1
|
Feng T, Xie F, Lyu Y, Yu P, Chen B, Yu J, Zhang G, To KF, Tsang CM, Kang W. The arginine metabolism and its deprivation in cancer therapy. Cancer Lett 2025; 620:217680. [PMID: 40157492 DOI: 10.1016/j.canlet.2025.217680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Arginine deprivation has emerged as a promising therapeutic strategy in cancer treatment due to the auxotrophy of certain tumors. Many cancers, such as pancreatic, colorectal, and hepatocellular carcinoma, exhibit downregulated argininosuccinate synthetase, making them reliant on external arginine sources. This dependency allows targeted therapies that deplete arginine, inhibiting tumor growth while sparing normal cells. Arginine is crucial for various cellular processes, including protein synthesis and immune function. Its deprivation affects both tumor metabolism and immune responses, potentially enhancing cancer therapy. Studies have explored using enzymes like arginine deiminase and arginase, often modified for increased stability and reduced immunogenicity, to effectively lower arginine levels in the tumor microenvironment. These approaches show promise, particularly in tumors with low argininosuccinate synthetase expression. However, the impact on immune cells and the potential for resistance highlight the need for further research. Combining arginine deprivation with other treatments might improve outcomes, offering a novel approach to combat arginine-dependent cancers.
Collapse
Affiliation(s)
- Tiejun Feng
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales Hospital, The Chinese University of Hong Kong, China
| | - Fuda Xie
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales Hospital, The Chinese University of Hong Kong, China; Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, China; CUHK-Shenzhen Research Institute, Shenzhen, China
| | - Yang Lyu
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales Hospital, The Chinese University of Hong Kong, China
| | - Peiyao Yu
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales Hospital, The Chinese University of Hong Kong, China
| | - Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales Hospital, The Chinese University of Hong Kong, China; Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, China; CUHK-Shenzhen Research Institute, Shenzhen, China
| | - Jun Yu
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, China
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales Hospital, The Chinese University of Hong Kong, China
| | - Chi Man Tsang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales Hospital, The Chinese University of Hong Kong, China.
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales Hospital, The Chinese University of Hong Kong, China; Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, China; CUHK-Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
2
|
Hannemann J, Oliveira-Ferrer L, Goele AK, Mileva Y, Kleinsang F, Röglin A, Witzel I, Müller V, Böger R. L-arginine dependence of breast cancer - molecular subtypes matter. BMC Cancer 2025; 25:546. [PMID: 40140975 PMCID: PMC11948839 DOI: 10.1186/s12885-025-13908-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
L-arginine limits proliferation in highly proliferative tissues. It is a substrate for nitric oxide synthases, arginases; its methylation by protein-L-arginine methyltransferases (PRMTs) leads to asymmetric (ADMA) and symmetric dimethylarginine (SDMA). We measured L-arginine and its metabolites L-ornithine, L-citrulline, ADMA, and SDMA in a prospective cohort of 243 women with primary breast cancer (BC) and their associations with mortality and disease recurrence during 88 (IQR, 82-93) months of follow-up. We quantified these metabolites and expression of genes involved in L-arginine metabolic pathways in MCF-7, BT-474, SK-BR-3, MDA-MB-231, and MDA-MB-468 cells representing ER-positive, HER2-positive, and triple-negative BC compared to MCF-12 A cells. Plasma L-arginine and ADMA concentrations were elevated in 47 patients with recurrent disease and in 34 non-survivors. ADMA was significantly associated with mortality and recurrent disease in Luminal A patients; low L-citrulline was significantly associated with survival in triple-negative BC. In all BC cells except MCF-7, DDAH1 and DDAH2 expression was higher than in MCF-12 A (DDAH1: 32-44 fold, DDAH2: 1.7-4.2 fold; p < 0.05). By contrast, MCF-7 cells showed low DDAH1 and DDAH2, but high PRMT4 and PRMT6 expression and high L-arginine content. BT-474 and MDA-MB-468 cells showed high ARG2 expression and high L-ornithine concentrations, and MDA-MB-468 cells had the highest L-citrulline/L-arginine ratio. In conclusion, regulation of L-arginine metabolic pathways shows a complex and differential pattern between BC subtypes. ADMA is a prognostic biomarker in Luminal A patients; its metabolizing enzyme, DDAH, is highly overexpressed in BC cells. Thus, fingerprinting of L-arginine metabolism may offer novel personalized treatment options within BC subtypes.
Collapse
Affiliation(s)
- Juliane Hannemann
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | | | - Anne Kathrin Goele
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yoana Mileva
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fiona Kleinsang
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Röglin
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Isabell Witzel
- Department of Gynecology, University Hospital Zürich, Zürich, Switzerland
| | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
3
|
Zarrella S, Miranda MR, Covelli V, Restivo I, Novi S, Pepe G, Tesoriere L, Rodriquez M, Bertamino A, Campiglia P, Tecce MF, Vestuto V. Endoplasmic Reticulum Stress and Its Role in Metabolic Reprogramming of Cancer. Metabolites 2025; 15:221. [PMID: 40278350 PMCID: PMC12029571 DOI: 10.3390/metabo15040221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Endoplasmic reticulum (ER) stress occurs when ER homeostasis is disrupted, leading to the accumulation of misfolded or unfolded proteins. This condition activates the unfolded protein response (UPR), which aims to restore balance or trigger cell death if homeostasis cannot be achieved. In cancer, ER stress plays a key role due to the heightened metabolic demands of tumor cells. This review explores how metabolomics can provide insights into ER stress-related metabolic alterations and their implications for cancer therapy. Methods: A comprehensive literature review was conducted to analyze recent findings on ER stress, metabolomics, and cancer metabolism. Studies examining metabolic profiling of cancer cells under ER stress conditions were selected, with a focus on identifying potential biomarkers and therapeutic targets. Results: Metabolomic studies highlight significant shifts in lipid metabolism, protein synthesis, and oxidative stress management in response to ER stress. These metabolic alterations are crucial for tumor adaptation and survival. Additionally, targeting ER stress-related metabolic pathways has shown potential in preclinical models, suggesting new therapeutic strategies. Conclusions: Understanding the metabolic impact of ER stress in cancer provides valuable opportunities for drug development. Metabolomics-based approaches may help identify novel biomarkers and therapeutic targets, enhancing the effectiveness of antitumor therapies.
