1
|
Hu SY, Lin TH, Chen CY, He YH, Huang WC, Hsieh CY, Chen YH, Chang WC. Stephania tetrandra and Its Active Compound Coclaurine Sensitize NSCLC Cells to Cisplatin through EFHD2 Inhibition. Pharmaceuticals (Basel) 2024; 17:1356. [PMID: 39458997 PMCID: PMC11510146 DOI: 10.3390/ph17101356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/19/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Adjuvant chemotherapy, particularly cisplatin, is recommended for non-small cell lung carcinoma (NSCLC) patients at high risk of recurrence. EF-hand domain-containing protein D2 (EFHD2) has been recently shown to increase cisplatin resistance and is significantly associated with recurrence in early-stage NSCLC patients. Natural products, commonly used as phytonutrients, are also recognized for their potential as pharmaceutical anticancer agents. RESULT In this study, a range of Chinese herbs known for their antitumor or chemotherapy-enhancing properties were evaluated for their ability to inhibit EFHD2 expression in NSCLC cells. Among the herbs tested, Stephania tetrandra (S. tetrandra) exhibited the highest efficacy in inhibiting EFHD2 and sensitizing cells to cisplatin. Through LC-MS identification and functional assays, coclaurine was identified as a key molecule in S. tetrandra responsible for EFHD2 inhibition. Coclaurine not only downregulated EFHD2-related NOX4-ABCC1 signaling and enhanced cisplatin sensitivity, but also suppressed the stemness and metastatic properties of NSCLC cells. Mechanistically, coclaurine disrupted the interaction between the transcription factor FOXG1 and the EFHD2 promoter, leading to a reduction in EFHD2 transcription. Silencing FOXG1 further inhibited EFHD2 expression and sensitized NSCLC cells to cisplatin. CONCLUSIONS S. tetrandra and its active compound coclaurine may serve as effective adjuvant therapies to improve cisplatin efficacy in the treatment of NSCLC.
Collapse
Affiliation(s)
- Shu-Yu Hu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan; (S.-Y.H.); (Y.-H.H.); (W.-C.H.)
| | - Tsai-Hui Lin
- Department of Chinese Medicine, China Medical University Hospital, Taichung 404327, Taiwan;
| | - Chung-Yu Chen
- Research Center for Cancer Biology, China Medical University, Taichung 406040, Taiwan;
| | - Yu-Hao He
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan; (S.-Y.H.); (Y.-H.H.); (W.-C.H.)
- Center for Molecular Medicine, China Medical University Hospital, Taichung 406040, Taiwan
- Program for Cancer Biology and Drug Discovery, China Medical University, Taichung 404333, Taiwan
| | - Wei-Chien Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan; (S.-Y.H.); (Y.-H.H.); (W.-C.H.)
- Center for Molecular Medicine, China Medical University Hospital, Taichung 406040, Taiwan
- Program for Cancer Biology and Drug Discovery, China Medical University, Taichung 404333, Taiwan
- School of Pharmacy, China Medical University, Taichung 404333, Taiwan
| | - Ching-Yun Hsieh
- Division of Hematology and Oncology, Department of internal medicine, China Medical University Hospital, Taichung 404327, Taiwan;
| | - Ya-Huey Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan; (S.-Y.H.); (Y.-H.H.); (W.-C.H.)
- Center for Molecular Medicine, China Medical University Hospital, Taichung 406040, Taiwan
- Program for Cancer Biology and Drug Discovery, China Medical University, Taichung 404333, Taiwan
| | - Wei-Chao Chang
- Center for Molecular Medicine, China Medical University Hospital, Taichung 406040, Taiwan
| |
Collapse
|
2
|
Soltani M, Zhao Y, Xia Z, Ganjalikhani Hakemi M, Bazhin AV. The Importance of Cellular Metabolic Pathways in Pathogenesis and Selective Treatments of Hematological Malignancies. Front Oncol 2021; 11:767026. [PMID: 34868994 PMCID: PMC8636012 DOI: 10.3389/fonc.2021.767026] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/20/2021] [Indexed: 02/05/2023] Open
Abstract
Despite recent advancements in the treatment of hematologic malignancies and the emergence of newer and more sophisticated therapeutic approaches such as immunotherapy, long-term overall survival remains unsatisfactory. Metabolic alteration, as an important hallmark of cancer cells, not only contributes to the malignant transformation of cells, but also promotes tumor progression and metastasis. As an immune-escape mechanism, the metabolic adaptation of the bone marrow microenvironment and leukemic cells is a major player in the suppression of anti-leukemia immune responses. Therefore, metabolic rewiring in leukemia would provide promising opportunities for newer therapeutic interventions. Several therapeutic agents which affect essential bioenergetic pathways in cancer cells including glycolysis, β-oxidation of fatty acids and Krebs cycle, or anabolic pathways such as lipid biosynthesis and pentose phosphate pathway, are being tested in various types of cancers. So far, numerous preclinical or clinical trial studies using such metabolic agents alone or in combination with other remedies such as immunotherapy are in progress and have demonstrated promising outcomes. In this review, we aim to argue the importance of metabolic alterations and bioenergetic pathways in different types of leukemia and their vital roles in disease development. Designing treatments based on targeting leukemic cells vulnerabilities, particularly in nonresponsive leukemia patients, should be warranted.
Collapse
Affiliation(s)
- Mojdeh Soltani
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yue Zhao
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | - Alexandr V. Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|
3
|
Mollinedo F, Gajate C. Direct Endoplasmic Reticulum Targeting by the Selective Alkylphospholipid Analog and Antitumor Ether Lipid Edelfosine as a Therapeutic Approach in Pancreatic Cancer. Cancers (Basel) 2021; 13:4173. [PMID: 34439330 PMCID: PMC8394177 DOI: 10.3390/cancers13164173] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/11/2021] [Accepted: 08/15/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common malignancy of the pancreas, shows a dismal and grim overall prognosis and survival rate, which have remained virtually unchanged for over half a century. PDAC is the most lethal of all cancers, with the highest mortality-to-incidence ratio. PDAC responds poorly to current therapies and remains an incurable malignancy. Therefore, novel therapeutic targets and drugs are urgently needed for pancreatic cancer treatment. Selective induction of apoptosis in cancer cells is an appealing approach in cancer therapy. Apoptotic cell death is highly regulated by different signaling routes that involve a variety of subcellular organelles. Endoplasmic reticulum (ER) stress acts as a double-edged sword at the interface of cell survival and death. Pancreatic cells exhibit high hormone and enzyme secretory functions, and thereby show a highly developed ER. Thus, pancreatic cancer cells display a prominent ER. Solid tumors have to cope with adverse situations in which hypoxia, lack of certain nutrients, and the action of certain antitumor agents lead to a complex interplay and crosstalk between ER stress and autophagy-the latter acting as an adaptive survival response. ER stress also mediates cell death induced by a number of anticancer drugs and experimental conditions, highlighting the pivotal role of ER stress in modulating cell fate. The alkylphospholipid analog prototype edelfosine is selectively taken up by tumor cells, accumulates in the ER of a number of human solid tumor cells-including pancreatic cancer cells-and promotes apoptosis through a persistent ER-stress-mediated mechanism both in vitro and in vivo. Here, we discuss and propose that direct ER targeting may be a promising approach in the therapy of pancreatic cancer, opening up a new avenue for the treatment of this currently incurable and deadly cancer. Furthermore, because autophagy acts as a cytoprotective response to ER stress, potentiation of the triggering of a persistent ER response by combination therapy, together with the use of autophagy blockers, could improve the current gloomy expectations for finding a cure for this type of cancer.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, C/Ramiro de Maeztu 9, E-28040 Madrid, Spain;
| | | |
Collapse
|
4
|
Kumari N, Bansal S. Arginine depriving enzymes: applications as emerging therapeutics in cancer treatment. Cancer Chemother Pharmacol 2021; 88:565-594. [PMID: 34309734 DOI: 10.1007/s00280-021-04335-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022]
Abstract
Cancer is the second leading cause of death globally. Chemotherapy and radiation therapy and other medications are employed to treat various types of cancer. However, each treatment has its own set of side effects, owing to its low specificity. As a result, there is an urgent need for newer therapeutics that do not disrupt healthy cells' normal functioning. Depriving nutrient or non/semi-essential amino acids to which cancerous cells are auxotrophic remains one such promising anticancer strategy. L-Arginine (Arg) is a semi-essential vital amino acid involved in versatile metabolic processes, signaling pathways, and cancer cell proliferation. Hence, the administration of Arg depriving enzymes (ADE) such as arginase, arginine decarboxylase (ADC), and arginine deiminase (ADI) could be effective in cancer therapy. The Arg auxotrophic cancerous cells like hepatocellular carcinoma, human colon cancer, leukemia, and breast cancer cells are sensitive to ADE treatment due to low expression of crucial enzymes argininosuccinate synthetase (ASS), argininosuccinate lyase (ASL), and ornithine transcarbamylase (OCT). These therapeutic enzyme treatments induce cell death through inducing autophagy, apoptosis, generation of oxidative species, i.e., oxidative stress, and arresting the progression and expansion of cancerous cells at certain cell cycle checkpoints. The enzymes are undergoing clinical trials and could be successfully exploited as potential anticancer agents in the future.
Collapse
Affiliation(s)
- Neha Kumari
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology Waknaghat, Solan, 173234, Himachal Pradesh, India
| | - Saurabh Bansal
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology Waknaghat, Solan, 173234, Himachal Pradesh, India.
| |
Collapse
|
5
|
Bhingarkar A, Vangapandu HV, Rathod S, Hoshitsuki K, Fernandez CA. Amino Acid Metabolic Vulnerabilities in Acute and Chronic Myeloid Leukemias. Front Oncol 2021; 11:694526. [PMID: 34277440 PMCID: PMC8281237 DOI: 10.3389/fonc.2021.694526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/15/2021] [Indexed: 12/24/2022] Open
Abstract
Amino acid (AA) metabolism plays an important role in many cellular processes including energy production, immune function, and purine and pyrimidine synthesis. Cancer cells therefore require increased AA uptake and undergo metabolic reprogramming to satisfy the energy demand associated with their rapid proliferation. Like many other cancers, myeloid leukemias are vulnerable to specific therapeutic strategies targeting metabolic dependencies. Herein, our review provides a comprehensive overview and TCGA data analysis of biosynthetic enzymes required for non-essential AA synthesis and their dysregulation in myeloid leukemias. Furthermore, we discuss the role of the general control nonderepressible 2 (GCN2) and-mammalian target of rapamycin (mTOR) pathways of AA sensing on metabolic vulnerability and drug resistance.
