDO), prevented acidification-induced cell death. In contrast, the caspase inhibitor zVAD.fmk and the ferroptosis inhibitor Ferrostatin-1 were ineffective. We conclude that extracellular acidification induces necroptotic cell death in HEK293 cells and that the latter involves intracellular acidification, mitochondrial functional impairment, increased ROS levels, mPTP opening and protein carbonylation. These findings suggest that acidosis of the extracellular environment (as observed in mitochondrial disorders, ischemia, acute inflammation and cancer) can induce cell death via a ROS- and mPTP opening-mediated pathogenic mechanism. Extracellular acidification induces mitochondrial dysfunction. Extracellular acidification increases intracellular ROS levels. Extracellular acidification stimulates protein carbonylation. Extracellular acidification induces mPTP opening- and ROS-dependent cell death. Acidosis-induced oxidative stress likely contributes to various pathologies.
Collapse
|
117
|
Jayaraman S, Chittiboyina S, Bai Y, Abad PC, Vidi PA, Stauffacher CV, Lelièvre SA. The nuclear mitotic apparatus protein NuMA controls rDNA transcription and mediates the nucleolar stress response in a p53-independent manner. Nucleic Acids Res 2017; 45:11725-11742. [PMID: 28981686 PMCID: PMC5714241 DOI: 10.1093/nar/gkx782] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 08/30/2017] [Indexed: 12/20/2022] Open
Abstract
The nuclear mitotic apparatus protein, NuMA, is involved in major cellular events such as DNA damage response, apoptosis and p53-mediated growth-arrest, all of which are under the control of the nucleolus upon stress. Proteomic investigation has identified NuMA among hundreds of nucleolar proteins. Yet, the precise link between NuMA and nucleolar function remains undetermined. We confirm that NuMA is present in the nucleolus and reveal redistribution of NuMA upon actinomycin D or doxorubicin-induced nucleolar stress. NuMA coimmunoprecipitates with RNA polymerase I, with ribosomal proteins RPL26 and RPL24, and with components of B-WICH, an ATP-dependent chromatin remodeling complex associated with rDNA transcription. NuMA also binds to 18S and 28S rRNAs and localizes to rDNA promoter regions. Downregulation of NuMA expression triggers nucleolar stress, as shown by decreased nascent pre-rRNA synthesis, fibrillarin perinucleolar cap formation and upregulation of p27kip1, but not p53. Physiologically relevant nucleolar stress induction with reactive oxygen species reaffirms a p53-independent p27kip1 response pathway and leads to nascent pre-rRNA reduction. It also promotes the decrease in the amount of NuMA. This previously uncharacterized function of NuMA in rDNA transcription and p53-independent nucleolar stress response supports a central role for this nuclear structural protein in cellular homeostasis.
Collapse
Affiliation(s)
- Swaathi Jayaraman
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907-2026, USA.,Department of Biological Sciences, Purdue University, West Lafayette, IN 47907-2026, USA
| | - Shirisha Chittiboyina
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907-2026, USA
| | - Yunfeng Bai
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907-2026, USA
| | - Patricia C Abad
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907-2026, USA
| | - Pierre-Alexandre Vidi
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907-2026, USA
| | - Cynthia V Stauffacher
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907-2026, USA.,Center for Cancer Research, Purdue University, West Lafayette, IN 47907-2026, USA
| | - Sophie A Lelièvre
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907-2026, USA.,Center for Cancer Research, Purdue University, West Lafayette, IN 47907-2026, USA
| |
Collapse
|
118
|
Liang C, Shi S, Meng Q, Liang D, Ji S, Zhang B, Qin Y, Xu J, Ni Q, Yu X. Complex roles of the stroma in the intrinsic resistance to gemcitabine in pancreatic cancer: where we are and where we are going. Exp Mol Med 2017; 49:e406. [PMID: 29611542 PMCID: PMC5750480 DOI: 10.1038/emm.2017.255] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/23/2017] [Accepted: 08/07/2017] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the most devastating human malignancies. The poor clinical outcome in PDAC is partly attributed to a growth-permissive tumor microenvironment. In the PDAC microenvironment, the stroma is characterized by the development of extensive fibrosis, with stromal components outnumbering pancreatic cancer cells. Each of the components within the stroma has a distinct role in conferring chemoresistance to PDAC, and intrinsic chemoresistance has further worsened this pessimistic prognosis. The nucleoside analog gemcitabine (GEM) is usually the recommended first-line chemotherapeutic agent for PDAC patients and is given alone or in combination with other agents. The mechanisms of intrinsic resistance to GEM are an active area of ongoing research. This review highlights the important role the complex structure of stroma in PDAC plays in the intrinsic resistance to GEM and discusses whether antistroma therapy improves the efficacy of GEM. The addition of antistroma therapy combined with GEM is expected to be a novel therapeutic strategy with significant survival benefits for PDAC patients.
Collapse
Affiliation(s)
- Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Qingcai Meng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Dingkong Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Quanxing Ni
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| |
Collapse
|
119
|
Ramello MC, Haura EB, Abate-Daga D. CAR-T cells and combination therapies: What's next in the immunotherapy revolution? Pharmacol Res 2017; 129:194-203. [PMID: 29203440 DOI: 10.1016/j.phrs.2017.11.035] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapies are dramatically reshaping the clinical management of oncologic patients. For many of these therapies, the guidelines for administration, monitoring, and management of associated toxicities are still being established. This is especially relevant for adoptively transferred, genetically-modified T cells, which have unique pharmacokinetic properties, due to their ability to replicate and persist long-term, following a single administration. Furthermore, in the case of CAR-T cells, the use of synthetic immune receptors may impact signaling pathways involved in T cell function and survival in unexpected ways. We, herein, comment on the most salient aspects of CAR-T cell design and clinical experience in the treatment of solid tumors. In addition, we discuss different possible scenarios for combinations of CAR-T cells and other treatment modalities, with a special emphasis on kinase inhibitors, elaborating on the strategies to maximize synergism. Finally, we discuss some of the technologies that are available to explore the molecular events governing the success of these therapies. The young fields of synthetic and systems biology are likely to be major players in the advancement of CAR-T cell therapies, providing the tools and the knowledge to engineer patients' T lymphocytes into intelligent cancer-fighting micromachines.
Collapse
Affiliation(s)
- Maria C Ramello
- Dept. of Immunology, H. Lee Moffitt Cancer Center and Research Institute. Tampa, FL, United States
| | - Eric B Haura
- Dept. of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, United States
| | - Daniel Abate-Daga
- Dept. of Immunology, H. Lee Moffitt Cancer Center and Research Institute. Tampa, FL, United States; Dept. of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, United States; Dept. of Oncological Sciences, Morsani School of Medicine, University of South Florida, United States
| |
Collapse
|
120
|
Limiting Injury During Saphenous Vein Graft Preparation For Coronary Arterial Bypass Prevents Metabolic Decompensation. Sci Rep 2017; 7:14179. [PMID: 29079734 PMCID: PMC5660200 DOI: 10.1038/s41598-017-13819-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 10/02/2017] [Indexed: 02/01/2023] Open
Abstract
Standard harvest and preparation of human saphenous vein (HSV) for autologous coronary and peripheral arterial bypass procedures is associated with injury and increased oxidative stress that negatively affect graft performance. In this study we investigated the global metabolomic profiles of HSV before (unprepared; UP) and after standard vein graft preparation (AP). AP-HSV showed impaired vasomotor function that was associated with increased oxidative stress, phospholipid hydrolysis and energy depletion that are characteristic of mechanical and chemical injury. A porcine model (PSV) was utilized to validate these metabolomic changes in HSV and to determine the efficacy of an improved preparation technique (OP) using pressure-regulated distension, a non-toxic vein marker, and graft storage in buffered PlasmaLyte solution in limiting metabolic decompensation due to graft preparation. Deficits in vasomotor function and metabolic signature observed in AP-PSV could be largely mitigated with the OP procedure. These findings suggest that simple strategies aimed at reducing injury during graft harvest and preparation represents a straightforward and viable strategy to preserve conduit function and possibly improve graft patency.