Collapse
Affiliation(s)
- Salvatore Zarrella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Maria Rosaria Miranda
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Verdiana Covelli
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (V.C.); (M.R.)
| | - Ignazio Restivo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (L.T.)
| | - Sara Novi
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Luisa Tesoriere
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (L.T.)
| | - Manuela Rodriquez
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (V.C.); (M.R.)
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Mario Felice Tecce
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| |
Collapse
|
4
|
Chen Y, Zhang M, Qi Y, Lin Y, Liu S, Deng C, Jiang S, Sun N. Efficient extraction via titanium organic frameworks facilitates in-depth profiling of urinary exosome metabolite fingerprints. Anal Bioanal Chem 2025; 417:1543-1555. [PMID: 39853354 DOI: 10.1007/s00216-025-05741-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025]
Abstract
Urinary exosome metabolite analysis has demonstrated notable advantages in uncovering disease status, yet its potential in decoding the intricacies of clear cell renal cell carcinoma (ccRCC) remains untapped. To address this, a core-shell magnetic titanium organic framework was designed to capture urinary exosomes and assist laser desorption/ionization mass spectrometry (LDI MS) to decipher the exosomal metabolic profile of ccRCC, with high sensitivity, throughput, and speed. A total of 492 urinary exosome metabolite fingerprints (UEMFs) from 176 samples were extracted for exploring the differences between ccRCC and healthy individuals. Leveraging machine learning algorithms, the exosomal metabolic profile was disclosed, achieving accurate differentiation and prediction of ccRCC patients versus healthy individuals, with an accuracy exceeding 97.3%. Furthermore, an optimized algorithm panel comprising five key features demonstrated consistent and high diagnosing accuracy rates of over 94.0% both in the training and blind test sets for ccRCC, underscoring the remarkable effectiveness and superiority of this strategy in ccRCC detection. This study not only refines the LDI MS method for metabolite analysis in urinary exosomes but also introduces a promising technical approach for unraveling the mysteries of ccRCC.
Collapse
Affiliation(s)
- Yijie Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Man Zhang
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Yu Qi
- Department of Urology, Zhongshan Hospital, Zhongshan Hospital Wusong Branch Fudan University, Shanghai, 200032, China
| | - Yiwen Lin
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Shasha Liu
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Chunhui Deng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, China.
| | - Shuai Jiang
- Department of Urology, Zhongshan Hospital, Zhongshan Hospital Wusong Branch Fudan University, Shanghai, 200032, China.
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
5
|
Khan T, Nagarajan M, Kang I, Wu C, Wangpaichitr M. Targeting Metabolic Vulnerabilities to Combat Drug Resistance in Cancer Therapy. J Pers Med 2025; 15:50. [PMID: 39997327 PMCID: PMC11856717 DOI: 10.3390/jpm15020050] [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: 12/20/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
Drug resistance remains a significant barrier to effective cancer therapy. Cancer cells evade treatment by reprogramming their metabolism, switching from glycolysis to oxidative phosphorylation (OXPHOS), and relying on alternative carbon sources such as glutamine. These adaptations not only enable tumor survival but also contribute to immune evasion through mechanisms such as reactive oxygen species (ROS) generation and the upregulation of immune checkpoint molecules like PD-L1. This review explores the potential of targeting metabolic weaknesses in drug-resistant cancers to enhance therapeutic efficacy. Key metabolic pathways involved in resistance, including glycolysis, glutamine metabolism, and the kynurenine pathway, are discussed. The combination of metabolic inhibitors with immune checkpoint inhibitors (ICIs), particularly anti-PD-1/PD-L1 therapies, represents a promising approach to overcoming both metabolic and immune evasion mechanisms. Clinical trials combining metabolic and immune therapies have shown early promise, but further research is needed to optimize treatment combinations and identify biomarkers for patient selection. In conclusion, targeting cancer metabolism in combination with immune checkpoint blockade offers a novel approach to overcoming drug resistance, providing a potential pathway to improved outcomes in cancer therapy. Future directions include personalized treatments based on tumor metabolic profiles and expanding research to other tumor types.
Collapse
Affiliation(s)
- Taranatee Khan
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL 33125, USA; (T.K.); (M.N.); (I.K.); (C.W.)
| | - Manojavan Nagarajan
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL 33125, USA; (T.K.); (M.N.); (I.K.); (C.W.)
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Irene Kang
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL 33125, USA; (T.K.); (M.N.); (I.K.); (C.W.)
- South Florida VA Foundation for Research and Education, Miami, FL 33125, USA
| | - Chunjing Wu
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL 33125, USA; (T.K.); (M.N.); (I.K.); (C.W.)
| | - Medhi Wangpaichitr
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL 33125, USA; (T.K.); (M.N.); (I.K.); (C.W.)