Collapse
Affiliation(s)
- Aboli Bhingarkar
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, United States
| | - Hima V. Vangapandu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, United States
| | - Sanjay Rathod
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, United States
| | - Keito Hoshitsuki
- Division of General Internal Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Christian A. Fernandez
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, United States
| |
Collapse
|
6
|
García-Navas R, Gajate C, Mollinedo F. Neutrophils drive endoplasmic reticulum stress-mediated apoptosis in cancer cells through arginase-1 release. Sci Rep 2021; 11:12574. [PMID: 34131176 PMCID: PMC8206108 DOI: 10.1038/s41598-021-91947-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 06/02/2021] [Indexed: 12/19/2022] Open
Abstract
Human neutrophils constitutively express high amounts of arginase-1, which depletes arginine from the surrounding medium and downregulates T-cell activation. Here, we have found that neutrophil arginase-1, released from activated human neutrophils or dead cells, induced apoptosis in cancer cells through an endoplasmic reticulum (ER) stress pathway. Silencing of PERK in cancer cells prevented the induction of ER stress and apoptosis. Arginase inhibitor Nω-hydroxy-nor-arginine inhibited apoptosis and ER stress response induced by conditioned medium from activated neutrophils. A number of tumor cell lines, derived from different tissues, were sensitive to neutrophil arginase-1, with pancreatic, breast, ovarian and lung cancer cells showing the highest sensitivity. Neutrophil-released arginase-1 and arginine deprivation potentiated the antitumor action against pancreatic cancer cells of the ER-targeted antitumor alkylphospholipid analog edelfosine. Our study demonstrates the involvement of neutrophil arginase-1 in cancer cell killing and highlights the importance and complex role of neutrophils in tumor surveillance and biology.
Collapse
Affiliation(s)
- Rósula García-Navas
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, 37007, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Salamanca, Spain
| | - Consuelo Gajate
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, 37007, Salamanca, Spain.,Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), C/Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Faustino Mollinedo
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, 37007, Salamanca, Spain. .,Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), C/Ramiro de Maeztu 9, 28040, Madrid, Spain.
| |
Collapse
|
7
|
Shuvayeva GY, Bobak YP, Vovk OI, Kunz-Schughart LA, Fletcher MT, Stasyk OV. Indospicine combined with arginine deprivation triggers cancer cell death via caspase-dependent apoptosis. Cell Biol Int 2020; 45:518-527. [PMID: 32068315 DOI: 10.1002/cbin.11321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 02/16/2020] [Indexed: 12/15/2022]
Abstract
Arginine-deprivation therapy is a rapidly developing metabolic anticancer approach. To overcome the resistance of some cancer cells to this monotherapy, rationally designed combination modalities are needed. In this report, we evaluated for the first time indospicine, an arginine analogue of Indigofera plant genus origin, as potential enhancer compound for the metabolic therapy that utilizes recombinant human arginase I. We demonstrate that indospicine at low micromolar concentrations is selectively toxic for human colorectal cancer cells only in the absence of arginine. In arginine-deprived cancer cells indospicine deregulates some prosurvival pathways (PI3K-Akt and MAPK) and activates mammalian target of rapamycin, exacerbates endoplasmic reticulum stress and triggers caspase-dependent apoptosis, which is reversed by the exposure to translation inhibitors. Simultaneously, indospicine is not degraded by recombinant human arginase I and does not inhibit this arginine-degrading enzyme at its effective dose. The obtained results emphasize the potential of arginine structural analogues as efficient components for combinatorial metabolic targeting of malignant cells.
Collapse
Affiliation(s)
- Galyna Y Shuvayeva
- Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov Street 14/16, 79005, Lviv, Ukraine
| | - Yaroslav P Bobak
- Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov Street 14/16, 79005, Lviv, Ukraine
| | - Olena I Vovk
- Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov Street 14/16, 79005, Lviv, Ukraine
| | - Leoni A Kunz-Schughart
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, TU Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstr 74, Dresden, 01307, Germany.,National Center for Tumor Diseases, Partner site Dresden (NCT), Dresden, 01307, Germany
| | - Mary T Fletcher
- Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, PO Box 156 Archerfield, QLD, 4108, Australia
| | - Oleh V Stasyk
- Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov Street 14/16, 79005, Lviv, Ukraine
| |
Collapse
|
8
|
Abstract
In this Review, Rashkovan et al. discuss the role of cancer metabolic circuitries feeding anabolism and redox potential in leukemia development and recent progress in translating these important findings to the clinic. Leukemia cell proliferation requires up-regulation and rewiring of metabolic pathways to feed anabolic cell growth. Oncogenic drivers directly and indirectly regulate metabolic pathways, and aberrant metabolism is central not only for leukemia proliferation and survival, but also mediates oncogene addiction with significant implications for the development of targeted therapies. This review explores leukemia metabolic circuitries feeding anabolism, redox potential, and energy required for tumor propagation with an emphasis on emerging therapeutic opportunities.
Collapse
Affiliation(s)
- Marissa Rashkovan
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Adolfo Ferrando
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA.,Department of Pediatrics, Columbia University, New York, NY 10032, USA.,Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
9
|
Use of Immunohistochemical Markers (HNF-1β, Napsin A, ER, CTH, and ASS1) to Distinguish Endometrial Clear Cell Carcinoma From Its Morphologic Mimics Including Arias-Stella Reaction. Int J Gynecol Pathol 2019; 39:344-353. [DOI: 10.1097/pgp.0000000000000609] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
10
|
Metabolic pathways of L-arginine and therapeutic consequences in tumors. Adv Med Sci 2019; 64:104-110. [PMID: 30605863 DOI: 10.1016/j.advms.2018.08.018] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 06/03/2018] [Accepted: 08/31/2018] [Indexed: 02/06/2023]
Abstract
Difference in the metabolism of normal and cancer cells inspires to search for new, more specific and less toxic therapies than those currently used. The development of tumors is conditioned by genetic changes in cancer-transformed cells, immunological tolerance and immunosuppression. At the initial stages of carcinogenesis, the immune system shows anti-tumor activity, however later, cancer disrupts the function of Th1/Th17/Th2 lymphocytes by regulatory T (Treg) cells, tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) and finally causes immunosuppression. Recently, much attention has been devoted to the influence of l-arginine metabolism disorders on both carcinogenesis and the immune system. l-Arginine is essential for the maturation of the T cell receptor zeta (TCRζ), and its absence deprives T-cells of the ability to interact with tumor antigens. MDSCs deplete l-arginine due to a high expression of arginase 1 (ARG1) and their number increases 4-10 times depending on the type of the cancer. L-Arginine has been shown to be essential for the survival and progression of arginine auxotrophic tumors. However, the progression of arginine non-auxotrophic tumors is independent of exogenous l-arginine, because these tumors have arginine-succinate synthetase (ASS1) activity and are available to produce l-arginine from citrulline. Clinical studies have confirmed the high efficacy of arginine auxotrophic tumors therapy based on the elimination of l-arginine. However, l-arginine supplementation may improve the results of treatment of patients with arginine non-auxotrophic cancer. This review is an attempt to explain the seemingly contradictory results of oncological therapies based on the deprivation or supplementation of l-arginine.
Collapse
|
11
|
Rona GB, Almeida NP, Santos GC, Fidalgo TKS, Almeida FCL, Eleutherio ECA, Pinheiro AS. 1
H NMR metabolomics reveals increased glutaminolysis upon overexpression of NSD3s or Pdp3 in
Saccharomyces cerevisiae. J Cell Biochem 2018; 120:5377-5385. [DOI: 10.1002/jcb.27816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 09/12/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Germana B Rona
- Department of Biochemistry Institute of Chemistry, Federal University of Rio de Janeiro Rio de Janeiro Brazil
| | - Natalia P Almeida
- Department of Biochemistry Institute of Chemistry, Federal University of Rio de Janeiro Rio de Janeiro Brazil
| | - Gilson C Santos
- National Center for Nuclear Magnetic Resonance Jiri Jonas (CNRMN), Structural Biology Program, Medical Biochemistry Institute and Center for Structural Biology and Bioimaging I (CENABIO I), Federal University of Rio de Janeiro Rio de Janeiro Brazil
| | - Tatiana KS Fidalgo
- Department of Preventive and Community Dentistry, School of Dentistry, State University of Rio de Janeiro Rio de Janeiro Brazil
| | - Fabio CL Almeida
- National Center for Nuclear Magnetic Resonance Jiri Jonas (CNRMN), Structural Biology Program, Medical Biochemistry Institute and Center for Structural Biology and Bioimaging I (CENABIO I), Federal University of Rio de Janeiro Rio de Janeiro Brazil
| | - Elis CA Eleutherio
- Department of Biochemistry Institute of Chemistry, Federal University of Rio de Janeiro Rio de Janeiro Brazil
| | - Anderson S Pinheiro
- Department of Biochemistry Institute of Chemistry, Federal University of Rio de Janeiro Rio de Janeiro Brazil
| |
Collapse
|
12
|
Tripathi SC, Fahrmann JF, Vykoukal JV, Dennison JB, Hanash SM. Targeting metabolic vulnerabilities of cancer: Small molecule inhibitors in clinic. Cancer Rep (Hoboken) 2018; 2:e1131. [PMID: 32721114 DOI: 10.1002/cnr2.1131] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Altered cell metabolism is an established hallmark of cancer. Advancement in our understanding of dysregulated cellular metabolism has aided drastically in identifying metabolic vulnerabilities that can be exploited therapeutically. Indeed, this knowledge has led to the development of a multitude of agents targeting various aspects of tumor metabolism. RECENT FINDINGS The intent of this review is to provide insight into small molecule inhibitors that target tumor metabolism and that are currently being explored in active clinical trials as either preventive, stand-alone, or adjuvant therapies for various malignancies. For each inhibitor, we outline the mechanism (s) of action, preclinical/clinical findings, and limitations. Sections are divided into three aspects based on the primary target of the small molecule inhibitor (s): those that impact (1) cancer cells directly, (2) immune cells present in the tumor microenvironment, or (3) both cancer cells and immune cells. We highlight small molecule targeting of metabolic pathways including de novo fatty acid synthesis, NAD+ biosynthesis, 2-hydroxyglutarate biosynthesis, polyamine metabolism, the kynurenine pathway, as well as glutamine and arginine metabolism. CONCLUSIONS Use of small molecule inhibitors aimed at exploiting tumor metabolic vulnerabilities continues to be an active area of research. Identifying metabolic dependencies specific to cancer cells and/or constituents of the tumor microenvironment is a viable area of therapeutic intervention that holds considerable clinical potential.