Collapse
|
121
|
Personalized Nanomedicine: A Revolution at the Nanoscale. J Pers Med 2017; 7:jpm7040012. [PMID: 29023366 PMCID: PMC5748624 DOI: 10.3390/jpm7040012] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/04/2017] [Accepted: 10/04/2017] [Indexed: 12/24/2022] Open
Abstract
Nanomedicine is an interdisciplinary research field that results from the application of nanotechnology to medicine and has the potential to significantly improve some current treatments. Specifically, in the field of personalized medicine, it is expected to have a great impact in the near future due to its multiple advantages, namely its versatility to adapt a drug to a cohort of patients. In the present review, the properties and requirements of pharmaceutical dosage forms at the nanoscale, so-called nanomedicines, are been highlighted. An overview of the main current nanomedicines in pre-clinical and clinical development is presented, detailing the challenges to the personalization of these therapies. Next, the process of development of novel nanomedicines is described, from their design in research labs to their arrival on the market, including considerations for the design of nanomedicines adapted to the requirements of the market to achieve safe, effective, and quality products. Finally, attention is given to the point of view of the pharmaceutical industry, including regulation issues applied to the specific case of personalized medicine. The authors expect this review to be a useful overview of the current state of the art of nanomedicine research and industrial production, and the future opportunities of personalized medicine in the upcoming years. The authors encourage the development and marketing of novel personalized nanomedicines.
Collapse
|
122
|
Glucose-6-phosphate dehydrogenase and transketolase modulate breast cancer cell metabolic reprogramming and correlate with poor patient outcome. Oncotarget 2017; 8:106693-106706. [PMID: 29290982 PMCID: PMC5739767 DOI: 10.18632/oncotarget.21601] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 09/20/2017] [Indexed: 12/13/2022] Open
Abstract
The pentose phosphate pathway is a fundamental metabolic pathway that provides cells with ribose and NADPH required for anabolic reactions — synthesis of nucleotides and fatty acids — and maintenance of intracellular redox homeostasis. It plays a key role in tumor metabolic reprogramming and has been reported to be deregulated in different types of tumors. Herein, we silenced the most important enzymes of this pathway — glucose-6-phosphate dehydrogenase (G6PD) and transketolase (TKT) — in the human breast cancer cell line MCF7. We demonstrated that inhibition of G6PD, the oxidative branch-controlling enzyme, reduced proliferation, cell survival and increased oxidative stress. At the metabolic level, silencing of both enzymes reduced ribose synthesis. G6PD silencing in particular, augmented the glycolytic flux, reduced lipid synthesis and increased glutamine uptake, whereas silencing of TKT reduced the glycolytic flux. Importantly, we showed using breast cancer patient datasets that expression of both enzymes is positively correlated and that high expression levels of G6PD and TKT are associated with decreased overall and relapse-free survival. Altogether, our results suggest that this metabolic pathway could be subjected to therapeutic intervention to treat breast tumors and warrant further investigation.
Collapse
|
123
|
Abstract
The high metabolic demand of cancer cells leads to an accumulation of H+ ions in the tumour microenvironment. The disorganized tumour vasculature prevents an efficient wash-out of H+ ions released into the extracellular medium but also favours the development of tumour hypoxic regions associated with a shift towards glycolytic metabolism. Under hypoxia, the final balance of glycolysis, including breakdown of generated ATP, is the production of lactate and a stoichiometric amount of H+ ions. Another major source of H+ ions results from hydration of CO2 produced in the more oxidative tumour areas. All of these events occur at high rates in tumours to fulfil bioenergetic and biosynthetic needs. This Review summarizes the current understanding of how H+-generating metabolic processes segregate within tumours according to the distance from blood vessels and inversely how ambient acidosis influences tumour metabolism, reducing glycolysis while promoting mitochondrial activity. The Review also presents novel insights supporting the participation of acidosis in cancer progression via stimulation of autophagy and immunosuppression. Finally, recent advances in the different therapeutic modalities aiming to either block pH-regulatory systems or exploit acidosis will be discussed.
Collapse
Affiliation(s)
- Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 53 Avenue Mounier B1.53.09, B-1200 Brussels, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 53 Avenue Mounier B1.53.09, B-1200 Brussels, Belgium
| |
Collapse
|
124
|
Peppicelli S, Andreucci E, Ruzzolini J, Laurenzana A, Margheri F, Fibbi G, Del Rosso M, Bianchini F, Calorini L. The acidic microenvironment as a possible niche of dormant tumor cells. Cell Mol Life Sci 2017; 74:2761-2771. [PMID: 28331999 PMCID: PMC11107711 DOI: 10.1007/s00018-017-2496-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 02/01/2017] [Accepted: 02/27/2017] [Indexed: 12/31/2022]
Abstract
Although surgical excision, chemo-, and radio-therapy are clearly advanced, tumors may relapse due to cells of the so-called "minimal residual disease". Indeed, small clusters of tumor cells persist in host tissues after treatment of the primary tumor elaborating strategies to survive and escape from immunological attacks before their relapse: this variable period of remission is known as "cancer dormancy". Therefore, it is crucial to understand and consider the major concepts addressing dormancy, to identify new targets and disclose potential clinical strategies. Here, we have particularly focused the relationships between tumor microenvironment and cancer dormancy, looking at a re-appreciated aspect of this compartment that is the low extracellular pH. Accumulating evidences indicate that acidity of tumor microenvironment is associated with a poor prognosis of tumor-bearing patients, stimulates a chemo- and radio-therapy resistant phenotype, and suppresses the tumoricidal activity of cytotoxic lymphocytes and natural killer cells, and all these aspects are useful for dormancy. Therefore, this review discusses the possibility that acidity of tumor microenvironment may provide a new, not previously suggested, adequate milieu for "dormancy" of tumor cells.
Collapse
MESH Headings
- Acidosis/complications
- Acidosis/immunology
- Acidosis/pathology
- Animals
- Apoptosis
- Cell Proliferation
- Humans
- Hydrogen-Ion Concentration
- Immunologic Surveillance
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Neoplasm Recurrence, Local/etiology
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/pathology
- Neoplasm, Residual/complications
- Neoplasm, Residual/immunology
- Neoplasm, Residual/pathology
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/pathology
- Neovascularization, Pathologic/etiology
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/pathology
- Prognosis
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/pathology
- Tumor Microenvironment
Collapse
Affiliation(s)
- Silvia Peppicelli
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Elena Andreucci
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Jessica Ruzzolini
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Anna Laurenzana
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Francesca Margheri
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Gabriella Fibbi
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Mario Del Rosso
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy
- Istituto Toscano Tumori, Firenze, Italy
| | - Francesca Bianchini
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy.
- Istituto Toscano Tumori, Firenze, Italy.
| | - Lido Calorini
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università di Firenze, Viale G.B. Morgagni, 50, 50134, Firenze, Italy.
- Istituto Toscano Tumori, Firenze, Italy.
| |
Collapse
|
125
|
Pedersen SF, Novak I, Alves F, Schwab A, Pardo LA. Alternating pH landscapes shape epithelial cancer initiation and progression: Focus on pancreatic cancer. Bioessays 2017; 39. [DOI: 10.1002/bies.201600253] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Stine F. Pedersen
- Section for Cell Biology and Physiology; Department of Biology; University of Copenhagen; Copenhagen Denmark
| | - Ivana Novak
- Section for Cell Biology and Physiology; Department of Biology; University of Copenhagen; Copenhagen Denmark
| | - Frauke Alves
- Max Planck Institute of Experimental Medicine; Göttingen Germany
- Institute for Diagnostic and Interventional Radiology; University Medical Center; Göttingen Germany
- Department of Hematology and Medical Oncology; University Medical Center; Göttingen Germany
| | - Albrecht Schwab
- Institute of Physiology II; University of Münster; Münster Germany
| | - Luis A. Pardo
- Max Planck Institute of Experimental Medicine; Göttingen Germany
| |
Collapse
|
126
|
Xu P, Qiao K, Stephanopoulos G. Engineering oxidative stress defense pathways to build a robust lipid production platform in Yarrowia lipolytica. Biotechnol Bioeng 2017; 114:1521-1530. [PMID: 28295166 DOI: 10.1002/bit.26285] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/05/2017] [Accepted: 03/09/2017] [Indexed: 12/22/2022]
Abstract
Microbially derived lipids have recently attracted renewed interests due to their broad applications in production of green diesels, cosmetic additives, and oleochemicals. Metabolic engineering efforts have targeted a large portfolio of biosynthetic pathways to efficiently convert sugar to lipids in oleaginous yeast. In the engineered overproducing strains, endogenous cell metabolism typically generates harmful electrophilic molecules that compromise cell fitness and productivity. Lipids, particularly unsaturated fatty acids, are highly susceptible to oxygen radical attack and the resulting oxidative species are detrimental to cell metabolism and limit lipid productivity. In this study, we investigated cellular oxidative stress defense pathways in Yarrowia lipolytica to further improve the lipid titer, yield, and productivity. Specifically, we determined that coupling glutathione disulfide reductase and glucose-6-phosphate dehydrogenase along with aldehyde dehydrogenase are efficient solutions to combat reactive oxygen and aldehyde stress in Y. lipolytica. With the reported engineering strategies, we were able to synchronize cell growth and lipid production, improve cell fitness and morphology, and achieved industrially-relevant level of lipid titer (72.7 g/L), oil content (81.4%) and productivity (0.97 g/L/h) in controlled bench-top bioreactors. The strategies reported here represent viable steps in the development of sustainable biorefinery platforms that potentially upgrade low value carbons to high value oleochemicals and biofuels. Biotechnol. Bioeng. 2017;114: 1521-1530. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Peng Xu
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139.,Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County, Baltimore, Maryland
| | - Kangjian Qiao
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Gregory Stephanopoulos
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
127
|
Gupta S, Roy A, Dwarakanath BS. Metabolic Cooperation and Competition in the Tumor Microenvironment: Implications for Therapy. Front Oncol 2017; 7:68. [PMID: 28447025 PMCID: PMC5388702 DOI: 10.3389/fonc.2017.00068] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/24/2017] [Indexed: 12/31/2022] Open
Abstract
The tumor microenvironment (TME) is an ensemble of non-tumor cells comprising fibroblasts, cells of the immune system, and endothelial cells, besides various soluble secretory factors from all cellular components (including tumor cells). The TME forms a pro-tumorigenic cocoon around the tumor cells where reprogramming of the metabolism occurs in tumor and non-tumor cells that underlies the nature of interactions as well as competitions ensuring steady supply of nutrients and anapleoretic molecules for the tumor cells that fuels its growth even under hypoxic conditions. This metabolic reprogramming also plays a significant role in suppressing the immune attack on the tumor cells and in resistance to therapies. Thus, the metabolic cooperation and competition among the different TME components besides the inherent alterations in the tumor cells arising out of genetic as well as epigenetic changes supports growth, metastasis, and therapeutic resistance. This review focuses on the metabolic remodeling achieved through an active cooperation and competition among the three principal components of the TME—the tumor cells, the T cells, and the cancer-associated fibroblasts while discussing about the current strategies that target metabolism of TME components. Further, we will also consider the probable therapeutic opportunities targeting the various metabolic pathways as well as the signaling molecules/transcription factors regulating them for the development of novel treatment strategies for cancer.