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- South Florida VA Foundation for Research and Education, Miami, FL 33125, USA
- Department of Surgery, Division of Thoracic Surgery, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
6
|
Carpentier J, Freitas M, Morales V, Bianchi K, Bomalaski J, Szlosarek P, Martin SA. Overcoming resistance to arginine deprivation therapy using GC7 in pleural mesothelioma. iScience 2025; 28:111525. [PMID: 39758821 PMCID: PMC11699351 DOI: 10.1016/j.isci.2024.111525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/12/2024] [Accepted: 11/29/2024] [Indexed: 01/07/2025] Open
Abstract
Pleural mesothelioma is a highly chemotherapy-resistant cancer. Approximately 50% of mesotheliomas do not express argininosuccinate synthetase 1 (ASS1), the rate-limiting enzyme in arginine biosynthesis, making arginine depletion with pegylated arginine deiminase (ADI-PEG20) an attractive therapeutic strategy. We investigated whether combinatory treatment composed of ADI-PEG20 and polyamine inhibitors constitutes a promising novel therapeutic strategy to overcome ADI-PEG20 resistance in mesothelioma patients. Treatment of ADI-PEG20-resistant cell lines with a range of different polyamine inhibitors demonstrated that ADI-PEG20-resistant cell lines were highly sensitive to the spermidine-analog GC7. We observed a synergistic effect of GC7 and ADI-PEG20 in both ADI-PEG20-sensitive and ADI-PEG20-resistant cell lines. Metabolomic analysis revealed that sensitivity to GC7 is due to inhibition of the Tricarboxylic (TCA) cycle. Significantly, combination of GC7 and ADI-PEG20 prevented the emergence of resistant cells in vitro. Taken together, we have identified the therapeutic potential of combinatorial treatment of ADI-PEG20 with GC7 for mesothelioma management.
Collapse
Affiliation(s)
- Josephine Carpentier
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Marta Freitas
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Valle Morales
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Katiuscia Bianchi
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | | | - Peter Szlosarek
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Sarah A. Martin
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
7
|
Hari K, Squitti R, Bosco J, Jayaraman V, Krishna K, Pal A, De Luca A, Di Veroli L, Mastromoro G, Rizzo G, Tondolo V, Rongioletti M. Classification of colorectal cancer patients based on serum micronutrients: An exploratory investigation. J Trace Elem Med Biol 2024; 86:127564. [PMID: 39579716 DOI: 10.1016/j.jtemb.2024.127564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is a growing global health challenge with a multifactorial etiology encompassing genetic susceptibility, nutrition, and inflammation in the bowel. OBJECTIVE To examine micronutrient status in CRC patients undergoing CRC resection. DESIGN We performed a case-control study including 13 consecutive CRC patients and 10 healthy controls (CTRL) comparing the serum levels of 29 micronutrients, namely Copper, Zinc, Selenium, Chromium, Manganese, Carnitine, Choline, Inositol, Methylmalonic acid (MMA), Vitamin (Vit) B1, Vit B2, Vit B3, Vit B5, Vit B6, Vit C, Vit A, Vit D3, Vit E, Vit K1, Vit K2 and the amino acids Serine, Valine, Leucine, Isoleucine, Asparagine, Glutamine, Arginine, Citrulline and Cysteine. RESULTS After considering the effect of age and sex, copper, arginine, and cysteine were increased, while zinc, selenium, chromium, Vit B1, Vit K1, and Vit A were decreased in CRC patients in comparison with CTRL. Zinc levels perfectly predicted the diagnosis of CRC, and were associated with lymph nodes (pN), of the pTNM staging. Copper levels in serum were strongly associated with the pathological pTNM staging of CRC. CONCLUSION Though this is a preliminary study that needs confirmation with a larger longitudinal cohort, our results show that serum micronutrients are linked to tumor growth, likely caused by increased demand from tumor tissue associated with an aberrant cell proliferation and changes in the antioxidant function.
Collapse
Affiliation(s)
| | - Rosanna Squitti
- Department of Laboratory Science, Research and Development Division, Ospedale Isola Tiberina-Gemelli Isola, Rome 00186, Italy; Department of Theoretical and Applied Sciences, eCampus University, Como, Novedrate, Italy.
| | - Jophi Bosco
- Vibrant America LLC, San Carlos, CA, United States of America
| | | | - Karthik Krishna
- Vibrant Sciences LLC, San Carlos, CA, United States of America
| | - Amit Pal
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Kalyani 741245, India.
| | - Anastasia De Luca
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, Roma 00133, Italy
| | - Laura Di Veroli
- Department of Laboratory Science, Research and Development Division, Ospedale Isola Tiberina-Gemelli Isola, Rome 00186, Italy
| | - Gioia Mastromoro
- Department of Laboratory Science, Research and Development Division, Ospedale Isola Tiberina-Gemelli Isola, Rome 00186, Italy
| | - Gianluca Rizzo
- Digestive and Colorectal Surgery, Ospedale Isola Tiberina-Gemelli Isola, Rome 00186, Italy; Digestive Surgery Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
| | - Vincenzo Tondolo
- Digestive and Colorectal Surgery, Ospedale Isola Tiberina-Gemelli Isola, Rome 00186, Italy; Digestive Surgery Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
| | - Mauro Rongioletti
- Department of Laboratory Science, Research and Development Division, Ospedale Isola Tiberina-Gemelli Isola, Rome 00186, Italy.
| |
Collapse
|
8
|
Chen C, Jiang X, Zhao Z. Inhibition or promotion, the potential role of arginine metabolism in immunotherapy for colorectal cancer. ALL LIFE 2023. [DOI: 10.1080/26895293.2022.2163306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Chengyang Chen
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Xia Jiang
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Zengren Zhao
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| |
Collapse
|
9
|
Liu D, Wang Y, Li X, Wang Y, Zhang Z, Wang Z, Zhang X. Participation of protein metabolism in cancer progression and its potential targeting for the management of cancer. Amino Acids 2023; 55:1223-1246. [PMID: 37646877 DOI: 10.1007/s00726-023-03316-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 08/11/2023] [Indexed: 09/01/2023]
Abstract
Cancer malignancies may broadly be described as heterogeneous disorders manifested by uncontrolled cellular growth/division and proliferation. Tumor cells utilize metabolic reprogramming to accomplish the upregulated nutritional requirements for sustaining their uncontrolled growth, proliferation, and survival. Metabolic reprogramming also called altered or dysregulated metabolism undergoes modification in normal metabolic pathways for anabolic precursor's generation that serves to continue biomass formation that sustains the growth, proliferation, and survival of carcinogenic cells under a nutrition-deprived microenvironment. A wide range of dysregulated/altered metabolic pathways encompassing different metabolic regulators have been described; however, the current review is focused to explain deeply the metabolic pathways modifications inducing upregulation of proteins/amino acids metabolism. The essential modification of various metabolic cycles with their consequent outcomes meanwhile explored promising therapeutic targets playing a pivotal role in metabolic regulation and is successfully employed for effective target-specific cancer treatment. The current review is aimed to understand the metabolic reprogramming of different proteins/amino acids involved in tumor progression along with potential therapeutic perspective elucidating targeted cancer therapy via these targets.