Collapse
Affiliation(s)
- Satyendra C Tripathi
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Johannes F Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Jody V Vykoukal
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Jennifer B Dennison
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| |
Collapse
|
13
|
Lee HJ, Jedrychowski MP, Vinayagam A, Wu N, Shyh-Chang N, Hu Y, Min-Wen C, Moore JK, Asara JM, Lyssiotis CA, Perrimon N, Gygi SP, Cantley LC, Kirschner MW. Proteomic and Metabolomic Characterization of a Mammalian Cellular Transition from Quiescence to Proliferation. Cell Rep 2018; 20:721-736. [PMID: 28723573 DOI: 10.1016/j.celrep.2017.06.074] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/22/2017] [Accepted: 06/25/2017] [Indexed: 12/28/2022] Open
Abstract
There exist similarities and differences in metabolism and physiology between normal proliferative cells and tumor cells. Once a cell enters the cell cycle, metabolic machinery is engaged to facilitate various processes. The kinetics and regulation of these metabolic changes have not been properly evaluated. To correlate the orchestration of these processes with the cell cycle, we analyzed the transition from quiescence to proliferation of a non-malignant murine pro-B lymphocyte cell line in response to IL-3. Using multiplex mass-spectrometry-based proteomics, we show that the transition to proliferation shares features generally attributed to cancer cells: upregulation of glycolysis, lipid metabolism, amino-acid synthesis, and nucleotide synthesis and downregulation of oxidative phosphorylation and the urea cycle. Furthermore, metabolomic profiling of this transition reveals similarities to cancer-related metabolic pathways. In particular, we find that methionine is consumed at a higher rate than that of other essential amino acids, with a potential link to maintenance of the epigenome.
Collapse
Affiliation(s)
- Ho-Joon Lee
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Ning Wu
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Ng Shyh-Chang
- Stem Cell & Regenerative Biology, Genome Institute of Singapore, S138672 Singapore, Singapore
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Chua Min-Wen
- Stem Cell & Regenerative Biology, Genome Institute of Singapore, S138672 Singapore, Singapore
| | - Jodene K Moore
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - John M Asara
- Division of Signal Transduction, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Costas A Lyssiotis
- Division of Gastroenterology, Department of Molecular and Integrative Physiology and Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Marc W Kirschner
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
14
|
Saeed H, Ali H, Soudan H, Embaby A, El-Sharkawy A, Farag A, Hussein A, Ataya F. Molecular cloning, structural modeling and production of recombinant Aspergillus terreusl. asparaginase in Escherichia coli. Int J Biol Macromol 2018; 106:1041-1051. [PMID: 28851634 DOI: 10.1016/j.ijbiomac.2017.08.110] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/16/2017] [Accepted: 08/17/2017] [Indexed: 11/15/2022]
Abstract
l-Asparaginase (EC 3.5.1.1) is an important medical enzyme that catalysis the hydrolysis of l-asparagine to aspartic acid and ammonium. For over four decades l. asparaginase utic agent for the treatment of a variety of lymphoproliferative disorders and lymphoma such as acute lymphoblastic leukemia. In the present study A. terreus full length l. asparaginase gene, 1179bp was optimized for expression in Escherichia coli BL21 (DE3) pLysS. The full length A. terreusl. asparaginase gene encoding a protein of 376 amino acids with estimated molecular weight of 42.0kDa and a theoretical isoelectric point (pI) of 5.0. BLAST and phylogeny analysis revealed that the A. terreusl. asparaginase shared high similarity with other known fungal l. asparaginase (75% homology with A. nomius and 71% with A. nidulans). The recombinant protein was overexpressed in the form of amorphous submicron proteinaceous inclusion bodies upon induction with 1mM IPTG at 37°C for 18h.
Collapse
Affiliation(s)
- Hesham Saeed
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt.
| | - Hadeer Ali
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Hadeer Soudan
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Amira Embaby
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Amany El-Sharkawy
- Marine Biotechnology and Natural Products Extract Laboratory, National Institute of Oceanography and Fisheries (NIOF), Alexandria, Egypt
| | - Aida Farag
- Marine Biotechnology and Natural Products Extract Laboratory, National Institute of Oceanography and Fisheries (NIOF), Alexandria, Egypt
| | - Ahmed Hussein
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Farid Ataya
- Biochemistry Department, College of Science, King Saud University, Bld. 5, Lab AA10, P.O. Box: 2454, Riyadh, Saudi Arabia; Molecular Biology Department, Genetic Engineering Division, National Research Centre, 33 El-Bohouth St. (former El-Tahrir St.), P.O. 12622, Dokki, Giza, Egypt(1)
| |
Collapse
|
15
|
Savaraj N, Wu C, Kuo MT, You M, Wangpaichitr M, Robles C, Spector S, Feun L. The Relationship of Arginine Deprivation, Argininosuccinate Synthetase and Cell Death in Melanoma. Drug Target Insights 2017. [DOI: 10.1177/117739280700200016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Niramol Savaraj
- VA Medical Center, Hematology-Oncology, Miami, Florida, U.S.A
| | - Chunjing Wu
- University of Miami, Hematology-Oncology, Miami, Florida, U.S.A
| | - Marcus Tien Kuo
- M.D. Anderson Cancer Center, Molecular Pathology, Houston, Texas, U.S.A
| | - Min You
- University of Miami, Hematology-Oncology, Miami, Florida, U.S.A
| | | | - Carlos Robles
- VA Medical Center, Hematology-Oncology, Miami, Florida, U.S.A
| | - Seth Spector
- VA Medical Center, Hematology-Oncology, Miami, Florida, U.S.A
| | - Lynn Feun
- University of Miami, Hematology-Oncology, Miami, Florida, U.S.A
| |
Collapse
|
16
|
Affiliation(s)
- Wissam Zam
- Al-Andalus University for Medical Sciences, Syrian Arab Republic
| |
Collapse
|
17
|
Starkova J, Hermanova I, Hlozkova K, Hararova A, Trka J. Altered Metabolism of Leukemic Cells: New Therapeutic Opportunity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 336:93-147. [PMID: 29413894 DOI: 10.1016/bs.ircmb.2017.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The cancer metabolic program alters bioenergetic processes to meet the higher demands of tumor cells for biomass production, nucleotide synthesis, and NADPH-balancing redox homeostasis. It is widely accepted that cancer cells mostly utilize glycolysis, as opposed to normal cells, in which oxidative phosphorylation is the most employed bioenergetic process. Still, studies examining cancer metabolism had been overlooked for many decades, and it was only recently discovered that metabolic alterations affect both the oncogenic potential and therapeutic response. Since most of the published works concern solid tumors, in this comprehensive review, we aim to summarize knowledge about the metabolism of leukemia cells. Leukemia is a malignant disease that ranks first and fifth in cancer-related deaths in children and adults, respectively. Current treatment has reached its limits due to toxicity, and there has been a need for new therapeutic approaches. One of the possible scenarios is improved use of established drugs and another is to introduce new druggable targets. Herein, we aim to describe the complexity of leukemia metabolism and highlight cellular processes that could be targeted therapeutically and enhance the effectiveness of current treatments.
Collapse
Affiliation(s)
- Julia Starkova
- CLIP-Childhood Leukaemia Investigation Prague, Charles University, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Ivana Hermanova
- CLIP-Childhood Leukaemia Investigation Prague, Charles University, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Katerina Hlozkova
- CLIP-Childhood Leukaemia Investigation Prague, Charles University, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alzbeta Hararova
- Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Trka
- CLIP-Childhood Leukaemia Investigation Prague, Charles University, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic; University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
18
|
Hinrichs CN, Ingargiola M, Käubler T, Löck S, Temme A, Köhn-Luque A, Deutsch A, Vovk O, Stasyk O, Kunz-Schughart LA. Arginine Deprivation Therapy: Putative Strategy to Eradicate Glioblastoma Cells by Radiosensitization. Mol Cancer Ther 2017; 17:393-406. [DOI: 10.1158/1535-7163.mct-16-0807] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 05/08/2017] [Accepted: 07/26/2017] [Indexed: 11/16/2022]
|
19
|
Fernandes HS, Silva Teixeira CS, Fernandes PA, Ramos MJ, Cerqueira NMFSA. Amino acid deprivation using enzymes as a targeted therapy for cancer and viral infections. Expert Opin Ther Pat 2016; 27:283-297. [DOI: 10.1080/13543776.2017.1254194] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- H. S. Fernandes
- UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - C. S. Silva Teixeira
- UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - P. A. Fernandes
- UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - M. J. Ramos
- UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - N. M. F. S. A. Cerqueira
- UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| |
Collapse
|
20
|
Arginine Metabolism in Bacterial Pathogenesis and Cancer Therapy. Int J Mol Sci 2016; 17:363. [PMID: 26978353 PMCID: PMC4813224 DOI: 10.3390/ijms17030363] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 01/29/2016] [Accepted: 03/04/2016] [Indexed: 02/06/2023] Open
Abstract
Antibacterial resistance to infectious diseases is a significant global concern for health care organizations; along with aging populations and increasing cancer rates, it represents a great burden for government healthcare systems. Therefore, the development of therapies against bacterial infection and cancer is an important strategy for healthcare research. Pathogenic bacteria and cancer have developed a broad range of sophisticated strategies to survive or propagate inside a host and cause infection or spread disease. Bacteria can employ their own metabolism pathways to obtain nutrients from the host cells in order to survive. Similarly, cancer cells can dysregulate normal human cell metabolic pathways so that they can grow and spread. One common feature of the adaption and disruption of metabolic pathways observed in bacterial and cancer cell growth is amino acid pathways; these have recently been targeted as a novel approach to manage bacterial infections and cancer therapy. In particular, arginine metabolism has been illustrated to be important not only for bacterial pathogenesis but also for cancer therapy. Therefore, greater insights into arginine metabolism of pathogenic bacteria and cancer cells would provide possible targets for controlling of bacterial infection and cancer treatment. This review will summarize the recent progress on the relationship of arginine metabolism with bacterial pathogenesis and cancer therapy, with a particular focus on arginase and arginine deiminase pathways of arginine catabolism.