Collapse
Affiliation(s)
- Seema Gupta
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Amrita Roy
- School of Life Sciences, B. S. Abdur Rahman Crescent University, Chennai, India
| | | |
Collapse
|
128
|
Kolosenko I, Avnet S, Baldini N, Viklund J, De Milito A. Therapeutic implications of tumor interstitial acidification. Semin Cancer Biol 2017; 43:119-133. [PMID: 28188829 DOI: 10.1016/j.semcancer.2017.01.008] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 01/25/2017] [Accepted: 01/31/2017] [Indexed: 12/12/2022]
Abstract
Interstitial acidification is a hallmark of solid tumor tissues resulting from the combination of different factors, including cellular buffering systems, defective tissue perfusion and high rates of cellular metabolism. Besides contributing to tumor pathogenesis and promoting tumor progression, tumor acidosis constitutes an important intrinsic and extrinsic mechanism modulating therapy sensitivity and drug resistance. In fact, pharmacological properties of anticancer drugs can be affected not only by tissue structure and organization but also by the distribution of the interstitial tumor pH. The acidic tumor environment is believed to create a chemical barrier that limits the effects and activity of many anticancer drugs. In this review article we will discuss the general protumorigenic effects of acidosis, the role of tumor acidosis in the modulation of therapeutic efficacy and potential strategies to overcome pH-dependent therapy-resistance.
Collapse
Affiliation(s)
- Iryna Kolosenko
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Sofia Avnet
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Nicola Baldini
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Angelo De Milito
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
129
|
Zhao M, Liu Q, Gong Y, Xu X, Zhang C, Liu X, Zhang C, Guo H, Zhang X, Gong Y, Shao C. GSH-dependent antioxidant defense contributes to the acclimation of colon cancer cells to acidic microenvironment. Cell Cycle 2017; 15:1125-33. [PMID: 26950675 PMCID: PMC4889284 DOI: 10.1080/15384101.2016.1158374] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Due to increased glycolysis and poor local perfusion, solid tumors are usually immersed in an acidic microenvironment. While extracellular acidosis is cytotoxic, cancer cells eventually become acclimated to it. While previous studies have addressed the acute effect of acidosis on cancer cells, little is known about how cancer cells survive chronic acidosis. In this study we exposed colorectal cancer (CRC) cells (HCT15, HCT116 and LoVo) to acidic pH (pH 6.5) continuously for over three months and obtained CRC cells that become acclimated to acidic pH, designated as CRC-acidosis-acclimated or CRC-AA. We unexpectedly found that while acute exposure to low pH resulted in an increase in the level of intracellular reactive oxygen species (ROS), CRC-AA cells exhibited a significantly reduced level of ROS when compared to ancestor cells. CRC-AA cells were found to maintain a higher level of reduced glutathione, via the upregulation of CD44 and glutathione reductase (GSR), among others, than their ancestor cells. Importantly, CRC-AA cells were more sensitive to agents that deplete GSH. Moreover, downregulation of GSR by RNA interference was more deleterious to CRC-AA cells than to control cells. Together, our results demonstrate a critical role of glutathione-dependent antioxidant defense in acclimation of CRC cells to acidic extracellular pH.
Collapse
Affiliation(s)
- Minnan Zhao
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Qiao Liu
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Yanchao Gong
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Xiuhua Xu
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Chen Zhang
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Xiaojie Liu
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Caibo Zhang
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Haiyang Guo
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Xiyu Zhang
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Yaoqin Gong
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Changshun Shao
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China.,b Department of Genetics/Human Genetics Institute of New Jersey , Rutgers University , Piscataway , NJ , USA
| |
Collapse
|
130
|
Katt WP, Lukey MJ, Cerione RA. A tale of two glutaminases: homologous enzymes with distinct roles in tumorigenesis. Future Med Chem 2017; 9:223-243. [PMID: 28111979 PMCID: PMC5558546 DOI: 10.4155/fmc-2016-0190] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/01/2016] [Indexed: 01/17/2023] Open
Abstract
Many cancer cells exhibit an altered metabolic phenotype, in which glutamine consumption is upregulated relative to healthy cells. This metabolic reprogramming often depends upon mitochondrial glutaminase activity, which converts glutamine to glutamate, a key precursor for biosynthetic and bioenergetic processes. Two isozymes of glutaminase exist, a kidney-type (GLS) and a liver-type enzyme (GLS2 or LGA). While a majority of studies have focused on GLS, here we summarize key findings on both glutaminases, describing their structure and function, their roles in cancer and pharmacological approaches to inhibiting their activities.
Collapse
Affiliation(s)
- William P Katt
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Michael J Lukey
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Richard A Cerione
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
131
|
Koshkin V, Ailles LE, Liu G, Krylov SN. Metabolic Suppression of a Drug-Resistant Subpopulation in Cancer Spheroid Cells. J Cell Biochem 2016; 117:59-65. [PMID: 26054050 DOI: 10.1002/jcb.25247] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 05/29/2015] [Indexed: 12/15/2022]
Abstract
Inhibition of metabolic features which distinguish cancer cells from their non-malignant counterparts is a promising approach to cancer treatment. Energy support for drug extrusion in multidrug resistance (MDR) is a potential target for metabolic inhibition. Two major sources of ATP-based metabolic energy are partial (glycolysis) and complete (mitochondrial oxidative phosphorylation) oxidation of metabolic fuels. In cancer cells, the balance between them tends to be shifted toward glycolysis; this shift is considered to be characteristic of the cancer metabolic phenotype. Numerous earlier studies, conducted with cells cultured in a monolayer (2-D model), suggested inhibition of glycolytic ATP production as an efficient tool to suppress MDR in cancer cells. Yet, more recent work challenged the appropriateness of the 2-D model for such studies and suggested that a more clinically relevant approach would utilize a more advanced cellular model such as a 3-D model. Here, we show that the transition from the 2-D model (cultured monolayer) to a 3-D model (cultured spheroids) introduces essential changes into the concept of energetic suppression of MDR. The 3-D cell organization leads to the formation of a discrete cell subpopulation (not formed in the 2-D model) with elevated MDR transport capacity. This subpopulation has a specific metabolic phenotype (mixed glycolytic/oxidative MDR support) different from that of cells cultured in the 2-D model. Finally, the shift to the oxidative phenotype becomes greater when the spheroids are grown under conditions of lactic acidosis that are typical for solid tumors. The potential clinical significance of these findings is discussed.