Collapse
Affiliation(s)
- Dalong Liu
- Department of Orthopedics, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| | - Yun Wang
- Department of Thoracic Surgery, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| | - Xiaojiang Li
- Department of Orthopedics, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| | - Yan Wang
- Department of Neurosurgery, People's Hospital of Jilin City, Jilin, 136200, China
| | - Zhiqiang Zhang
- Department of Orthopedics, Baishan Hospital of Traditional Chinese Medicine, Baishan, 134300, China
| | - Zhifeng Wang
- Department of Traditional Chinese Medicine, Changchun Chaoyang District Hospital of Traditional Chinese Medicine, Changchun, 130000, China
| | - Xudong Zhang
- Department of Brain Surgery, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China.
| |
Collapse
|
10
|
Zhang X, Wang C, Li C, Zhao H. Development and internal validation of nomograms based on plasma metabolites to predict non-small cell lung cancer risk in smoking and nonsmoking populations. Thorac Cancer 2023; 14:1719-1731. [PMID: 37150808 PMCID: PMC10290921 DOI: 10.1111/1759-7714.14917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/09/2023] Open
Abstract
BACKGROUND Lung cancer has significantly higher incidence and mortality rates worldwide. In this study, we analyzed the metabolic profiles of non-small cell lung cancer (NSCLC) patients and constructed prediction models for smokers and nonsmokers with internal validation. METHODS Plasma was collected from all patients enrolled for metabolic profiling by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The total population was divided into two groups according to smoking or not. Statistical analysis of metabolites was performed separately for each group and prediction models were constructed. RESULTS A total of 1723 patients (1109 NSCLC patients and 614 healthy controls) were enrolled from the affiliated hospital during 2018 to 2021. After grouping by smoking history, each group was statistically analyzed and prediction models were constructed, which resulted in eight indicators (propionylcarnitine, arginine, citrulline, etc.) significantly associated with lung cancer risk for smokers and eight indicators (dodecanoylcarnitine, hydroxybutyrylcarnitine, asparagine, etc.) for nonsmokers (p < 0.05). The smoker model indicated an AUC of 0.860 in the training set and 0.850 in the validation set. The nonsmoker model showed an AUC of 0.783 in the training set and 0.762 in the validation set. Further calibration tests for both models indicated excellent goodness-of-fit results. CONCLUSIONS In this study, we found a series of metabolites significantly associated with lung cancer incidence and constructed respectively prediction models for NSCLC risk in smokers and nonsmokers, with internal validation to confirm the efficiency to discriminate lung cancer risk in both smoking and nonsmoking states.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Health Examination CenterThe Second Hospital of Dalian Medical UniversityDalianLiaoningChina
- Department of Respiratory MedicineThe Second Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Cuicui Wang
- Department of Health Examination CenterThe Second Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Chenwei Li
- Department of Respiratory MedicineThe Second Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Hui Zhao
- Department of Health Examination CenterThe Second Hospital of Dalian Medical UniversityDalianLiaoningChina
| |
Collapse
|
11
|
Fantone S, Ermini L, Piani F, Di Simone N, Barbaro G, Giannubilo SR, Gesuita R, Tossetta G, Marzioni D. Downregulation of argininosuccinate synthase 1 (ASS1) is associated with hypoxia in placental development. Hum Cell 2023; 36:1190-1198. [PMID: 36995581 DOI: 10.1007/s13577-023-00901-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
Argininosuccinate synthase (ASS1) is involved in nitric oxide production, which has a key role in placental development improving pregnancy outcomes. Syncytiotrophoblast and extravillous trophoblast differentiations are milestones of placental development and their impairment can cause pathologies, such as preeclampsia (PE) and fetal growth restriction (FGR). Immunohistochemistry and Western blotting were used to localize and quantify ASS1 in first trimester (8.2 ± 1.8 weeks), third trimester (38.6 ± 1.1 weeks), and PE (36.3 ± 1.5 weeks) placentas. In addition, cell cultures were used to evaluate ASS1 expression under hypoxic conditions and the syncytialization process. Our data showed that ASS1 is localized in the villous cytotrophoblast of first trimester, third trimester, and PE placentas, while the villous cytotrophoblast adjacent to the extravillous trophoblast of cell columns as well as the extravillous trophoblast were negative for ASS1 in first trimester placentas. In addition, ASS1 was decreased in third trimester compared to the first trimester placentas (p = 0.003) and no differences were detected between third trimester and PE placentas. Moreover, ASS1 expression was decreased in hypoxic conditions and syncytialized cells compared to those not syncytialized. In conclusion, we suggest that the expression of ASS1 in villous cytotrophoblast is related to maintaining proliferative phenotype, while ASS1 absence may be involved in promoting the differentiation of villous cytotrophoblast in extravillous cytotrophoblast of cell columns in first trimester placentas.
Collapse
Affiliation(s)
- Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Leonardo Ermini
- Department of Life Science, University of Siena, 53100, Siena, Italy
| | - Federica Piani
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40128, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, 40138, Bologna, Italy
| | - Nicoletta Di Simone
- Department of Biomedical Sciences, Humanitas University, 20072, Milan, Italy
- IRCCS Humanitas Research Hospital, 20089, Milan, Italy
| | - Greta Barbaro
- Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Stefano Raffaele Giannubilo
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, 60126, Ancona, Italy
| | - Rosaria Gesuita
- Centre of Epidemiology and Biostatistics, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy.