Collapse
|
21
|
Bol S, Bunnik EM. Lysine supplementation is not effective for the prevention or treatment of feline herpesvirus 1 infection in cats: a systematic review. BMC Vet Res 2015; 11:284. [PMID: 26573523 PMCID: PMC4647294 DOI: 10.1186/s12917-015-0594-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 11/05/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Feline herpesvirus 1 is a highly contagious virus that affects many cats. Virus infection presents with flu-like signs and irritation of ocular and nasal regions. While cats can recover from active infections without medical treatment, examination by a veterinarian is recommended. Lysine supplementation appears to be a popular intervention (recommended by > 90 % of veterinarians in cat hospitals). We investigated the scientific merit of lysine supplementation by systematically reviewing all relevant literature. METHODS NCBI's PubMed database was used to search for published work on lysine and feline herpesvirus 1, as well as lysine and human herpesvirus 1. Seven studies on lysine and feline herpesvirus 1 (two in vitro studies and 5 studies with cats), and 10 publications on lysine and human herpesvirus 1 (three in vitro studies and 7 clinical trials) were included for qualitative analysis. RESULTS There is evidence at multiple levels that lysine supplementation is not effective for the prevention or treatment of feline herpesvirus 1 infection in cats. Lysine does not have any antiviral properties, but is believed to act by lowering arginine levels. However, lysine does not antagonize arginine in cats, and evidence that low intracellular arginine concentrations would inhibit viral replication is lacking. Furthermore, lowering arginine levels is highly undesirable since cats cannot synthesize this amino acid themselves. Arginine deficiency will result in hyperammonemia, which may be fatal. In vitro studies with feline herpesvirus 1 showed that lysine has no effect on the replication kinetics of the virus. Finally, and most importantly, several clinical studies with cats have shown that lysine is not effective for the prevention or the treatment of feline herpesvirus 1 infection, and some even reported increased infection frequency and disease severity in cats receiving lysine supplementation. CONCLUSION We recommend an immediate stop of lysine supplementation because of the complete lack of any scientific evidence for its efficacy.
Collapse
Affiliation(s)
- Sebastiaan Bol
- Department of Botany and Plant Sciences, University of California, Riverside, 900 University Avenue, Riverside, CA, 92521, USA.
| | - Evelien M Bunnik
- Department of Cell Biology and Neuroscience, University of California, Riverside, 900 University Avenue, Riverside, CA, 92521, USA.
| |
Collapse
|
22
|
Anishkin A, Vanegas JM, Rogers DM, Lorenzi PL, Chan WK, Purwaha P, Weinstein JN, Sukharev S, Rempe SB. Catalytic Role of the Substrate Defines Specificity of Therapeutic l-Asparaginase. J Mol Biol 2015; 427:2867-85. [DOI: 10.1016/j.jmb.2015.06.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 05/20/2015] [Accepted: 06/26/2015] [Indexed: 12/23/2022]
|
23
|
Mussai F, Egan S, Higginbotham-Jones J, Perry T, Beggs A, Odintsova E, Loke J, Pratt G, U KP, Lo A, Ng M, Kearns P, Cheng P, De Santo C. Arginine dependence of acute myeloid leukemia blast proliferation: a novel therapeutic target. Blood 2015; 125:2386-96. [PMID: 25710880 PMCID: PMC4416943 DOI: 10.1182/blood-2014-09-600643] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 02/11/2015] [Indexed: 12/14/2022] Open
Abstract
Acute myeloid leukemia (AML) is one of the most common acute leukemias in adults and children, yet significant numbers of patients relapse and die of disease. In this study, we identify the dependence of AML blasts on arginine for proliferation. We show that AML blasts constitutively express the arginine transporters CAT-1 and CAT-2B, and that the majority of newly diagnosed patients' blasts have deficiencies in the arginine-recycling pathway enzymes argininosuccinate synthase and ornithine transcarbamylase, making them arginine auxotrophic. BCT-100, a pegylated human recombinant arginase, leads to a rapid depletion in extracellular and intracellular arginine concentrations, resulting in arrest of AML blast proliferation and a reduction in AML engraftment in vivo. BCT-100 as a single agent causes significant death of AML blasts from adults and children, and acts synergistically in combination with cytarabine. Using RNA sequencing, 20 further candidate genes which correlated with resistance have been identified. Thus, AML blasts are dependent on arginine for survival and proliferation, as well as depletion of arginine with BCT-100 of clinical value in the treatment of AML.
Collapse
MESH Headings
- Adolescent
- Aged
- Animals
- Antimetabolites, Antineoplastic/therapeutic use
- Arginase/therapeutic use
- Arginine/metabolism
- Child
- Child, Preschool
- Cytarabine/therapeutic use
- Enzyme Therapy
- Female
- Humans
- Infant
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice, SCID
- Middle Aged
- Recombinant Proteins/therapeutic use
- Tumor Cells, Cultured
- Young Adult
Collapse
Affiliation(s)
- Francis Mussai
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Sharon Egan
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | | | - Tracey Perry
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Andrew Beggs
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Elena Odintsova
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Justin Loke
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Guy Pratt
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Kin Pong U
- Bio-cancer Treatment International Ltd, Hong Kong Science Park, Shatin, New Territories, Hong Kong; and
| | - Anthony Lo
- Department of Anatomic Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Margaret Ng
- Department of Anatomic Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Pamela Kearns
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Cheng
- Bio-cancer Treatment International Ltd, Hong Kong Science Park, Shatin, New Territories, Hong Kong; and
| | - Carmela De Santo
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
24
|
Williams MD, Zhang X, Belton AS, Xian L, Huso T, Park JJ, Siems WF, Gang DR, Resar LMS, Reeves R, Hill HH. HMGA1 drives metabolic reprogramming of intestinal epithelium during hyperproliferation, polyposis, and colorectal carcinogenesis. J Proteome Res 2015; 14:1420-31. [PMID: 25643065 DOI: 10.1021/pr501084s] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Although significant progress has been made in the diagnosis and treatment of colorectal cancer (CRC), it remains a leading cause of cancer death worldwide. Early identification and removal of polyps that may progress to overt CRC is the cornerstone of CRC prevention. Expression of the High Mobility Group A1 (HMGA1) gene is significantly elevated in CRCs as compared with adjacent, nonmalignant tissues. We investigated metabolic aberrations induced by HMGA1 overexpression in small intestinal and colonic epithelium using traveling wave ion mobility mass spectrometry (TWIMMS) in a transgenic model in which murine Hmga1 was misexpressed in colonic epithelium. To determine if these Hmga1-induced metabolic alterations in mice were relevant to human colorectal carcinogenesis, we also investigated tumors from patients with CRC and matched, adjacent, nonmalignant tissues. Multivariate statistical methods and manual comparisons were used to identify metabolites specific to Hmga1 and CRC. Statistical modeling of data revealed distinct metabolic patterns in Hmga1 transgenics and human CRC samples as compared with the control tissues. We discovered that 13 metabolites were specific for Hmga1 in murine intestinal epithelium and also found in human CRC. Several of these metabolites function in fatty acid metabolism and membrane composition. Although further validation is needed, our results suggest that high levels of HMGA1 protein drive metabolic alterations that contribute to CRC pathogenesis through fatty acid synthesis. These metabolites could serve as potential biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Michael D Williams
- Department of Chemistry, Washington State University , 100 Dairy Road, Pullman, Washington 99164, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Zeng J, Yin P, Tan Y, Dong L, Hu C, Huang Q, Lu X, Wang H, Xu G. Metabolomics study of hepatocellular carcinoma: discovery and validation of serum potential biomarkers by using capillary electrophoresis-mass spectrometry. J Proteome Res 2014; 13:3420-31. [PMID: 24853826 DOI: 10.1021/pr500390y] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignancies. The lack of effective screening methods for early diagnosis has been a longstanding bottleneck to improve the survival rate. In the present study, a capillary electrophoresis-time-of-flight mass spectrometry (CE-TOF/MS)-based metabolomics method was employed to discover novel biomarkers for HCC. A total of 183 human serum specimens (77 sera in discovery set and 106 sera in external validation set) were enrolled in this study, and a "serum biomarker model" including tryptophan, glutamine, and 2-hydroxybutyric acid was finally established based on the comprehensive screening and validation workflow. This model was evaluated as an effective tool in that area under the receiver operating characteristic curve reached 0.969 in the discovery set and 0.99 in the validation set for diagnosing HCC from non-HCC (health and cirrhosis). Furthermore, this model enabled the discrimination of small HCC from precancer cirrhosis with an AUC of 0.976, highlighting the potential of early diagnosis. The biomarker model is effective for those a-fetoprotein (AFP) false-negative and false-postive subjects, indicating the complementary function to conventional tumor marker AFP. This study demonstrates the promising potential of CE-MS-based metabolomics approach in finding biomarkers for disease diagnosis and providing special insights into tumor metabolism.
Collapse
Affiliation(s)
- Jun Zeng
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences , 457 Zhongshan Road, Dalian 116023, China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Mass spectrometry-based metabolic profiling of gemcitabine-sensitive and gemcitabine-resistant pancreatic cancer cells. Pancreas 2014; 43:311-8. [PMID: 24518513 DOI: 10.1097/mpa.0000000000000092] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Gemcitabine resistance (GR) is one of the critical issues for therapy for pancreatic cancer, but the mechanism still remains unclear. Our aim was to increase the understanding of GR by metabolic profiling approach. METHODS To establish GR cells, 2 human pancreatic cancer cell lines, SUIT-2 and CAPAN-1, were exposed to increasing concentration of gemcitabine. Both parental and chemoresistant cells obtained by this treatment were subjected to metabolic profiling based on liquid chromatography-mass spectrometry. RESULTS Multivariate statistical analyses, both principal component analysis and orthogonal partial least squares discriminant analysis, distinguished metabolic signature of responsiveness and resistance to gemcitabine in both SUIT-2 and CAPAN-1 cells. Among significantly different (P < 0.005) metabolite peaks between parental and GR cells, we identified metabolites related to several metabolic pathways such as amino acid, nucleotide, energy, cofactor, and vitamin pathways. Decreases in glutamine and proline levels as well as increases in aspartate, hydroxyproline, creatine, and creatinine levels were observed in chemoresistant cells from both cell lines. CONCLUSIONS These results suggest that metabolic profiling can isolate distinct features of pancreatic cancer in the metabolome of gemcitabine-sensitive and GR cells. These findings may contribute to the biomarker discovery and an enhanced understanding of GR in pancreatic cancer.