Collapse
Affiliation(s)
- Vasilij Koshkin
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario, Canada, M3J 1P3
| | - Laurie E Ailles
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada, N5G 1L7
| | - Geoffrey Liu
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Hospital, Toronto, Ontario, Canada, M5G 2C4
| | - Sergey N Krylov
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario, Canada, M3J 1P3
| |
Collapse
|
132
|
Chen B, Liu J, Ho TT, Ding X, Mo YY. ERK-mediated NF-κB activation through ASIC1 in response to acidosis. Oncogenesis 2016; 5:e279. [PMID: 27941930 PMCID: PMC5177778 DOI: 10.1038/oncsis.2016.81] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/30/2016] [Accepted: 11/03/2016] [Indexed: 12/16/2022] Open
Abstract
Acidic microenvironment is a common feature of solid tumors. We have previously shown that neuron specific acid-sensing ion channel 1 (ASIC1) is expressed in breast cancer, and it is responsible for acidosis-induced cellular signaling through AKT, leading to nuclear factor-κB (NF-κB) activation, and cell invasion and metastasis. However, AKT is frequently activated in cancer. Thus, a key question is whether ASIC1-mediated cell signaling still takes place in the cancer cells carrying constitutively active AKT. In the present study, we show that among four prostate cancer cell lines tested, 22Rv1 cells express the highest level of phosphorylated AKT that is not impacted by acidosis. However, acidosis can still induce NF-κB activation during which extracellular signal-regulated kinase (ERK) serves as an alternative pathway for ASIC-mediated cell signaling. Inhibition of ERK by chemical inhibitors or small interfering RNAs suppresses the acidosis-induced NF-κB activity through regulation of the inhibitory subunit IκBα phosphorylation. Furthermore, suppression of ASIC1-mediated generation of reactive oxygen species (ROS) by ROS scavengers, such as glutathione or N-acetyl-cysteine causes a decrease in ERK phosphorylation and degradation of IκBα. Finally, ASIC1 is upregulated in a subset of prostate cancer cases and ASIC1 knockout by CRISPR/Cas9 significantly suppresses cell invasion, and castration resistance both in vitro and in vivo. Together, these results support the significance of ASIC1-ROS-ERK-IκBα-NF-κB axis in prostate tumorigenesis, especially in the constitutively active AKT background.
Collapse
Affiliation(s)
- B Chen
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - J Liu
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Emergency Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - T-T Ho
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, USA
| | - X Ding
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA.,College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Y-Y Mo
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Pharmacology/Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
133
|
Liang C, Qin Y, Zhang B, Ji S, Shi S, Xu W, Liu J, Xiang J, Liang D, Hu Q, Ni Q, Xu J, Yu X. Metabolic plasticity in heterogeneous pancreatic ductal adenocarcinoma. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1866:177-188. [PMID: 27600832 DOI: 10.1016/j.bbcan.2016.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/01/2016] [Accepted: 09/02/2016] [Indexed: 01/17/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal malignant neoplasms. The recognized hallmarks of PDA are regarded to be downstream events of metabolic reprogramming. Because PDA is a heterogeneous disease that is influenced by genetic polymorphisms and changes in the microenvironment, metabolic plasticity is a novel feature of PDA. As intrinsic factors for metabolic plasticity, K-ras activation and mutations in other tumor suppressor genes induce abnormal mitochondrial metabolism and enhance glycolysis, with alterations in glutamine and lipid metabolism. As extrinsic factors, the acidic and oxygen/nutrient-deprived microenvironment also induces cancer cells to reprogram their metabolic pathway and hijack stromal cells (mainly cancer-associated fibroblasts and immunocytes) to communicate, thereby adapting to metabolic stress. Therefore, a better understanding of the metabolic features of PDA will contribute to the development of novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Wenyan Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Jinfeng Xiang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Dingkong Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Qiangsheng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Quanxing Ni
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China.
| |
Collapse
|
134
|
Is GERD a Factor in Osteonecrosis of the Jaw? Evidence of Pathology Linked to G6PD Deficiency and Sulfomucins. DISEASE MARKERS 2016; 2016:8376979. [PMID: 27773962 PMCID: PMC5059643 DOI: 10.1155/2016/8376979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/18/2016] [Accepted: 07/20/2016] [Indexed: 11/30/2022]
Abstract
Osteonecrosis of the jaw (ONJ), a rare side effect of bisphosphonate therapy, is a debilitating disorder with a poorly understood etiology. FDA's Adverse Event Reporting System (FAERS) provides the opportunity to investigate this disease. Our goals were to analyze FAERS data to discover possible relationships between ONJ and specific conditions and drugs and then to consult the scientific literature to deduce biological explanations. Our methodology revealed a very strong association between gastroesophageal reflux and bisphosphonate-induced ONJ, suggesting acidosis as a key factor. Overgrowth of acidophilic species, particularly Streptococcus mutans, in the oral microbiome in the context of insufficient acid buffering due to impaired salivary glands maintains the low pH that sustains damage to the mucosa. Significant associations between ONJ and adrenal insufficiency, vitamin C deficiency, and Sjögren's syndrome were found. Glucose 6 phosphate dehydrogenase (G6PD) deficiency can explain much of the pathology. An inability to maintain vitamin C and other antioxidants in the reduced form leads to vascular oxidative damage and impaired adrenal function. Thus, pathogen-induced acidosis, hypoxia, and insufficient antioxidant defenses together induce ONJ. G6PD deficiency and adrenal insufficiency are underlying factors. Impaired supply of adrenal-derived sulfated sterols such as DHEA sulfate may drive the disease process.
Collapse
|
135
|
The Role of Autophagy in the Maintenance of Stemness and Differentiation of Mesenchymal Stem Cells. Stem Cell Rev Rep 2016; 12:621-633. [DOI: 10.1007/s12015-016-9690-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
136
|
Gorad SS, Ellingsen C, Bathen TF, Mathiesen BS, Moestue SA, Rofstad EK. Identification of Metastasis-Associated Metabolic Profiles of Tumors by (1)H-HR-MAS-MRS. Neoplasia 2016; 17:767-75. [PMID: 26585232 PMCID: PMC4656806 DOI: 10.1016/j.neo.2015.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/25/2015] [Accepted: 10/05/2015] [Indexed: 12/30/2022] Open
Abstract
Tumors develop an abnormal microenvironment during growth, and similar to the metastatic phenotype, the metabolic phenotype of cancer cells is tightly linked to characteristics of the tumor microenvironment (TME). In this study, we explored relationships between metabolic profile, metastatic propensity, and hypoxia in experimental tumors in an attempt to identify metastasis-associated metabolic profiles. Two human melanoma xenograft lines (A-07, R-18) showing different TMEs were used as cancer models. Metabolic profile was assessed by proton high resolution magic angle spinning magnetic resonance spectroscopy (1H-HR-MAS-MRS). Tumor hypoxia was detected in immunostained histological preparations by using pimonidazole as a hypoxia marker. Twenty-four samples from 10 A-07 tumors and 28 samples from 10 R-18 tumors were analyzed. Metastasis was associated with hypoxia in both A-07 and R-18 tumors, and 1H-HR-MAS-MRS discriminated between tissue samples with and tissue samples without hypoxic regions in both models, primarily because hypoxia was associated with high lactate resonance peaks in A-07 tumors and with low lactate resonance peaks in R-18 tumors. Similarly, metastatic and non-metastatic R-18 tumors showed significantly different metabolic profiles, but not metastatic and non-metastatic A-07 tumors, probably because some samples from the metastatic A-07 tumors were derived from tumor regions without hypoxic tissue. This study suggests that 1H-HR-MAS-MRS may be a valuable tool for evaluating the role of hypoxia and lactate in tumor metastasis as well as for identification of metastasis-associated metabolic profiles.