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, 60126, Ancona, Italy.
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| |
Collapse
|
12
|
Afshinpour M, Mahdiuni H. Arginine transportation mechanism through cationic amino acid transporter 1: insights from molecular dynamics studies. J Biomol Struct Dyn 2023; 41:13580-13594. [PMID: 36762692 DOI: 10.1080/07391102.2023.2175374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/28/2023] [Indexed: 02/11/2023]
Abstract
Metabolic and signaling mechanisms in mammalian cells are facilitated by the transportation of L-arginine (Arg) across the plasma membrane through cationic amino acid transporter (CAT) proteins. Due to a lack of argininosuccinate synthase (ASS) activity in various tumor cells such as acute myeloid leukemia, acute lymphocytic leukemia, and chronic lymphocytic leukemia, these tumor entities are arginine-auxotrophic and therefore depend on the uptake of the amino acid arginine. Cationic amino acid transporter-1 (CAT-1) is the leading arginine importer expressed in the aforementioned tumor entities. Hence, in the present study, to investigate the transportation mechanism of arginine in CAT-1, we performed molecular dynamics (MD) simulation methods on the modeled human CAT-1. The MM-PBSA approach was conducted to determine the critical residues interacting with arginine within the corresponding binding site of CAT-1. In addition, we found out that the water molecules have the leading role in forming the transportation channel within CAT-1. The conductive structure of CAT-1 was formed only when the water molecules were continuously distributed across the channel. Steered molecular dynamics (SMD) simulation approach showed various energy barriers against arginine transportation through CAT-1, especially while crossing the bottlenecks of the related channel. These findings at the molecular level might shed light on identifying the crucial amino acids in the binding of arginine to eukaryotic CATs and also provide fundamental insights into the arginine transportation mechanisms through CAT-1. Understanding the transportation mechanism of arginine is essential to developing CAT-1 blockers, which can be potential medications for some types of cancers.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Maral Afshinpour
- Bioinformatics Lab., Department of Biology, School of Sciences, Razi University, Kermanshah, Iran
| | - Hamid Mahdiuni
- Bioinformatics Lab., Department of Biology, School of Sciences, Razi University, Kermanshah, Iran
| |
Collapse
|
13
|
Rebsamen M, Girardi E, Sedlyarov V, Scorzoni S, Papakostas K, Vollert M, Konecka J, Guertl B, Klavins K, Wiedmer T, Superti-Furga G. Gain-of-function genetic screens in human cells identify SLC transporters overcoming environmental nutrient restrictions. Life Sci Alliance 2022; 5:e202201404. [PMID: 36114003 PMCID: PMC9481932 DOI: 10.26508/lsa.202201404] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
Abstract
Solute carrier (SLC) transporters control fluxes of nutrients and metabolites across membranes and thereby represent a critical interface between the microenvironment and cellular and subcellular metabolism. Because of substantial functional overlap, the interplay and relative contributions of SLCs in response to environmental stresses remain poorly elucidated. To infer functional relationships between SLCs and metabolites, we developed a strategy to identify SLCs able to sustain cell viability and proliferation under growth-limiting concentrations of essential nutrients. One-by-one depletion of 13 amino acids required for cell proliferation enabled gain-of-function genetic screens using a SLC-focused CRISPR/Cas9-based transcriptional activation approach to uncover transporters relieving cells from growth-limiting metabolic bottlenecks. Among the transporters identified, we characterized the cationic amino acid transporter SLC7A3 as a gene that, when up-regulated, overcame low availability of arginine and lysine by increasing their uptake, whereas SLC7A5 was able to sustain cellular fitness upon deprivation of several neutral amino acids. Moreover, we identified metabolic compensation mediated by the glutamate/aspartate transporters SLC1A2 and SLC1A3 under glutamine-limiting conditions. Overall, this gain-of-function approach using human cells uncovered functional transporter-nutrient relationships and revealed that transport activity up-regulation may be sufficient to overcome environmental metabolic restrictions.
Collapse
Affiliation(s)
- Manuele Rebsamen
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Enrico Girardi
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Vitaly Sedlyarov
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Stefania Scorzoni
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Konstantinos Papakostas
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Manuela Vollert
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Justyna Konecka
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Bettina Guertl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Kristaps Klavins
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Tabea Wiedmer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
14
|
Kumar R, Mishra A, Gautam P, Feroz Z, Vijayaraghavalu S, Likos EM, Shukla GC, Kumar M. Metabolic Pathways, Enzymes, and Metabolites: Opportunities in Cancer Therapy. Cancers (Basel) 2022; 14:5268. [PMID: 36358687 PMCID: PMC9656396 DOI: 10.3390/cancers14215268] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/09/2022] [Accepted: 10/19/2022] [Indexed: 07/30/2023] Open
Abstract
Metabolic reprogramming enables cancer cells to proliferate and produce tumor biomass under a nutrient-deficient microenvironment and the stress of metabolic waste. A cancer cell adeptly undergoes a variety of adaptations in metabolic pathways and differential expression of metabolic enzyme genes. Metabolic adaptation is mainly determined by the physiological demands of the cancer cell of origin and the host tissue. Numerous metabolic regulators that assist cancer cell proliferation include uncontrolled anabolism/catabolism of glucose metabolism, fatty acids, amino acids metabolism, nucleotide metabolism, tumor suppressor genes, microRNAs, and many regulatory enzymes and genes. Using this paradigm, we review the current understanding of metabolic reprogramming in tumors and discuss the new strategies of cancer metabolomics that can be tapped into for cancer therapeutics.