Collapse
|
27
|
Lan J, Tai HC, Lee SW, Chen TJ, Huang HY, Li CF. Deficiency in expression and epigenetic DNA Methylation of ASS1 gene in nasopharyngeal carcinoma: negative prognostic impact and therapeutic relevance. Tumour Biol 2014; 35:161-169. [PMID: 23897555 DOI: 10.1007/s13277-013-1020-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 07/11/2013] [Indexed: 11/28/2022] Open
Abstract
The risk stratification and final outcomes in patients with nasopharyngeal carcinomas (NPC) still remain suboptimal. Our principal goals were to identify and validate targetable metabolic drivers relevant to pathogenesis of NPC using a published transcriptome. One prominently downregulated gene regulating amino acid metabolism was found to be argininosuccinate synthetase (ASS1). Attributable to epigenetic DNA methylation, ASS1 deficiency may link to the therapeutic sensitivity to the arginine-depriving agents and promote tumor aggressiveness through its newly identified tumor suppressor function. ASS1 immunohistochemistry was therefore examined in a well-defined cohort of 124 NPC biopsy specimens and in the neck lymph node metastases of another ten independent cases. For the latter, bisulphite pyrosequencing was performed to evaluate the extent of ASS1 gene methylation. ASS1 protein deficiency was identified in 64 of 124 cases (51.6%), significantly related to T3-T4 status (p = 0.006), and univariately associated with inferior local recurrence-free survival (p = 0.0427), distant metastasis-free survival (DMFS; p = 0.0036), and disease-specific survival (DSS; p = 0.0069). Together with advanced AJCC stages III-IV, ASS1 protein deficiency was also independently predictive of worse outcomes for the DFMS (p = 0.010, hazard ratio = 2.241) and DSS (p = 0.020, hazard ratio = 1.900). ASS1 promoter hypermethylation was detected in eight of ten neck nodal metastatic lesions by bisulphite pyrosequencing and associated with ASS1 protein deficiency (p < 0.001). In summary, ASS1 protein deficiency was seen in approximately a half of NPCs and associated with advanced T classification, DNA methylation, and clinical aggressiveness, consistent with its tumor suppressor role. This aberration may render pegylated arginine deiminase as a promising strategy for ASS1-deficient NPCs.
Collapse
Affiliation(s)
- Jui Lan
- Departments of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | | | | | | | | |
Collapse
|
28
|
Involvement of autophagy in recombinant human arginase-induced cell apoptosis and growth inhibition of malignant melanoma cells. Appl Microbiol Biotechnol 2013; 98:2485-94. [PMID: 23917632 DOI: 10.1007/s00253-013-5118-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Revised: 06/15/2013] [Accepted: 07/10/2013] [Indexed: 12/11/2022]
Abstract
Recombinant human arginase (rhArg) has been developed for arginine derivation therapy of cancer and is currently in clinical trials for a variety of malignant solid tumors. In this study, we reported for the first time that rhArg could induce obvious autophagy in human melanoma cells; inhibition of autophagy by chloroquine (CQ) significantly increased rhArg-induced cell apoptosis and growth inhibition of A375 cells. A significant increase in mitochondrial membrane potential loss and elevated intracellular reactive oxygen species (ROS) levels were detected in A375 cells after rhArg treatment when compared with control. Membrane transition inhibitor cyclosporine A blocked autophagy and accelerated cell death induced by rhArg, indicating that rhArg induced autophagy via mitochondria pathway. Furthermore, antioxidant N-acetyl-L-cysteine suppressed rhArg-induced autophagy and rescued cells from cell growth inhibition, suggesting that ROS played an important role in rhArg-induced A375 cell growth inhibition and autophagy. Akt/mTOR signaling pathway was involved in autophagy induced by rhArg in a time-dependent manner. Moreover, rhArg could induce ERK1/2 activation in a dose- and time-dependent manner and rhArg-induced autophagy was attenuated when p-ERK1/2 was inhibited by MEK 1/2 inhibitor, U0126. Taken together, this study provides new insight into the molecular mechanism of autophagy involved in rhArg-induced cell apoptosis and growth inhibition, which facilitates the development of rhArg in combination with CQ as a potential therapy for malignant melanoma.
Collapse
|
29
|
Huang HY, Wu WR, Wang YH, Wang JW, Fang FM, Tsai JW, Li SH, Hung HC, Yu SC, Lan J, Shiue YL, Hsing CH, Chen LT, Li CF. ASS1 as a novel tumor suppressor gene in myxofibrosarcomas: aberrant loss via epigenetic DNA methylation confers aggressive phenotypes, negative prognostic impact, and therapeutic relevance. Clin Cancer Res 2013; 19:2861-2872. [PMID: 23549872 DOI: 10.1158/1078-0432.ccr-12-2641] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The principal goals were to identify and validate targetable metabolic drivers relevant to myxofibrosarcoma pathogenesis using a published transcriptome. EXPERIMENTAL DESIGN As the most significantly downregulated gene regulating amino acid metabolism, argininosuccinate synthetase (ASS1) was selected for further analysis by methylation-specific PCR, pyrosequencing, and immunohistochemistry of myxofibrosarcoma samples. The roles of ASS1 in tumorigenesis and the therapeutic relevance of the arginine-depriving agent pegylated arginine deiminase (ADI-PEG20) were elucidated in ASS1-deficient myxofibrosarcoma cell lines and xenografts with and without stable ASS1 reexpression. RESULTS ASS1 promoter hypermethylation was detected in myxofibrosarcoma samples and cell lines and was strongly linked to ASS1 protein deficiency. The latter correlated with increased tumor grade and stage and independently predicted a worse survival. ASS1-deficient cell lines were auxotrophic for arginine and susceptible to ADI-PEG20 treatment, with dose-dependent reductions in cell viability and tumor growth attributable to cell-cycle arrest in the S-phase. ASS1 expression was restored in 2 of 3 ASS1-deficient myxofibrosarcoma cell lines by 5-aza-2'-deoxycytidine, abrogating the inhibitory effect of ADI-PEG20. Conditioned media following ASS1 reexpression attenuated HUVEC tube-forming capability, which was associated with suppression of MMP-9 and an antiangiogenic effect in corresponding myxofibrosarcoma xenografts. In addition to delayed wound closure and fewer invading cells in a Matrigel assay, ASS1 reexpression reduced tumor cell proliferation, induced G1-phase arrest, and downregulated cyclin E with corresponding growth inhibition in soft agar and xenograft assays. CONCLUSIONS Our findings highlight ASS1 as a novel tumor suppressor in myxofibrosarcomas, with loss of expression linked to promoter methylation, clinical aggressiveness, and sensitivity to ADI-PEG20.
Collapse
Affiliation(s)
- Hsuan-Ying Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Adam J, Yang M, Bauerschmidt C, Kitagawa M, O'Flaherty L, Maheswaran P, Özkan G, Sahgal N, Baban D, Kato K, Saito K, Iino K, Igarashi K, Stratford M, Pugh C, Tennant DA, Ludwig C, Davies B, Ratcliffe PJ, El-Bahrawy M, Ashrafian H, Soga T, Pollard PJ. A role for cytosolic fumarate hydratase in urea cycle metabolism and renal neoplasia. Cell Rep 2013; 3:1440-8. [PMID: 23643539 PMCID: PMC3675675 DOI: 10.1016/j.celrep.2013.04.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/24/2013] [Accepted: 04/08/2013] [Indexed: 11/28/2022] Open
Abstract
The identification of mutated metabolic enzymes in hereditary cancer syndromes has established a direct link between metabolic dysregulation and cancer. Mutations in the Krebs cycle enzyme, fumarate hydratase (FH), predispose affected individuals to leiomyomas, renal cysts, and cancers, though the respective pathogenic roles of mitochondrial and cytosolic FH isoforms remain undefined. On the basis of comprehensive metabolomic analyses, we demonstrate that FH1-deficient cells and tissues exhibit defects in the urea cycle/arginine metabolism. Remarkably, transgenic re-expression of cytosolic FH ameliorated both renal cyst development and urea cycle defects associated with renal-specific FH1 deletion in mice. Furthermore, acute arginine depletion significantly reduced the viability of FH1-deficient cells in comparison to controls. Our findings highlight the importance of extramitochondrial metabolic pathways in FH-associated oncogenesis and the urea cycle/arginine metabolism as a potential therapeutic target.
Collapse
Affiliation(s)
- Julie Adam
- Cancer Biology and Metabolism Group, Nuffield Department of Medicine, Henry Wellcome Building for Molecular Physiology, University of Oxford, Oxford OX3 7BN, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Wells JW, Evans CH, Scott MC, Rütgen BC, O'Brien TD, Modiano JF, Cvetkovic G, Tepic S. Arginase treatment prevents the recovery of canine lymphoma and osteosarcoma cells resistant to the toxic effects of prolonged arginine deprivation. PLoS One 2013; 8:e54464. [PMID: 23365669 PMCID: PMC3554772 DOI: 10.1371/journal.pone.0054464] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 12/11/2012] [Indexed: 11/18/2022] Open
Abstract
Rapidly growing tumor cells require a nutrient-rich environment in order to thrive, therefore, restricting access to certain key amino acids, such as arginine, often results in the death of malignant cells, which frequently display defective cell cycle check-point control. Healthy cells, by contrast, become quiescent and remain viable under arginine restriction, displaying full recovery upon return to arginine-rich conditions. The use of arginase therapy to restrict available arginine for selectively targeting malignant cells is currently under investigation in human clinical trials. However, the suitability of this approach for veterinary uses is unexplored. As a prelude to in vivo studies in canine malignancies, we examined the in vitro effects of arginine-deprivation on canine lymphoid and osteosarcoma cell lines. Two lymphoid and 2 osteosarcoma cell lines were unable to recover following 6 days of arginine deprivation, but all remaining cell lines displayed full recovery upon return to arginine-rich culture conditions. These remaining cell lines all proved susceptible to cell death following the addition of arginase to the cultures. The lymphoid lines were particularly sensitive to arginase, becoming unrecoverable after just 3 days of treatment. Two of the osteosarcoma lines were also susceptible over this time-frame; however the other 3 lines required 6-8 days of arginase treatment to prevent recovery. In contrast, adult progenitor cells from the bone marrow of a healthy dog were able to recover fully following 9 days of culture in arginase. Over 3 days in culture, arginase was more effective than asparaginase in inducing the death of lymphoid lines. These results strongly suggest that short-term arginase treatment warrants further investigation as a therapy for lymphoid malignancies and osteosarcomas in dogs.