Collapse
Affiliation(s)
- Saurabh S Gorad
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; St. Olavs University Hospital, Trondheim, Norway
| | - Christine Ellingsen
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Tone F Bathen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Berit S Mathiesen
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Siver A Moestue
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; St. Olavs University Hospital, Trondheim, Norway
| | - Einar K Rofstad
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
137
|
Corbet C, Pinto A, Martherus R, Santiago de Jesus JP, Polet F, Feron O. Acidosis Drives the Reprogramming of Fatty Acid Metabolism in Cancer Cells through Changes in Mitochondrial and Histone Acetylation. Cell Metab 2016; 24:311-23. [PMID: 27508876 DOI: 10.1016/j.cmet.2016.07.003] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 06/09/2016] [Accepted: 07/05/2016] [Indexed: 12/11/2022]
Abstract
Bioenergetic preferences of cancer cells foster tumor acidosis that in turn leads to dramatic reduction in glycolysis and glucose-derived acetyl-coenzyme A (acetyl-CoA). Here, we show that the main source of this critical two-carbon intermediate becomes fatty acid (FA) oxidation in acidic pH-adapted cancer cells. FA-derived acetyl-CoA not only fuels the tricarboxylic acid (TCA) cycle and supports tumor cell respiration under acidosis, but also contributes to non-enzymatic mitochondrial protein hyperacetylation, thereby restraining complex I activity and ROS production. Also, while oxidative metabolism of glutamine supports the canonical TCA cycle in acidic conditions, reductive carboxylation of glutamine-derived α-ketoglutarate sustains FA synthesis. Concomitance of FA oxidation and synthesis is enabled upon sirtuin-mediated histone deacetylation and consecutive downregulation of acetyl-CoA carboxylase ACC2 making mitochondrial fatty acyl-CoA degradation compatible with cytosolic lipogenesis. Perturbations of these regulatory processes lead to tumor growth inhibitory effects further identifying FA metabolism as a critical determinant of tumor cell proliferation under acidosis.
Collapse
Affiliation(s)
- Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, 53 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Adán Pinto
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, 53 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Ruben Martherus
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, 53 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - João Pedro Santiago de Jesus
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, 53 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Florence Polet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, 53 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, 53 Avenue Mounier B1.53.09, 1200 Brussels, Belgium.
| |
Collapse
|
138
|
Sanhueza C, Araos J, Naranjo L, Barros E, Subiabre M, Toledo F, Gutiérrez J, Chiarello DI, Pardo F, Leiva A, Sobrevia L. Nitric oxide and pH modulation in gynaecological cancer. J Cell Mol Med 2016; 20:2223-2230. [PMID: 27469435 PMCID: PMC5134382 DOI: 10.1111/jcmm.12921] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/05/2016] [Indexed: 01/09/2023] Open
Abstract
Nitric oxide plays several roles in cellular physiology, including control of the vascular tone and defence against pathogen infection. Neuronal, inducible and endothelial nitric oxide synthase (NOS) isoforms synthesize nitric oxide. Cells generate acid and base equivalents, whose physiological intracellular concentrations are kept due to membrane transport systems, including Na+/H+ exchangers and Na+/HCO3− transporters, thus maintaining a physiological pH at the intracellular (~7.0) and extracellular (~7.4) medium. In several pathologies, including cancer, cells are exposed to an extracellular acidic microenvironment, and the role for these membrane transport mechanisms in this phenomenon is likely. As altered NOS expression and activity is seen in cancer cells and because this gas promotes a glycolytic phenotype leading to extracellular acidosis in gynaecological cancer cells, a pro‐inflammatory microenvironment increasing inducible NOS expression in this cell type is feasible. However, whether abnormal control of intracellular and extracellular pH by cancer cells regards with their ability to synthesize or respond to nitric oxide is unknown. We, here, discuss a potential link between pH alterations, pH controlling membrane transport systems and NOS function. We propose a potential association between inducible NOS induction and Na+/H+ exchanger expression and activity in human ovary cancer. A potentiation between nitric oxide generation and the maintenance of a low extracellular pH (i.e. acidic) is proposed to establish a sequence of events in ovarian cancer cells, thus preserving a pro‐proliferative acidic tumour extracellular microenvironment. We suggest that pharmacological therapeutic targeting of Na+/H+ exchangers and inducible NOS may have benefits in human epithelial ovarian cancer.
Collapse
Affiliation(s)
- Carlos Sanhueza
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Joaquín Araos
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luciano Naranjo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eric Barros
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mario Subiabre
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernando Toledo
- Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán, Chile
| | - Jaime Gutiérrez
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Cellular Signalling and Differentiation Laboratory (CSDL), School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago, Chile
| | - Delia I Chiarello
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fabián Pardo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Andrea Leiva
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville, Spain.,University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD, Australia
| |
Collapse
|
139
|
Böhme I, Bosserhoff AK. Acidic tumor microenvironment in human melanoma. Pigment Cell Melanoma Res 2016; 29:508-23. [PMID: 27233233 DOI: 10.1111/pcmr.12495] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/25/2016] [Indexed: 12/18/2022]
Abstract
One characteristic of solid tumors such as malignant melanoma is the acidification of the tumor microenvironment. The deregulation of cancer cell metabolism is considered a main cause of extracellular acidosis. Here, cancer cells utilize aerobic glycolysis instead of oxidative phosphorylation even under normoxic conditions, as originally described by Otto Warburg. These metabolic alterations cause enhanced acid production, especially of lactate and carbon dioxide (CO2 ). The extensive production of acidic metabolites and the enhanced acid export to the extracellular space cause a consistent acidification of the tumor microenvironment, thus promoting the formation of an acid-resistant tumor cell population with increased invasive and metastatic potential. As melanoma is one of the deadliest and most metastatic forms of cancer, understanding the effects of this extracellular acidosis on human melanoma cells with distinct metastatic properties is important. The aim of this review was to summarize recent studies of the acidification of the tumor microenvironment, focusing on the specific effects of the acidic milieu on melanoma cells and to give a short overview of therapeutic approaches.
Collapse
Affiliation(s)
- Ines Böhme
- Institute of Biochemistry, Emil-Fischer-Centrum, Friedrich Alexander University Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Anja Katrin Bosserhoff
- Institute of Biochemistry, Emil-Fischer-Centrum, Friedrich Alexander University Erlangen-Nürnberg, Erlangen-Nürnberg, Germany. .,Comprehensive Cancer Center Erlangen-EMN, University of Erlangen, Erlangen, Germany.
| |
Collapse
|
140
|
Peppicelli S, Toti A, Giannoni E, Bianchini F, Margheri F, Del Rosso M, Calorini L. Metformin is also effective on lactic acidosis-exposed melanoma cells switched to oxidative phosphorylation. Cell Cycle 2016; 15:1908-18. [PMID: 27266957 DOI: 10.1080/15384101.2016.1191706] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Low extracellular pH promotes in melanoma cells a malignant phenotype characterized by an epithelial-to-mesenchymal transition (EMT) program, endowed with mesenchymal markers, high invasiveness and pro-metastatic property. Here, we demonstrate that melanoma cells exposed to an acidic extracellular microenvironment, 6.7±0.1, shift to an oxidative phosphorylation (Oxphos) metabolism. Metformin, a biguanide commonly used for type 2 diabetes, inhibited the most relevant features of acid-induced phenotype, including EMT and Oxphos. When we tested effects of lactic acidosis, to verify whether sodium lactate might have additional effects on acidic melanoma cells, we found that EMT and Oxphos also characterized lactic acid-treated cells. An increased level of motility was the only gained property of lactic acidic-exposed melanoma cells. Metformin treatment inhibited both EMT markers and Oxphos and, when its concentration raised to 10 mM, it induced a striking inhibition of proliferation and colony formation of acidic melanoma cells, both grown in protons enriched medium or lactic acidosis. Thus, our study provides the first evidence that metformin may target either proton or lactic acidosis-exposed melanoma cells inhibiting EMT and Oxphox metabolism. These findings disclose a new potential rationale of metformin addition to advanced melanoma therapy, e.g. targeting acidic cell subpopulation.