Collapse
Affiliation(s)
- Rishabh Kumar
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, UP, India
| | - Anurag Mishra
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, UP, India
| | - Priyanka Gautam
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, UP, India
| | - Zainab Feroz
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, UP, India
| | | | - Eviania M. Likos
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, Cleveland State University, 2121 Euclid Avenue, Cleveland, OH 44115, USA
| | - Girish C. Shukla
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, Cleveland State University, 2121 Euclid Avenue, Cleveland, OH 44115, USA
| | - Munish Kumar
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, UP, India
| |
Collapse
|
15
|
Hou X, Chen S, Zhang P, Guo D, Wang B. Targeted Arginine Metabolism Therapy: A Dilemma in Glioma Treatment. Front Oncol 2022; 12:938847. [PMID: 35898872 PMCID: PMC9313538 DOI: 10.3389/fonc.2022.938847] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/20/2022] [Indexed: 11/29/2022] Open
Abstract
Efforts in the treatment of glioma which is the most common primary malignant tumor of the central nervous system, have not shown satisfactory results despite a comprehensive treatment model that combines various treatment methods, including immunotherapy. Cellular metabolism is a determinant of the viability and function of cancer cells as well as immune cells, and the interplay of immune regulation and metabolic reprogramming in tumors has become an active area of research in recent years. From the perspective of metabolism and immunity in the glioma microenvironment, we elaborated on arginine metabolic reprogramming in glioma cells, which leads to a decrease in arginine levels in the tumor microenvironment. Reduced arginine availability significantly inhibits the proliferation, activation, and function of T cells, thereby promoting the establishment of an immunosuppressive microenvironment. Therefore, replenishment of arginine levels to enhance the anti-tumor activity of T cells is a promising strategy for the treatment of glioma. However, due to the lack of expression of argininosuccinate synthase, gliomas are unable to synthesize arginine; thus, they are highly dependent on the availability of arginine in the extracellular environment. This metabolic weakness of glioma has been utilized by researchers to develop arginine deprivation therapy, which ‘starves’ tumor cells by consuming large amounts of arginine in circulation. Although it has shown good results, this treatment modality that targets arginine metabolism in glioma is controversial. Exploiting a suitable strategy that can not only enhance the antitumor immune response, but also “starve” tumor cells by regulating arginine metabolism to cure glioma will be promising.
Collapse
|
16
|
Du Z, Li T, Huang J, Chen Y, Chen C. Arginase: Mechanisms and Clinical Application in Hematologic Malignancy. Front Oncol 2022; 12:905893. [PMID: 35814439 PMCID: PMC9260017 DOI: 10.3389/fonc.2022.905893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Compared to normal tissues and cells, the metabolic patterns of tumor illnesses are more complex, and there are hallmarks of metabolic reprogramming in energy metabolism, lipid metabolism, and amino acid metabolism. When tumor cells are in a state of fast growth, they are susceptible to food shortage, resulting in growth suppression. Using this metabolic sensitivity of tumor cells to construct amino acid consumption therapy does not harm the function of normal cells, which is the focus of metabolic therapy research at the moment. As a non-essential amino acid, arginine is involved in numerous crucial biological processes, including the signaling system, cell proliferation, and material metabolism. Rapidly dividing tumor cells are more likely to be deficient in arginine; hence, utilizing arginase to consume arginine can suppress tumor growth. Due to the absence of arginine succinate synthase, arginine succinate lyase, and ornithine carbamoyl transferase in some blood tumors, arginases may be employed to treat blood tumors. By investigating the mechanism of arginase treatment and the mechanism of drug resistance in greater depth, arginase treatment becomes more successful in hematological cancers and a new anti-cancer agent in clinical practice.
Collapse
Affiliation(s)
- Zefan Du
- Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Tianwen Li
- Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Junbin Huang
- Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Yun Chen
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- *Correspondence: Yun Chen, ; Chun Chen,
| | - Chun Chen
- Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
- *Correspondence: Yun Chen, ; Chun Chen,
| |
Collapse
|
17
|
Liu N, Shi F, Yang L, Liao W, Cao Y. Oncogenic viral infection and amino acid metabolism in cancer progression: Molecular insights and clinical implications. Biochim Biophys Acta Rev Cancer 2022; 1877:188724. [DOI: 10.1016/j.bbcan.2022.188724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/15/2022] [Accepted: 03/15/2022] [Indexed: 02/08/2023]
|
18
|
Wu X, Liu Y, Ao H, Yang P, Zhu Z. A metabolomics strategy to identify potential biomarkers associated with human laryngeal cancer based on dried blood spot mass spectrometry approach. Medicine (Baltimore) 2022; 101:e28820. [PMID: 35212275 PMCID: PMC8878607 DOI: 10.1097/md.0000000000028820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/03/2022] [Accepted: 01/25/2022] [Indexed: 01/04/2023] Open
Abstract
ABSTRACT Laryngeal cancer (LC) as one of common malignant tumors in the head and neck region accounted for 1% to 5% of new cancer cases and was ranked as the third otolaryngology cancer. However, some patients with LC were diagnosed at the advanced stage, which can cause delayed diagnosis and treatment. It is an urgent task to seek effective biomarkers for the early diagnosis of LC aimed at alleviating suffering.A combination of dried blood spot sampling and direct infusion mass spectrometry technology was applied to 39 patients with LC and 53 healthy individuals. Multiple algorithms towards 93 metabolites including amino acids and carnitine/acylcarnitines were run for selecting differential metabolites. Furthermore, leave-one-out cross-validation method was used to evaluate diagnostic performance of selected metabolite biomarkers.A biomarker panel consisting of arginine, proline, hexacosanoic carnitine, ornithine /citrulline, and 3-hydroxy-octadecenoylcarnitine exhibited potential to distinguish patients with LC from healthy individuals, with a sensitivity of 0.8974 and a specificity of 0.8302 in leave-one-out cross-validation model.The metabolomic analysis of LC patients is beneficial to screen disease-associated biomarkers and develop new diagnostic approaches.