Collapse
Affiliation(s)
- James W Wells
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Syed N, Langer J, Janczar K, Singh P, Lo Nigro C, Lattanzio L, Coley HM, Hatzimichael E, Bomalaski J, Szlosarek P, Awad M, O'Neil K, Roncaroli F, Crook T. Epigenetic status of argininosuccinate synthetase and argininosuccinate lyase modulates autophagy and cell death in glioblastoma. Cell Death Dis 2013; 4:e458. [PMID: 23328665 PMCID: PMC3563985 DOI: 10.1038/cddis.2012.197] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Arginine deprivation, either by nutritional starvation or exposure to ADI-PEG20, induces adaptive transcriptional upregulation of ASS1 and ASL in glioblastoma multiforme ex vivo cultures and cell lines. This adaptive transcriptional upregulation is blocked by neoplasia-specific CpG island methylation in either gene, causing arginine auxotrophy and cell death. In cells with methylated ASS1 or ASL CpG islands, ADI-PEG20 initially induces a protective autophagic response, but abrogation of this by chloroquine accelerates and potentiates cytotoxicity. Concomitant methylation in the CpG islands of both ASS1 and ASL, observed in a subset of cases, confers hypersensitivity to ADI-PEG20. Cancer stem cells positive for CD133 and methylation in the ASL CpG island retain sensitivity to ADI-PEG20. Our results show for the first time that epigenetic changes occur in both of the two key genes of arginine biosynthesis in human cancer and confer sensitivity to therapeutic arginine deprivation. We demonstrate that methylation status of the CpG islands, rather than expression levels per se of the genes, predicts sensitivity to arginine deprivation. Our results suggest a novel therapeutic strategy for this invariably fatal central nervous system neoplasm for which we have identified robust biomarkers and which overcomes the limitations to conventional chemotherapy imposed by the blood/brain barrier.
Collapse
Affiliation(s)
- N Syed
- John Fulcher Neuro-oncology Laboratory, Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Arginine deprivation as a new treatment strategy for head and neck cancer. Oral Oncol 2012; 48:1227-35. [DOI: 10.1016/j.oraloncology.2012.06.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 06/02/2012] [Accepted: 06/08/2012] [Indexed: 11/18/2022]
|
34
|
Abstract
OBJECTIVES In this study, our aim was to test whether asparagine synthetase (ASNS) deficiency in pancreatic malignant cells can lead to sensitivity to asparagine starvation. We also investigated, in tumor-bearing mice, the efficacy of L-asparaginase entrapped in red blood cells (RBCs), a safe formulation, to induce asparagine depletion. METHODS First, ASNS expression was evaluated by immunohistochemistry in sporadic pancreatic ductal adenocarcinoma. Then, 4 pancreatic carcinoma cell lines were examined by Western blot, immunocytochemistry, and cytotoxicity assay to L-asparaginase and in asparagine-free or reduced-asparagine media. Finally, mice bearing the most in vitro sensitive cell line received RBC-entrapped L-asparaginase to investigate the anticancer efficacy of serum asparagine depletion in vivo. RESULTS Approximately 52% of pancreatic adenocarcinomas expressed no or low ASNS. The highest in vitro cytotoxicity to L-asparaginase or to reduced asparagine medium was observed with SW1990 line when ASNS expression was the lowest. In vivo sensitivity was confirmed for this cell line. CONCLUSIONS Plasma asparagine depletion by RBC-entrapped L-asparaginase in selected patients having no low or no ASNS may be a promising therapeutic approach for pancreatic cancer.
Collapse
|
35
|
Hsueh EC, Knebel SM, Lo WH, Leung YC, Cheng PNM, Hsueh CT. Deprivation of arginine by recombinant human arginase in prostate cancer cells. J Hematol Oncol 2012; 5:17. [PMID: 22546217 PMCID: PMC3403903 DOI: 10.1186/1756-8722-5-17] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 04/30/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recombinant human arginase (rhArg) has been developed for arginine deprivation therapy in cancer, and is currently under clinical investigation. During pre-clinical evaluation, rhArg has exhibited significant anti-proliferative activity in cancer cells deficient in the expression of ornithine carbamoyl transferase (OCT). Interestingly, a variety of cancer cells such as melanoma and prostate cancer deficient in argininosuccinate synthetase (ASS) are sensitive to arginine deprivation by arginine deiminase. In this study, we investigated levels of gene expression of OCT and ASS, and the effects of rhArg in human prostate cancer cells: LNCaP (androgen-dependent), PC-3 and DU-145 (both androgen-independent). RESULTS Quantitative real-time PCR showed minimal to absent gene expression of OCT, but ample expression of ASS expression in all 3 cell lines. Cell viability assay after 72-h exposure of rhArg showed all 3 lines had half maximal inhibitory concentration less than or equal to 0.02 U/ml. Addition of ornithine to cell culture media failed to rescue these cells from rhArg-mediated cytotoxicity.Decreased phosphorylation of 4E-BP1, a downstream effector of mammalian target of rapamycin (mTOR), was noted in DU-145 and PC-3 after exposure to rhArg. Moreover, there was no significant apoptosis induction after arginine deprivation by rhArg in all 3 prostate cancer cell lines. CONCLUSION rhArg causes significant cytotoxicity in LNCaP, DU-145 and PC-3 prostate cancer cells which all demonstrate decreased OCT expression. Inhibition of mTOR manifested by hypophosphorylation of 4E-BP1 suggests autophagy is involved as alternative cell death mechanism. rhArg demonstrates a promising novel agent for prostate cancer treatment.
Collapse
Affiliation(s)
- Eddy C Hsueh
- Department of Surgery, Saint Louis University, St. Louis, MO, USA
| | | | | | | | | | | |
Collapse
|
36
|
Stone E, Chantranupong L, Gonzalez C, O'Neal J, Rani M, VanDenBerg C, Georgiou G. Strategies for optimizing the serum persistence of engineered human arginase I for cancer therapy. J Control Release 2011; 158:171-9. [PMID: 22001609 DOI: 10.1016/j.jconrel.2011.09.097] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 08/17/2011] [Accepted: 09/29/2011] [Indexed: 11/16/2022]
Abstract
Systemic L-arginine depletion following intravenous administration of l-arginine hydrolyzing enzymes has been shown to selectively impact tumors displaying urea cycle defects including a large fraction of hepatocellular carcinomas, metastatic melanomas and small cell lung carcinomas. However, the human arginases display poor serum stability (t(1/2)=4.8h) whereas a bacterial arginine deiminase evaluated in phase II clinical trials was reported to be immunogenic, eliciting strong neutralizing antibody responses. Recently, we showed that substitution of the Mn(2+) metal center in human Arginase I with Co(2+) (Co-hArgI) results in an enzyme that displays 10-fold higher catalytic efficiency for L-Arg hydrolysis, 12-15 fold reduction in the IC(50) towards a variety of malignant cell lines and, importantly a t(1/2)=22h in serum. To investigate the utility of Co-hArgI for L-Arg depletion therapy in cancer we systematically investigated three strategies for enhancing the persistence of the enzyme in circulation: (i) site specific conjugation of Co-hArgI engineered with an accessible N-terminal Cys residue to 20kDa PEG-maleimide (Co-hArgI-C(PEG-20K)); (ii) engineering of the homotrimeric Co-hArgI into a linked, monomeric 110kDa polypeptide (Co-hArgI x3) and (iii) lysyl conjugation of 5kDa PEG-N-hydroxysuccinimide (NHS) ester (Co-hArgI-K(PEG-5K)). Surprisingly, even though all three formulations resulted in proteins with a predicted hydrodynamic radius larger than the cut-off for renal filtration, only Co-hArgI amine conjugated to 5kDa PEG remained in circulation for sufficiently long durations. Using Co-hArgI-K(PEG-5K) labeled with an end-terminal fluorescein for easy detection, we demonstrated that following intraperitoneal administration at 6mg/kg weight, a well tolerated dose, the circulation t(1/2) of the protein in Balb/c mice is 63±10h. Very low levels of serum L-Arg (<5μM) could be sustained for over 75h after injection, representing a 9-fold increase in pharmacodynamic efficacy relative to similarly prepared Mn(2+)-containing hArgI conjugated to 5kDa PEG-NHS ester (Mn-hArgI-K(PEG-5K)). The favorable pharmacokinetic and pharmacodynamic properties of Co-hArgI-K(PEG-5K) reported here, coupled with its human origin which should reduce the likelihood of adverse immune responses, make it a promising candidate for cancer therapy.
Collapse
Affiliation(s)
- Everett Stone
- Department of Chemical Engineering, Center for Molecular, College of Pharmacy, University of Texas, Austin, TX 78712, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
McConathy J, Yu W, Jarkas N, Seo W, Schuster DM, Goodman MM. Radiohalogenated nonnatural amino acids as PET and SPECT tumor imaging agents. Med Res Rev 2011; 32:868-905. [DOI: 10.1002/med.20250] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Jonathan McConathy
- Mallinckrodt Institute of Radiology; Washington University School of Medicine; St. Louis Missouri
| | - Weiping Yu
- Department of Radiology and Imaging Sciences; School of Medicine, Emory University; Atlanta Georgia
| | - Nachwa Jarkas
- Department of Radiology and Imaging Sciences; School of Medicine, Emory University; Atlanta Georgia
| | - Wonewoo Seo
- Department of Radiology and Imaging Sciences; School of Medicine, Emory University; Atlanta Georgia
| | - David M. Schuster
- Department of Radiology and Imaging Sciences; School of Medicine, Emory University; Atlanta Georgia
| | - Mark M. Goodman
- Department of Radiology and Imaging Sciences; School of Medicine, Emory University; Atlanta Georgia
| |
Collapse
|
38
|
Robertson DG, Watkins PB, Reily MD. Metabolomics in toxicology: preclinical and clinical applications. Toxicol Sci 2010; 120 Suppl 1:S146-70. [PMID: 21127352 DOI: 10.1093/toxsci/kfq358] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Donald G Robertson
- Applied and Investigative Metabolomics, Bristol-Myers Squibb Co., Princeton, New Jersey 08543, USA.