Collapse
Affiliation(s)
- Silvia Peppicelli
- a Department of Experimental and Clinical Biomedical Sciences , University of Florence, Istituto Toscano Tumori and Center of Excellence for Research, Transfer and High Education DenoTHE , Florence , Italy
| | - Alessandra Toti
- a Department of Experimental and Clinical Biomedical Sciences , University of Florence, Istituto Toscano Tumori and Center of Excellence for Research, Transfer and High Education DenoTHE , Florence , Italy
| | - Elisa Giannoni
- a Department of Experimental and Clinical Biomedical Sciences , University of Florence, Istituto Toscano Tumori and Center of Excellence for Research, Transfer and High Education DenoTHE , Florence , Italy
| | - Francesca Bianchini
- a Department of Experimental and Clinical Biomedical Sciences , University of Florence, Istituto Toscano Tumori and Center of Excellence for Research, Transfer and High Education DenoTHE , Florence , Italy
| | - Francesca Margheri
- a Department of Experimental and Clinical Biomedical Sciences , University of Florence, Istituto Toscano Tumori and Center of Excellence for Research, Transfer and High Education DenoTHE , Florence , Italy
| | - Mario Del Rosso
- a Department of Experimental and Clinical Biomedical Sciences , University of Florence, Istituto Toscano Tumori and Center of Excellence for Research, Transfer and High Education DenoTHE , Florence , Italy
| | - Lido Calorini
- a Department of Experimental and Clinical Biomedical Sciences , University of Florence, Istituto Toscano Tumori and Center of Excellence for Research, Transfer and High Education DenoTHE , Florence , Italy
| |
Collapse
|
141
|
Lho Y, le Roux CW, Park HS, Kim GS, Jung J, Hwang GS, Seo YK, Ha TK, Ha E. Changes in Glucose Metabolism in Vertical Sleeve Gastrectomy. Obes Surg 2016; 25:2002-10. [PMID: 25726321 DOI: 10.1007/s11695-015-1636-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND We evaluated metabolic changes after vertical sleeve gastrectomy (VSG) surgery in a rat model using proteomics and metabolomic profiling in liver and serum. METHODS Rats were randomly divided into two groups: sham (n = 10) and VSG (n = 12). Food intake, body weight, blood glucose, insulin, and thyroid hormone levels were measured. Two-dimensional electrophoresis, nuclear resonance spectroscopy, mass spectroscopy, immunofluorescence, and immunoblot analyses were used to determine and validate changes in metabolites and proteins in liver tissue and serum samples. RESULTS Food intake and body weight decreased after VSG group (p < 0.05 and p < 0.05, respectively). Random blood glucose (sham, 183.3 ± 5.6 mg/dL; VSG, 138.5 ± 3.7 mg/dL) decreased while random insulin (sham, 0.45 ± 0.16 μg/L; VSG, 1.05 ± 0.18 μg/L) increased after VSG (p < 0.05 and p < 0.01, respectively). We found that expressions of gluconeogenic enzymes (phosphoenolpyruvate carboxykinase-1 and glucose-6-phosphatase) and concentrations of pyruvate and malate decreased while lactate, NADH, NADPH, glucose, and AMP/ATP ratio increased after VSG. Thyroid hormones, triiodothyronine (T3) and free thyroxine (fT4), decreased after VSG. CONCLUSION This study proves that VSG suppresses hepatic glucose production.
Collapse
Affiliation(s)
- Yunmee Lho
- Department of Biochemistry, Pain Research Center, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Carel W le Roux
- Diabetes Complications Research Center, UCD Conway Institute, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Hyeon Soo Park
- Research Institute of Life Science, College of Veterinary Medicine (BK21 plus project), Kyeongsang National University, Jinju, Republic of Korea
| | - Gon Sup Kim
- Research Institute of Life Science, College of Veterinary Medicine (BK21 plus project), Kyeongsang National University, Jinju, Republic of Korea
| | - Jeeyoun Jung
- KM Health Technology Research Group, Medical Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group of Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Youn Kyoung Seo
- Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Tae Kyung Ha
- Department of Surgery, College of Medicine, Hanyang University, Seoul, Republic of Korea.
| | - Eunyoung Ha
- Department of Biochemistry, Pain Research Center, School of Medicine, Keimyung University, Daegu, Republic of Korea.
| |
Collapse
|
142
|
Dong T, Kang X, Liu Z, Zhao S, Ma W, Xuan Q, Liu H, Wang Z, Zhang Q. Altered glycometabolism affects both clinical features and prognosis of triple-negative and neoadjuvant chemotherapy-treated breast cancer. Tumour Biol 2016; 37:8159-68. [PMID: 26715276 DOI: 10.1007/s13277-015-4729-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/21/2015] [Indexed: 12/31/2022] Open
Abstract
Glycometabolism is a distinctive aspect of energy metabolism in breast cancer, and key glycometabolism enzymes/pathways (glycolysis, hexosamine biosynthetic pathway, and pentose phosphate pathway) may directly or indirectly affect the clinical features. In this study, we analyzed the particular correlation between the altered glycometabolism and clinical features of breast cancer to instruct research and clinical treatment. Tissue microarrays containing 189 hollow needle aspiration samples and 295 triple-negative breast cancer tissues were used to test the expression of M2 isoform of pyruvate kinase (PKM2), glutamine-fructose-6-phosphate transaminase 1 (GFPT1), glucose-6-phosphate dehydrogenase (G6PD), and p53 by immunohistochemistry and the intensity of these glycometabolism-related protein was evaluated. Chi-square test, Kaplan-Meier estimates, and Cox proportional hazards model were used to analyze the relationship between the expression of these factors and major clinical features. PKM2, GFPT1, and G6PD affect the pathologic complete response rate of neoadjuvant chemotherapy patients in different ways; pyruvate kinase muscle isozyme 2 (PKM2) and G6PD are closely associated with the molecular subtypes, whereas GFPT1 is correlated with cancer size. All these three factors as well as p53 have impacts on the progression-free survival and overall survival of triple-negative breast cancer patients. Cancer size shows significant association with PKM2 and GFPT1 expression, while the pN stage and grade are associated with PKM2 and G6PD expression. Our study support that clinical characteristics are reflections of specific glycometabolism pathways, so their relationships may shed light on the orientation of research or clinical treatment. The expression of PKM2, GFPT1, and G6PD are hazardous factors for prognosis: high expression of these proteins predict worse progression-free survival and overall survival in triple-negative breast cancer, as well as worse pathologic complete response rate in neoadjuvant chemotherapy breast cancer. However, p53 appears as a protective factor only in the patients receiving neoadjuvant chemotherapy. All the four proteins, PKM2, GFPT1, G6PD and p53, are prognostic markers of breast cancer. The correlation among them suggests that there may be crosstalk of the four proteins in breast cancer.
Collapse
Affiliation(s)
- Tieying Dong
- Department of Internal Medicine, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Xinmei Kang
- Department of Internal Medicine, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Zhaoliang Liu
- Cancer Research Institute, Harbin Medical University, Harbin, China
- Cancer Research Institute of Heilongjiang, Harbin, China
| | - Shu Zhao
- Department of Internal Medicine, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Wenjie Ma
- Department of Internal Medicine, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Qijia Xuan
- Department of Internal Medicine, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Hang Liu
- Department of Internal Medicine, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Zhipeng Wang
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qingyuan Zhang
- Department of Internal Medicine, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150081, China.
- Cancer Research Institute of Heilongjiang, Harbin, China.
| |
Collapse
|
143
|
Iizumi T, Takahashi S, Mashima K, Minami K, Izawa Y, Abe T, Hishiki T, Suematsu M, Kajimura M, Suzuki N. A possible role of microglia-derived nitric oxide by lipopolysaccharide in activation of astroglial pentose-phosphate pathway via the Keap1/Nrf2 system. J Neuroinflammation 2016; 13:99. [PMID: 27143001 PMCID: PMC4855896 DOI: 10.1186/s12974-016-0564-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/26/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Toll-like receptor 4 (TLR4) plays a pivotal role in the pathophysiology of stroke-induced inflammation. Both astroglia and microglia express TLR4, and endogenous ligands produced in the ischemic brain induce inflammatory responses. Reactive oxygen species (ROS), nitric oxide (NO), and inflammatory cytokines produced by TLR4 activation play harmful roles in neuronal damage after stroke. Although astroglia exhibit pro-inflammatory responses upon TLR4 stimulation by lipopolysaccharide (LPS), they may also play cytoprotective roles via the activation of the pentose phosphate pathway (PPP), reducing oxidative stress by glutathione peroxidase. We investigated the mechanisms by which astroglia reduce oxidative stress via the activation of PPP, using TLR4 stimulation and hypoxia in concert with microglia. METHODS In vitro experiments were performed using cells prepared from Sprague-Dawley rats. Coexisting microglia in the astroglial culture were chemically eliminated using L-leucine methyl ester (LME). Cells were exposed to LPS (0.01 μg/mL) or hypoxia (1 % O2) for 12-15 h. PPP activity was measured using [1-(14)C]glucose and [6-(14)C]glucose. ROS and NO production were measured using 2',7'-dichlorodihydrofluorescein diacetate and diaminofluorescein-FM diacetate, respectively. The involvement of nuclear factor-erythroid-2-related factor 2 (Nrf2), a cardinal transcriptional factor under stress conditions that regulates glucose 6-phosphate dehydrogenase, the rate-limiting enzyme of PPP, was evaluated using immunohistochemistry. RESULTS Cultured astroglia exposed to LPS elicited 20 % increases in PPP flux, and these actions of astroglia appeared to involve Nrf2. However, the chemical depletion of coexisting microglia eliminated both increases in PPP and astroglial nuclear translocation of Nrf2. LPS induced ROS and NO production in the astroglial culture containing microglia but not in the microglia-depleted astroglial culture. LPS enhanced astroglial ROS production after glutathione depletion. U0126, an upstream inhibitor of mitogen-activated protein kinase, eliminated LPS-induced NO production, whereas ROS production was unaffected. U0126 also eliminated LPS-induced PPP activation in astroglial-microglial culture, indicating that microglia-derived NO mediated astroglial PPP activation. Hypoxia induced astroglial PPP activation independent of the microglia-NO pathway. Elimination of ROS and NO production by sulforaphane, a natural Nrf2 activator, confirmed the astroglial protective mechanism. CONCLUSIONS Astroglia in concert with microglia may play a cytoprotective role for countering oxidative stress in stroke.