Collapse
Affiliation(s)
- Xue Wu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Research Centre for Southern Deer at Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Research Centre for Medical data at Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yongting Liu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Huaixuan Ao
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Research Centre for Medical data at Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Peng Yang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Research Centre for Medical data at Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zhitu Zhu
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
19
|
Fouda AY, Eldahshan W, Xu Z, Lemtalsi T, Shosha E, Zaidi SA, Abdelrahman AA, Cheng PNM, Narayanan SP, Caldwell RW, Caldwell RB. Preclinical investigation of Pegylated arginase 1 as a treatment for retina and brain injury. Exp Neurol 2022; 348:113923. [PMID: 34780773 PMCID: PMC9122100 DOI: 10.1016/j.expneurol.2021.113923] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/12/2021] [Accepted: 11/08/2021] [Indexed: 02/03/2023]
Abstract
Arginase 1 (A1) is the enzyme that hydrolyzes the amino acid, L-arginine, to ornithine and urea. We have previously shown that A1 deletion worsens retinal ischemic injury, suggesting a protective role of A1. In this translational study, we aimed to study the utility of systemic pegylated A1 (PEG-A1, recombinant human arginase linked to polyethylene glycol) treatment in mouse models of acute retinal and brain injury. Cohorts of WT mice were subjected to retinal ischemia-reperfusion (IR) injury, traumatic optic neuropathy (TON) or brain cerebral ischemia via middle cerebral artery occlusion (MCAO) and treated with intraperitoneal injections of PEG-A1 or vehicle (PEG only). Drug penetration into retina and brain tissues was measured by western blotting and immunolabeling for PEG. Neuroprotection was measured in a blinded fashion by quantitation of NeuN (neuronal marker) immunolabeling of retina flat-mounts and brain infarct area using triphenyl tetrazolium chloride (TTC) staining. Furthermore, ex vivo retina explants and in vitro retina neuron cultures were subjected to oxygen-glucose deprivation (OGD) followed by reoxygenation (R) and treated with PEG-A1. PEG-A1 given systemically did not cross the intact blood-retina/brain barriers in sham controls but reached the retina and brain after injury. PEG-A1 provided neuroprotection after retinal IR injury, TON and cerebral ischemia. PEG-A1 treatment was also neuroprotective in retina explants subjected to OGD/R but did not improve survival in retinal neuronal cultures exposed to OGD/R. In summary, systemic PEG-A1 administration is neuroprotective and provides an excellent route to deliver the drug to the retina and the brain after acute injury.
Collapse
Affiliation(s)
- Abdelrahman Y Fouda
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Wael Eldahshan
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Zhimin Xu
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Tahira Lemtalsi
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Esraa Shosha
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Syed Ah Zaidi
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Ammar A Abdelrahman
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Paul Ning-Man Cheng
- Bio-cancer Treatment International, 511-513, Bioinformatics Building, Hong Kong Science Park, Tai Po, Hong Kong, China
| | - S Priya Narayanan
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Cellular Biology & Anatomy, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA; Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, GA, United States
| | - R William Caldwell
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Cellular Biology & Anatomy, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
20
|
Chen Y, Li Y. Metabolic reprogramming and immunity in cancer. CANCER IMMUNOLOGY AND IMMUNOTHERAPY 2022:137-196. [DOI: 10.1016/b978-0-12-823397-9.00006-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
21
|
El-Mais N, Fakhoury I, Al Haddad M, Nohra S, Abi-Habib R, El-Sibai M. Human Recombinant Arginase I [HuArgI(Co)-PEG5000]-Induced Arginine Depletion Inhibits Pancreatic Cancer Cell Migration and Invasion Through Autophagy. Pancreas 2021; 50:1187-1194. [PMID: 34714283 DOI: 10.1097/mpa.0000000000001891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Pancreatic cancer is one of the most aggressive solid cancers and the fourth leading cause of cancer death in men and women. We previously showed that arginine depletion, using arginase I [HuArgI(Co)-PEG5000], selectively triggers cell death by autophagy in PANC-1 pancreatic cancer cells. The mechanism of action of [HuArgI(Co)-PEG5000], however, has remained poorly understood. In this study, we investigated the effects of arginine depletion on PANC-1 cell migration, adhesion, and invasion and determined the main molecular targets, which mediate PANC-1 cell response to treatment with HuArgI(Co)-PEG5000. METHODS This was done through examining 2-dimensional (2D) cell motility assays (wound healing and time lapse), cell adhesion, and cell invasion assays, as well as immunostaining for focal adhesions and invadopodia in cells without or with the treatment with arginase. RESULTS We demonstrate that arginine depletion decreases PANC-1 2D cell migration, adhesion, and 3D invasion. Moreover, our data suggest that these effects are mediated by autophagy and subsequent decrease in the activation of members of Ras homolog gene family (Rho) GTPase family. CONCLUSIONS Altogether, these findings uncover the mechanism of action of [HuArgI(Co)-PEG5000] and highlight the promising and selective anticancer potential for arginine depletion in the treatment of pancreatic cancer cells.