| | | | | |
Collapse
|
39
|
McConathy J, Zhou D, Shockley SE, Jones LA, Griffin EA, Lee H, Adams SJ, Mach RH. Click Synthesis and Biologic Evaluation of (
R
)- and (
S
)-2-Amino-3-[1-(2-[
18
F]Fluoroethyl)-1
H
-[1,2,3]Triazol-4-yl]Propanoic Acid for Brain Tumor Imaging with Positron Emission Tomography. Mol Imaging 2010. [DOI: 10.2310/7290.2010.00025] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Jonathan McConathy
- Department of Radiology (Radiologic Sciences), Washington University School of Medicine, St. Louis, MO; Division of Radiological Sciences, Mallinckrodt Institute of Radiology, St. Louis, MO; and Molecular Neuroimaging, New Haven, CT
| | - Dong Zhou
- Department of Radiology (Radiologic Sciences), Washington University School of Medicine, St. Louis, MO; Division of Radiological Sciences, Mallinckrodt Institute of Radiology, St. Louis, MO; and Molecular Neuroimaging, New Haven, CT
| | - Stephany E. Shockley
- Department of Radiology (Radiologic Sciences), Washington University School of Medicine, St. Louis, MO; Division of Radiological Sciences, Mallinckrodt Institute of Radiology, St. Louis, MO; and Molecular Neuroimaging, New Haven, CT
| | - Lynne A. Jones
- Department of Radiology (Radiologic Sciences), Washington University School of Medicine, St. Louis, MO; Division of Radiological Sciences, Mallinckrodt Institute of Radiology, St. Louis, MO; and Molecular Neuroimaging, New Haven, CT
| | - Elizabeth A. Griffin
- Department of Radiology (Radiologic Sciences), Washington University School of Medicine, St. Louis, MO; Division of Radiological Sciences, Mallinckrodt Institute of Radiology, St. Louis, MO; and Molecular Neuroimaging, New Haven, CT
| | - Hsiaoju Lee
- Department of Radiology (Radiologic Sciences), Washington University School of Medicine, St. Louis, MO; Division of Radiological Sciences, Mallinckrodt Institute of Radiology, St. Louis, MO; and Molecular Neuroimaging, New Haven, CT
| | - Susan J. Adams
- Department of Radiology (Radiologic Sciences), Washington University School of Medicine, St. Louis, MO; Division of Radiological Sciences, Mallinckrodt Institute of Radiology, St. Louis, MO; and Molecular Neuroimaging, New Haven, CT
| | - Robert H. Mach
- Department of Radiology (Radiologic Sciences), Washington University School of Medicine, St. Louis, MO; Division of Radiological Sciences, Mallinckrodt Institute of Radiology, St. Louis, MO; and Molecular Neuroimaging, New Haven, CT
| |
Collapse
|
40
|
Cancer cell sensitivity to arginine deprivationin vitrois not determined by endogenous levels of arginine metabolic enzymes. Cell Biol Int 2010; 34:1085-9. [DOI: 10.1042/cbi20100451] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
41
|
Savaraj N, You M, Wu C, Wangpaichitr M, Kuo MT, Feun LG. Arginine deprivation, autophagy, apoptosis (AAA) for the treatment of melanoma. Curr Mol Med 2010; 10:405-12. [PMID: 20459375 PMCID: PMC3096550 DOI: 10.2174/156652410791316995] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Accepted: 08/12/2009] [Indexed: 01/27/2023]
Abstract
The majority of melanoma cells do not express argininosuccinate synthetase (ASS), and hence cannot synthesize arginine from citrulline. Their growth and proliferation depend on exogenous supply of arginine. Arginine degradation using arginine deiminase (ADI) leads to growth inhibition and eventually cell death while normal cell which express ASS can survive. This notion has been translated into clinical trial. Pegylated ADI (ADI-PEG20) has shown antitumor activity in melanoma. However, the sensitivity to ADI is different among ASS(-) melanoma cells. We have investigated and reviewed the signaling pathways which are affected by arginine deprivation and their consequences which lead to cell death. We have found that arginine deprivation inhibits mTOR signaling but leads to activation of MEK and ERK with no changes in BRAF. These changes most likely lead to autophagy, a possible mechanism to survive by recycling intracellular arginine. However apoptosis does occur which can be both caspase dependent or independent In order to increase the therapeutic efficacy of this form of treatment, one should consider adding other agent(s) which can drive the cells toward apoptosis or inhibit the autophagic process.
Collapse
Affiliation(s)
- N Savaraj
- VA Medical Center, Miami, FL 33125, USA.
| | | | | | | | | | | |
Collapse
|
42
|
You M, Savaraj N, Wangpaichitr M, Wu C, Kuo TM, Varona-Santos J, Nguyen D, Feun L. The combination of ADI-PEG20 and TRAIL effectively increases cell death in melanoma cell lines. Biochem Biophys Res Commun 2010; 394:760-6. [PMID: 20227389 PMCID: PMC2860865 DOI: 10.1016/j.bbrc.2010.03.066] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Accepted: 03/10/2010] [Indexed: 11/25/2022]
Abstract
Current treatment for advanced, metastatic melanoma is not very effective, and new modalities are needed. ADI-PEG20 is a drug that specifically targets ASS-negative malignant melanomas while sparing the ASS-expressing normal cells. Although laboratory research and clinical trials showed promising results, there are some ASS-negative cell lines and patients that can develop resistance to this drug. In this report, we combined ADI-PEG20 with another antitumor drug TRAIL to increase the killing of malignant melanoma cells. This combination can greatly inhibit cell growth (to over 80%) and also enhanced cell death (to over 60%) in four melanoma cell lines tested compared with control. We found that ADI-PEG20 could increase the cell surface receptors DR4/5 for TRAIL and that caspase activity correlated with the increased cell death. These two drugs could also increase the level of Noxa while decrease that of survivin. We propose that these two drugs can complement each other by activating the intrinsic and extrinsic apoptosis pathways, thus enhance the killing of melanoma cells.
Collapse
Affiliation(s)
- Min You
- Sylverster Comprehensive Cancer Center, University of Miami Miller School of Medicine; 1475 NW 12th AVE., Miami, FL 33136, USA
| | - Niramol Savaraj
- Sylverster Comprehensive Cancer Center, University of Miami Miller School of Medicine; 1475 NW 12th AVE., Miami, FL 33136, USA
- Division of Hematology and Oncology, Miami Veterans Affairs Healthcare System; 1201 NW 16th St., Miami, FL 33125, USA
| | - Medhi Wangpaichitr
- Division of Hematology and Oncology, Miami Veterans Affairs Healthcare System; 1201 NW 16th St., Miami, FL 33125, USA
| | - Chunjing Wu
- Division of Hematology and Oncology, Miami Veterans Affairs Healthcare System; 1201 NW 16th St., Miami, FL 33125, USA
| | - Tien M. Kuo
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Javier Varona-Santos
- Sylverster Comprehensive Cancer Center, University of Miami Miller School of Medicine; 1475 NW 12th AVE., Miami, FL 33136, USA
| | - Dao Nguyen
- Sylverster Comprehensive Cancer Center, University of Miami Miller School of Medicine; 1475 NW 12th AVE., Miami, FL 33136, USA
| | - Lynn Feun
- Sylverster Comprehensive Cancer Center, University of Miami Miller School of Medicine; 1475 NW 12th AVE., Miami, FL 33136, USA
| |
Collapse
|
43
|
Tsai WB, Aiba I, Lee SY, Feun L, Savaraj N, Kuo MT. Resistance to arginine deiminase treatment in melanoma cells is associated with induced argininosuccinate synthetase expression involving c-Myc/HIF-1alpha/Sp4. Mol Cancer Ther 2010; 8:3223-33. [PMID: 19934275 DOI: 10.1158/1535-7163.mct-09-0794] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Arginine deiminase (ADI)-based arginine depletion is a novel strategy under clinical trials for the treatment of malignant melanoma with promising results. The sensitivity of melanoma to ADI treatment is based on its auxotrophy for arginine due to a lack of argininosuccinate synthetase (AS) expression, the rate-limiting enzyme for the de novo biosynthesis of arginine. We show here that AS expression can be transcriptionally induced by ADI in melanoma cell lines A2058 and SK-MEL-2 but not in A375 cells, and this inducibility was correlated with resistance to ADI treatment. The proximal region of the AS promoter contains an E-box that is recognized by c-Myc and HIF-1alpha and a GC-box by Sp4. Through ChIP assays, we showed that under noninduced conditions, the E-box was bound by HIF-1alpha in all the three melanoma cell lines. Under arginine depletion conditions, HIF-1alpha was replaced by c-Myc in A2058 and SK-MEL-2 cells but not in A375 cells. Sp4 was constitutively bound to the GC-box regardless of arginine availability in all three cell lines. Overexpressing c-Myc by transfection upregulated AS expression in A2058 and SK-MEL-2 cells, whereas cotransfection with HIF-1alpha suppressed c-Myc-induced AS expression. These results suggest that regulation of AS expression involves interplay among positive transcriptional regulators c-Myc and Sp4, and negative regulator HIF-1alpha that confers resistance to ADI treatment in A2058 and SK-MEL-2 cells. Inability of AS induction in A375 cells under arginine depletion conditions was correlated by the failure of c-Myc to interact with the AS promoter.
Collapse
Affiliation(s)
- Wen-Bin Tsai
- Department of Molecular Pathology, Unit 951, The University of Texas M. D. Anderson Cancer Center, 7435 Fannin Street, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
44
|
Rebeca R, Bracht L, Noleto GR, Martinez GR, Cadena SMSC, Carnieri EGS, Rocha MEM, de Oliveira MBM. Production of cachexia mediators by Walker 256 cells from ascitic tumors. Cell Biochem Funct 2008; 26:731-8. [PMID: 18646274 DOI: 10.1002/cbf.1497] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In neoplasic cachexia, chemical mediators seem to act as initiators or perpetuators of this process. Walker 256 cells, whose metabolic properties have so far been little studied with respect to cancer cachexia, are used as a model for the study of this syndrome. The main objective of this research was to pinpoint the substances secreted by these cells that may contribute to the progression of the cachectic state. Since inflammatory mediators seem to be involved in the manifestation of this syndrome, the in vitro production of nitric oxide (NO), cytokines (tumor necrosis factor alpha (TNF-alpha) and interleukin-6 (IL-6)), and prostaglandin E2 (PGE2) was evaluated in Walker 256 cells isolated from ascitic tumors. After 4 or 5 h, a significant increase in NO production was observed (2.55 +/- 1.56 and 4.05 +/- 1.99 nmol NO per 10(7) cells, respectively). When isolated from a 6-day-old tumor, a significantly lower production of IL-6 and higher production of TNF-alpha than in cells from a 4-day-old tumor were observed, indicating a relationship between the production of cytokines and the time of tumor development after implantation. Considerable production of PGE(2) by Walker 256 cells isolated from the 6-day-old tumor was also observed. Polyamines were also determined in Walker 256 cells. Levels of putrescine, spermidine, and spermine did not show significant differences in tumors developed during 4 or 6 days. Direct evidence of the release of proinflammatory cytokines and PGE2 by Walker 256 cells suggests that these mediators can drive the cachectic syndrome in the host, the effect being dependent on tumor development time.