Collapse
Affiliation(s)
- Takuya Iizumi
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Shinichi Takahashi
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan.
| | - Kyoko Mashima
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Kazushi Minami
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Yoshikane Izawa
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Takato Abe
- Department of Neurology, Osaka City University Graduate School of Medicine, Osaka-shi, 545-8585, Osaka , Japan
| | - Takako Hishiki
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, 160-8582, Tokyo, Japan.,Clinical and Translational Research Center, Keio University School of Medicine, Shinjuku-ku, 160-8582, Tokyo, Japan.,JST Exploratory Research for Advanced Technology (ERATO) Suematsu Gas Biology Project, Shinjuku-ku, 160-8582, Tokyo , Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, 160-8582, Tokyo, Japan.,JST Exploratory Research for Advanced Technology (ERATO) Suematsu Gas Biology Project, Shinjuku-ku, 160-8582, Tokyo , Japan
| | - Mayumi Kajimura
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, 160-8582, Tokyo, Japan.,JST Exploratory Research for Advanced Technology (ERATO) Suematsu Gas Biology Project, Shinjuku-ku, 160-8582, Tokyo , Japan
| | - Norihiro Suzuki
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| |
Collapse
|
144
|
Allen E, Miéville P, Warren CM, Saghafinia S, Li L, Peng MW, Hanahan D. Metabolic Symbiosis Enables Adaptive Resistance to Anti-angiogenic Therapy that Is Dependent on mTOR Signaling. Cell Rep 2016; 15:1144-60. [PMID: 27134166 PMCID: PMC4872464 DOI: 10.1016/j.celrep.2016.04.029] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 01/28/2016] [Accepted: 04/07/2016] [Indexed: 01/01/2023] Open
Abstract
Therapeutic targeting of tumor angiogenesis with VEGF inhibitors results in demonstrable, but transitory efficacy in certain human tumors and mouse models of cancer, limited by unconventional forms of adaptive/evasive resistance. In one such mouse model, potent angiogenesis inhibitors elicit compartmental reorganization of cancer cells around remaining blood vessels. The glucose and lactate transporters GLUT1 and MCT4 are induced in distal hypoxic cells in a HIF1α-dependent fashion, indicative of glycolysis. Tumor cells proximal to blood vessels instead express the lactate transporter MCT1, and p-S6, the latter reflecting mTOR signaling. Normoxic cancer cells import and metabolize lactate, resulting in upregulation of mTOR signaling via glutamine metabolism enhanced by lactate catabolism. Thus, metabolic symbiosis is established in the face of angiogenesis inhibition, whereby hypoxic cancer cells import glucose and export lactate, while normoxic cells import and catabolize lactate. mTOR signaling inhibition disrupts this metabolic symbiosis, associated with upregulation of the glucose transporter GLUT2. Angiogenesis inhibitors causing acute hypoxia elicit metabolic compartmentalization Hypoxic cancer cells import and metabolize glucose, secreting lactate Normoxic vessel-proximal cancer cells import and metabolize lactate, involving mTOR Co-inhibiting mTOR disrupts the metabolic symbiosis
Collapse
Affiliation(s)
- Elizabeth Allen
- The Swiss Institute for Experimental Cancer Research (ISREC), EPFL SV ISREC, Station 19, 1015 Lausanne, Switzerland
| | - Pascal Miéville
- The Institute of Chemical Sciences and Engineering (ISIC-SB-EPFL), Ecole Polytechnique Fédérale de Lausanne, EPFL SB ISIC-Direction, CH A3 398 Station 6, 1015 Lausanne, Switzerland
| | - Carmen M Warren
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA 90095, USA
| | - Sadegh Saghafinia
- The Swiss Institute for Experimental Cancer Research (ISREC), EPFL SV ISREC, Station 19, 1015 Lausanne, Switzerland
| | - Leanne Li
- The Swiss Institute for Experimental Cancer Research (ISREC), EPFL SV ISREC, Station 19, 1015 Lausanne, Switzerland
| | - Mei-Wen Peng
- The Swiss Institute for Experimental Cancer Research (ISREC), EPFL SV ISREC, Station 19, 1015 Lausanne, Switzerland
| | - Douglas Hanahan
- The Swiss Institute for Experimental Cancer Research (ISREC), EPFL SV ISREC, Station 19, 1015 Lausanne, Switzerland; The Swiss Cancer Center Lausanne (SCCL), Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
145
|
Chano T, Avnet S, Kusuzaki K, Bonuccelli G, Sonveaux P, Rotili D, Mai A, Baldini N. Tumour-specific metabolic adaptation to acidosis is coupled to epigenetic stability in osteosarcoma cells. Am J Cancer Res 2016; 6:859-875. [PMID: 27186436 PMCID: PMC4859889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 03/10/2016] [Indexed: 06/05/2023] Open
Abstract
The glycolytic-based metabolism of cancers promotes an acidic microenvironment that is responsible for increased aggressiveness. However, the effects of acidosis on tumour metabolism have been almost unexplored. By using capillary electrophoresis with time-of-flight mass spectrometry, we observed a significant metabolic difference associated with glycolysis repression (dihydroxyacetone phosphate), increase of amino acid catabolism (phosphocreatine and glutamate) and urea cycle enhancement (arginino succinic acid) in osteosarcoma (OS) cells compared with normal fibroblasts. Noteworthy, metabolites associated with chromatin modification, like UDP-glucose and N(8)-acetylspermidine, decreased more in OS cells than in fibroblasts. COBRA assay and acetyl-H3 immunoblotting indicated an epigenetic stability in OS cells than in normal cells, and OS cells were more sensitive to an HDAC inhibitor under acidosis than under neutral pH. Since our data suggest that acidosis promotes a metabolic reprogramming that can contribute to the epigenetic maintenance under acidosis only in tumour cells, the acidic microenvironment should be considered for future therapies.
Collapse
Affiliation(s)
- Tokuhiro Chano
- Department of Clinical Laboratory Medicine, Shiga University of Medical ScienceOtsu, Shiga, Japan
| | - Sofia Avnet
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico RizzoliBologna, Italy
| | - Katsuyuki Kusuzaki
- Department of Musculoskeletal Oncology, Takai HospitalTennri, Nara, Japan
| | - Gloria Bonuccelli
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico RizzoliBologna, Italy
| | - Pierre Sonveaux
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology (FATH), Université Catholique de Louvain (UCL)Brussels, Belgium
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of RomaRoma, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of RomaRoma, Italy
- Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of RomaRoma, Italy
| | - Nicola Baldini
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico RizzoliBologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of BolognaBologna, Italy
| |
Collapse
|
146
|
Estrela JM, Ortega A, Mena S, Sirerol JA, Obrador E. Glutathione in metastases: From mechanisms to clinical applications. Crit Rev Clin Lab Sci 2016; 53:253-67. [DOI: 10.3109/10408363.2015.1136259] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- José M. Estrela
- Department of Physiology, Faculty of Medicine and Odontology and
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Angel Ortega
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Salvador Mena
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - J. Antoni Sirerol
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Elena Obrador
- Department of Physiology, Faculty of Medicine and Odontology and
| |
Collapse
|
147
|
Tang X, Wu J, Ding CK, Lu M, Keenan MM, Lin CC, Lin CA, Wang CC, George D, Hsu DS, Chi JT. Cystine Deprivation Triggers Programmed Necrosis in VHL-Deficient Renal Cell Carcinomas. Cancer Res 2016; 76:1892-903. [PMID: 26833124 DOI: 10.1158/0008-5472.can-15-2328] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 01/14/2016] [Indexed: 12/27/2022]
Abstract
Oncogenic transformation may reprogram tumor metabolism and render cancer cells addicted to extracellular nutrients. Deprivation of these nutrients may therefore represent a therapeutic opportunity, but predicting which nutrients cancer cells become addicted remains difficult. Here, we performed a nutrigenetic screen to determine the phenotypes of isogenic pairs of clear cell renal cancer cells (ccRCC), with or without VHL, upon the deprivation of individual amino acids. We found that cystine deprivation triggered rapid programmed necrosis in VHL-deficient cell lines and primary ccRCC tumor cells, but not in VHL-restored counterparts. Blocking cystine uptake significantly delayed xenograft growth of ccRCC. Importantly, cystine deprivation triggered similar metabolic changes regardless of VHL status, suggesting that metabolic responses alone are not sufficient to explain the observed distinct fates of VHL-deficient and restored cells. Instead, we found that increased levels of TNFα associated with VHL loss forced VHL-deficient cells to rely on intact RIPK1 to inhibit apoptosis. However, the preexisting elevation in TNFα expression rendered VHL-deficient cells susceptible to necrosis triggered by cystine deprivation. We further determined that reciprocal amplification of the Src-p38 (MAPK14)-Noxa (PMAIP1) signaling and TNFα-RIP1/3 (RIPK1/RIPK3)-MLKL necrosis pathways potentiated cystine-deprived necrosis. Together, our findings reveal that cystine deprivation in VHL-deficient RCCs presents an attractive therapeutic opportunity that may bypass the apoptosis-evading mechanisms characteristic of drug-resistant tumor cells. Cancer Res; 76(7); 1892-903. ©2016 AACR.