Collapse
Affiliation(s)
- Rosilene Rebeca
- Department of Biochemistry, UFPR-Federal University of Paraná, Curitiba, CEP, PR, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Bowles TL, Kim R, Galante J, Parsons CM, Virudachalam S, Kung HJ, Bold RJ. Pancreatic cancer cell lines deficient in argininosuccinate synthetase are sensitive to arginine deprivation by arginine deiminase. Int J Cancer 2008; 123:1950-5. [PMID: 18661517 PMCID: PMC4294549 DOI: 10.1002/ijc.23723] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Eukaryotic cells can synthesize the non-essential amino acid arginine from aspartate and citrulline using the enzyme argininosuccinate synthetase (ASS). It has been observed that ASS is underexpressed in various types of cancers ASS, for which arginine become auxotrophic. Arginine deiminase (ADI) is a prokaryotic enzyme that metabolizes arginine to citrulline and has been found to inhibit melanoma and hepatoma cancer cells deficient of ASS. We tested the hypothesis that pancreatic cancers have low ASS expression and therefore arginine deprivation by ADI will inhibit cell growth. ASS expression was examined in 47 malignant and 20 non-neoplastic pancreatic tissues as well as a panel of human pancreatic cancer cell lines. Arginine deprivation was achieved by treatment with a recombinant form of ADI formulated with polyethylene glycol (PEG-ADI). Effects on caspase activation, cell growth and cell death were examined. Furthermore, the effect of PEG-ADI on the in vivo growth of pancreatic xenografts was examined. Eighty-seven percent of the tumors lacked ASS expression; 5 of 7 cell lines similarly lacked ASS expression. PEG-ADI specifically inhibited growth of those cell lines lacking ASS. PEG-ADI treatment induced caspase activation and induction of apoptosis. PEG-ADI was well tolerated in mice despite complete elimination of plasma arginine; tumor growth was inhibited by approximately 50%. Reduced expression of ASS occurs in pancreatic cancer and predicts sensitivity to arginine deprivation achieved by PEG-ADI treatment. Therefore, these findings suggest that arginine deprivation by ADI could provide a beneficial strategy for the treatment of pancreatic cancer, a malignancy in which new therapy is desperately needed.
Collapse
Affiliation(s)
- Tawnya L. Bowles
- Department of Surgery, University of California, Davis Medical Center, Sacramento, CA
| | - Randie Kim
- Department of Biochemistry and Molecular Medicine, University of California, Davis Medical Center, Sacramento, CA
| | - Joseph Galante
- Department of Surgery, University of California, Davis Medical Center, Sacramento, CA
| | - Colin M. Parsons
- Department of Surgery, University of California, Davis Medical Center, Sacramento, CA
| | | | - Hsing-Jien Kung
- Department of Biochemistry and Molecular Medicine, University of California, Davis Medical Center, Sacramento, CA
| | - Richard J. Bold
- Department of Surgery, University of California, Davis Medical Center, Sacramento, CA
| |
Collapse
|
46
|
Lagarde SM, Ver Loren van Themaat PE, Moerland PD, Gilhuijs-Pederson LA, Ten Kate FJW, Reitsma PH, van Kampen AHC, Zwinderman AH, Baas F, van Lanschot JJB. Analysis of gene expression identifies differentially expressed genes and pathways associated with lymphatic dissemination in patients with adenocarcinoma of the esophagus. Ann Surg Oncol 2008; 15:3459-70. [PMID: 18825457 DOI: 10.1245/s10434-008-0165-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2008] [Revised: 08/22/2008] [Accepted: 08/23/2008] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The presence of lymphatic dissemination is an important predictor of survival in esophageal adenocarcinoma (EA). The aim of this study was to discover a prognostic gene expression profile for lymphatic dissemination in EA and to identify genes and pathways that provide oncological insight in lymphatic dissemination. METHODS Patients who had lymphatic dissemination (N = 55) were compared with patients without lymphatic dissemination (N = 22). Whole-genome oligonucleotide microarrays were used to evaluate the genetic signature of 77 esophageal cancers. Multiple random validation was used to analyze the stability of the molecular signature and predictive power. Gene set enrichment analysis (GSEA) was applied to elucidate oncogenetic pathways. RESULTS Lymphatic dissemination was correctly predicted in 75 +/- 14% of lymph node positive patients. The absence of lymphatic dissemination was correctly predicted in 41 +/- 23% of lymph-node-negative patients. Argininosuccinate synthetase (ASS) was selected for validation on the protein level because it was present in most prognostic signatures as well as the list of differentially expressed genes. ASS expression was lower (P = 0.048) in patients with lymphatic dissemination than in patients without. GSEA identified that arginine metabolism pathways and lipid metabolism pathways are related to less chance of developing lymphatic dissemination. DISCUSSION The predictive profile does not outperform current clinical practice to predict the presence of lymphatic dissemination in patients with EA. Several genes, including ASS, and genetic pathways which are important in the development of lymphatic dissemination in EA, were identified.
Collapse
Affiliation(s)
- S M Lagarde
- Department of Surgery, Academic Medical Center at the University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Mobley AS, Lucero MT, Michel WC. Cross-species comparison of metabolite profiles in chemosensory epithelia: an indication of metabolite roles in chemosensory cells. Anat Rec (Hoboken) 2008; 291:410-32. [PMID: 18361450 PMCID: PMC2576748 DOI: 10.1002/ar.20666] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Comparative studies of chemosensory systems in vertebrates and invertebrates have greatly enhanced our understanding of anatomical and physiological constraints of chemical detection. Immunohistochemical comparisons of chemosensory systems are difficult to make across species due to limited cross-reactivity of mammalian-based antibodies. Immunostaining chemosensory tissues with glutaraldehyde-based antibodies generated against small metabolites in combination with hierarchical cluster analyses provide a novel approach for identifying and classifying cell types regardless of species. We used this "metabolite profiling" technique to determine whether metabolite profiles can be used to identify cell classes within and across different species including mouse, zebrafish, lobster and squid. Within a species, metabolite profiles for distinct cell classes were generally consistent. We found several metabolite-based cell classifications that mirrored function or receptor protein-based classifications. Although profiles of all six metabolites differed across species, we found that specific metabolites were associated with certain cell types. For example, elevated levels of glutathione were characteristic of nonsensory cells from vertebrates, suggesting an antioxidative role in non-neuronal cells in sensory tissues. Collectively, we found significantly different metabolite profiles for distinct cell populations in chemosensory tissue within all of the species studied. Based on their roles in other systems or cells, we discuss the roles of L-arginine, L-aspartate, L-glutamate, glycine, glutathione, and taurine within chemosensory epithelia.
Collapse
|
48
|
Arginine deiminase, a potential anti-tumor drug. Cancer Lett 2008; 261:1-11. [PMID: 18179862 DOI: 10.1016/j.canlet.2007.11.038] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Revised: 11/21/2007] [Accepted: 11/23/2007] [Indexed: 11/24/2022]
Abstract
Arginine deiminase (ADI; EC 3.5.3.6), an arginine-degrading enzyme, has been studied as a potential anti-tumor drug for the treatment of arginine-auxotrophic tumors, such as hepatocellular carcinomas (HCCs) and melanomas. Studies with human lymphatic leukemia cell lines further suggest that ADI is a potential anti-angiogenic agent and is effective in the treatment of leukemia. For instance ADI-PEG-20, patented by Pheonix Pharmacologic Inc., is currently in clinical trials for the treatment of HCC (Phase II/III) and melanoma (Phase I/II). This review summarizes results on recombinant expression, structural analysis, PEG (polyethylene glycerol) modification, in vivo anti-cancer activities, and clinical studies of ADI. Discussions on heterogeneous expression of ADI, directed evolution for improving enzymatic properties, and HSA-fusion for increased in vivo activity conclude this review.
Collapse
|
49
|
Keun HC. Biomarker discovery for drug development and translational medicine using metabonomics. ERNST SCHERING FOUNDATION SYMPOSIUM PROCEEDINGS 2008:79-98. [PMID: 18811054 DOI: 10.1007/2789_2008_090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
There exists at present an urgent desire for better biomarkers, especially in the context of pharmaceutical drug development and in the detection and management of disease. Many researchers in the area of biomarker discovery and development have turned to the "-omics" sciences as a way of addressing these needs. Metabolic profiling, or metabonomics, defines the metabolic phenotype and offers a source of novel biomarkers that have better potential to translate effectively. This review will discuss the broad philosophy and motivations behind metabonomics, and illustrate the case with applications relevant to pharmaceutical development and patient management. Particular focus will be paid to the potential of metabonomics to contribute to biomarker discovery in toxicology and cancer research.
Collapse
Affiliation(s)
- H C Keun
- Biomolecular Medicine, Division of Surgery, Oncology, Reproductive Biology and Anaesthetics, Imperial College Faculty of Medicine, Sir Alexander Fleming Building, Exhibition Road, SW7 2AZ South Kensington, London, UK.
| |
Collapse
|
50
|
Feun L, You M, Wu CJ, Kuo MT, Wangpaichitr M, Spector S, Savaraj N. Arginine deprivation as a targeted therapy for cancer. Curr Pharm Des 2008; 14:1049-57. [PMID: 18473854 PMCID: PMC3096551 DOI: 10.2174/138161208784246199] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Certain cancers may be auxotrophic for a particular amino acid, and amino acid deprivation is one method to treat these tumors. Arginine deprivation is a novel approach to target tumors which lack argininosuccinate synthetase (ASS) expression. ASS is a key enzyme which converts citrulline to arginine. Tumors which usually do not express ASS include melanoma, hepatocellular carcinoma, some mesotheliomas and some renal cell cancers. Arginine can be degraded by several enzymes including arginine deiminase (ADI). Although ADI is a microbial enzyme from mycoplasma, it has high affinity to arginine and catalyzes arginine to citrulline and ammonia. Citrulline can be recycled back to arginine in normal cells which express ASS, whereas ASS(-) tumor cells cannot. A pegylated form of ADI (ADI-PEG20) has been formulated and has shown in vitro and in vivo activity against melanoma and hepatocellular carcinoma. ADI-PEG20 induces apoptosis in melanoma cell lines. However, arginine deprivation can also induce ASS expression in certain melanoma cell lines which can lead to in vitro drug resistance. Phase I and II clinical trials with ADI-PEG20 have been conducted in patients with melanoma and hepatocellular carcinoma, and antitumor activity has been demonstrated in both cancers. This article reviews our laboratory and clinical experience as well as that from others with ADI-PEG20 as an antineoplastic agent. Future direction in utilizing this agent is also discussed.
Collapse
Affiliation(s)
- L Feun
- Hematology/Oncology, University of Miami School of Medicine, 1201 N.W. 16th Street, Miami, FL. 33136, USA.
| | | | | | | | | | | | | |
Collapse
|