Collapse
Affiliation(s)
- Xiaohu Tang
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina. Center for Genomic and Computational Biology Duke University, Durham, North Carolina
| | - Jianli Wu
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina. Center for Genomic and Computational Biology Duke University, Durham, North Carolina
| | - Chien-Kuang Ding
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina. Center for Genomic and Computational Biology Duke University, Durham, North Carolina
| | - Min Lu
- Center for Genomic and Computational Biology Duke University, Durham, North Carolina. Department of Medicine, Duke University, Durham, North Carolina
| | - Melissa M Keenan
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina. Center for Genomic and Computational Biology Duke University, Durham, North Carolina
| | - Chao-Chieh Lin
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina. Center for Genomic and Computational Biology Duke University, Durham, North Carolina
| | - Chih-An Lin
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina. Center for Genomic and Computational Biology Duke University, Durham, North Carolina
| | - Charles C Wang
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina. Center for Genomic and Computational Biology Duke University, Durham, North Carolina
| | - Daniel George
- Department of Medicine, Duke University, Durham, North Carolina
| | - David S Hsu
- Center for Genomic and Computational Biology Duke University, Durham, North Carolina. Department of Medicine, Duke University, Durham, North Carolina
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina. Center for Genomic and Computational Biology Duke University, Durham, North Carolina.
| |
Collapse
|
148
|
Boros LG, D'Agostino DP, Katz HE, Roth JP, Meuillet EJ, Somlyai G. Submolecular regulation of cell transformation by deuterium depleting water exchange reactions in the tricarboxylic acid substrate cycle. Med Hypotheses 2016; 87:69-74. [PMID: 26826644 PMCID: PMC4733494 DOI: 10.1016/j.mehy.2015.11.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/23/2015] [Indexed: 02/08/2023]
Abstract
The naturally occurring isotope of hydrogen ((1)H), deuterium ((2)H), could have an important biological role. Deuterium depleted water delays tumor progression in mice, dogs, cats and humans. Hydratase enzymes of the tricarboxylic acid (TCA) cycle control cell growth and deplete deuterium from redox cofactors, fatty acids and DNA, which undergo hydride ion and hydrogen atom transfer reactions. A model is proposed that emphasizes the terminal complex of mitochondrial electron transport chain reducing molecular oxygen to deuterium depleted water (DDW); this affects gluconeogenesis as well as fatty acid oxidation. In the former, the DDW is thought to diminish the deuteration of sugar-phosphates in the DNA backbone, helping to preserve stability of hydrogen bond networks, possibly protecting against aneuploidy and resisting strand breaks, occurring upon exposure to radiation and certain anticancer chemotherapeutics. DDW is proposed here to link cancer prevention and treatment using natural ketogenic diets, low deuterium drinking water, as well as DDW production as the mitochondrial downstream mechanism of targeted anti-cancer drugs such as Avastin and Glivec. The role of (2)H in biology is a potential missing link to the elusive cancer puzzle seemingly correlated with cancer epidemiology in western populations as a result of excessive (2)H loading from processed carbohydrate intake in place of natural fat consumption.
Collapse
Affiliation(s)
- László G Boros
- Department of Pediatrics, UCLA School of Medicine Harbor-UCLA Medical Center, Torrance, CA, USA; The Los Angeles Biomedical Research Institute (LABiOMED), Torrance, CA, USA; SIDMAP, LLC, Los Angeles, CA, USA.
| | - Dominic P D'Agostino
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, Hyperbaric Biomedical Research Laboratory, University of South Florida, Tampa, FL, USA
| | - Howard E Katz
- Department of Materials Science & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Justine P Roth
- Department of Chemistry, Johns Hopkins University, Baltimore, MD, USA
| | - Emmanuelle J Meuillet
- The University of Arizona Comprehensive Cancer Center, The University of Arizona, Tucson, AZ, USA; Department of Nutritional Sciences, The University of Arizona, Tucson, AZ, USA
| | - Gábor Somlyai
- HYD, LLC for Cancer Research & Drug Development, Budapest, Hungary
| |
Collapse
|
149
|
Abstract
Tumor metabolism is significantly altered to support the various metabolic needs of tumor cells. The most prominent change is the increased tumor glycolysis that leads to increased glucose uptake and utilization. However, it has become obvious that many non-glucose nutrients, such as amino acids, lactate, acetate, and macromolecules, can serve as alternative fuels for cancer cells. This knowledge reveals an unexpected flexibility and evolutionarily conserved model in which cancer cells uptake nutrients from their external environment to fulfill their necessary energetic needs. Tumor cells may have evolved the ability to utilize different carbon sources because of the limited supply of nutrients in their microenvironment, which can be driven by oncogenic mutations or tumor microenvironmental stresses. In certain cases, these factors permanently alter the tumor cells' metabolism, causing certain nutrients to become indispensable and thus creating opportunities for therapeutic intervention to eradicate tumors by their metabolic vulnerabilities.
Collapse
|
150
|
Bai H, Chen G, Fang C, Yang X, Yu S, Hai C. Osteosarcoma cell proliferation and migration are partly regulated by redox-activated NHE-1. J Clin Transl Res 2015; 1:168-179. [PMID: 30873452 PMCID: PMC6410620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/30/2015] [Accepted: 12/04/2015] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Osteosarcoma (OS) is the most common primary malignant bone tumor in children and adolescents. OS is associated with locally aggressive growth and high metastatic potential. The mechanisms that underlie these processes are currently elusive. Reactive oxygen species (ROS) and Na+/H+ exchanger 1 (NHE1) have been suggested to regulate proliferation and migration of tumor cells. However, the relationship between NHE1 and ROS in OS proliferation and migration has not been investigated before. AIM To investigate the role of NHE1 and ROS in the proliferation and migration of OS. METHODS ROS levels and NHE1 expression were studied in cultured human OS cells and human OS xenografts in nude mice. In vitro, OS cells were treated with different doses of tert-butyl hydroperoxide (tBHP), a ROS inducer, and cariporide, an NHE1 inhibitor, to study the effect on cell proliferation and migration. In vivo, nude mice bearing OS cells were administrated with NHE1 inhibitor or antioxidant and the tumor weights were measured. RESULTS This study reported for the first time that the expression of NHE1 and intracellular ROS level were both increased in OS tissues and cells. Exposure of OS cell to ROS derived from tBHP was able to accelerate cell proliferation and migration and also up-regulate NHE1 protein expression. Moreover, tBHP significantly increased intracellular pH (pHi), decreased extracellular pH (pHe) and induced upregulation of ERK, MMP2, and MMP9. Lowering of ROS levels with the anti-oxidant DMTU or inhibiting NHE1 activity via cariporide abolished the stimulatory effect of tBHP. However, there cariporide did not affect intracellular ROS levels. In vivo study we further confirmed that cariporide could inhibit tumor growth in the nude mouse xenografts of OS cells. CONCLUSIONS The data demonstrate that up-regulation of NHE1 was induced by low concentrations of ROS contributes to the regulation of tumor proliferation and invasion of OS. RELEVANCE FOR PATIENTS There is potential application for cariporide as an effective antitumor agent during the development of human osteosarcoma. In addition, redox modulation on proton transport may represent a novel target of osteosarcoma prevention, and open a new avenues for future research.
Collapse
Affiliation(s)
- Hua Bai
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Guojing Chen
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Congwen Fang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xuekang Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Sixun Yu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Chunxu Hai
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|