Collapse
Affiliation(s)
- Nour El-Mais
- From the Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Isabelle Fakhoury
- From the Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Maria Al Haddad
- From the Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Sarah Nohra
- Department of Biosciences, School of Science and Technology, Università degli Studi di Milano, Milan, Italy
| | - Ralph Abi-Habib
- From the Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Mirvat El-Sibai
- From the Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| |
Collapse
|
22
|
Zhang Q, Yang X, Liu H. Extracellular Vesicles in Cancer Metabolism: Implications for Cancer Diagnosis and Treatment. Technol Cancer Res Treat 2021; 20:15330338211037821. [PMID: 34427131 PMCID: PMC8388228 DOI: 10.1177/15330338211037821] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Metabolic reprogramming is one of the most common characteristics of cancer cells. The metabolic alterations of glucose, amino acids and lipids can support the aggressive phenotype of cancer cells. Exosomes, a kind of extracellular vesicles, participate in the intercellular communication through transferring bioactive molecules. Increasing evidence has demonstrated that enzymes, metabolites and non-coding RNAs in exosomes are responsible for the metabolic alteration of cancer cells. In this review, we summarize the past and recent findings of exosomes in altering cancer metabolism and elaborate on the role of the specific enzymes, metabolites and non-coding RNAs transferred by exosomes. Moreover, we give evidence of the role of exosomes in cancer diagnosis and treatment. Finally, we discuss the existing problems in the study and application of exosomes in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiangling Yang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huanliang Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
23
|
Wangpaichitr M, Theodoropoulos G, Nguyen DJM, Wu C, Spector SA, Feun LG, Savaraj N. Cisplatin Resistance and Redox-Metabolic Vulnerability: A Second Alteration. Int J Mol Sci 2021; 22:7379. [PMID: 34298999 PMCID: PMC8304747 DOI: 10.3390/ijms22147379] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 01/17/2023] Open
Abstract
The development of drug resistance in tumors is a major obstacle to effective cancer chemotherapy and represents one of the most significant complications to improving long-term patient outcomes. Despite early positive responsiveness to platinum-based chemotherapy, the majority of lung cancer patients develop resistance. The development of a new combination therapy targeting cisplatin-resistant (CR) tumors may mark a major improvement as salvage therapy in these patients. The recent resurgence in research into cellular metabolism has again confirmed that cancer cells utilize aerobic glycolysis ("the Warburg effect") to produce energy. Hence, this observation still remains a characteristic hallmark of altered metabolism in certain cancer cells. However, recent evidence promotes another concept wherein some tumors that acquire resistance to cisplatin undergo further metabolic alterations that increase tumor reliance on oxidative metabolism (OXMET) instead of glycolysis. Our review focuses on molecular changes that occur in tumors due to the relationship between metabolic demands and the importance of NAD+ in redox (ROS) metabolism and the crosstalk between PARP-1 (Poly (ADP ribose) polymerase-1) and SIRTs (sirtuins) in CR tumors. Finally, we discuss a role for the tumor metabolites of the kynurenine pathway (tryptophan catabolism) as effectors of immune cells in the tumor microenvironment during acquisition of resistance in CR cells. Understanding these concepts will form the basis for future targeting of CR cells by exploiting redox-metabolic changes and their consequences on immune cells in the tumor microenvironment as a new approach to improve overall therapeutic outcomes and survival in patients who fail cisplatin.
Collapse
Affiliation(s)
- Medhi Wangpaichitr
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service (151), Miami, FL 33125, USA; (G.T.); (D.J.M.N.); (C.W.); (S.A.S.)
- Department of Surgery, Cardiothoracic Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - George Theodoropoulos
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service (151), Miami, FL 33125, USA; (G.T.); (D.J.M.N.); (C.W.); (S.A.S.)
| | - Dan J. M. Nguyen
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service (151), Miami, FL 33125, USA; (G.T.); (D.J.M.N.); (C.W.); (S.A.S.)
| | - Chunjing Wu
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service (151), Miami, FL 33125, USA; (G.T.); (D.J.M.N.); (C.W.); (S.A.S.)
| | - Sydney A. Spector
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service (151), Miami, FL 33125, USA; (G.T.); (D.J.M.N.); (C.W.); (S.A.S.)
| | - Lynn G. Feun
- Department of Medicine, Hematology/Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (L.G.F.); (N.S.)
| | - Niramol Savaraj
- Department of Medicine, Hematology/Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (L.G.F.); (N.S.)
- Department of Veterans Affairs, Miami VA Healthcare System, Hematology/Oncology, 1201 NW 16 Street, Room D1010, Miami, FL 33125, USA
| |
Collapse
|
24
|
Hsu SC, Chen CL, Cheng ML, Chu CY, Changou CA, Yu YL, Yeh SD, Kuo TC, Kuo CC, Chuu CP, Li CF, Wang LH, Chen HW, Yen Y, Ann DK, Wang HJ, Kung HJ. Arginine starvation elicits chromatin leakage and cGAS-STING activation via epigenetic silencing of metabolic and DNA-repair genes. Theranostics 2021; 11:7527-7545. [PMID: 34158865 PMCID: PMC8210599 DOI: 10.7150/thno.54695] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 05/13/2021] [Indexed: 12/15/2022] Open
Abstract
Rationale: One of the most common metabolic defects in cancers is the deficiency in arginine synthesis, which has been exploited therapeutically. Yet, challenges remain, and the mechanisms of arginine-starvation induced killing are largely unclear. Here, we sought to demonstrate the underlying mechanisms by which arginine starvation-induced cell death and to develop a dietary arginine-restriction xenograft model to study the in vivo effects. Methods: Multiple castration-resistant prostate cancer cell lines were treated with arginine starvation followed by comprehensive analysis of microarray, RNA-seq and ChIP-seq were to identify the molecular and epigenetic pathways affected by arginine starvation. Metabolomics and Seahorse Flux analyses were used to determine the metabolic profiles. A dietary arginine-restriction xenograft mouse model was developed to assess the effects of arginine starvation on tumor growth and inflammatory responses. Results: We showed that arginine starvation coordinately and epigenetically suppressed gene expressions, including those involved in oxidative phosphorylation and DNA repair, resulting in DNA damage, chromatin-leakage and cGAS-STING activation, accompanied by the upregulation of type I interferon response. We further demonstrated that arginine starvation-caused depletion of α-ketoglutarate and inactivation of histone demethylases are the underlying causes of epigenetic silencing. Significantly, our dietary arginine-restriction model showed that arginine starvation suppressed prostate cancer growth in vivo, with evidence of enhanced interferon responses and recruitment of immune cells. Conclusions: Arginine-starvation induces tumor cell killing by metabolite depletion and epigenetic silencing of metabolic genes, leading to DNA damage and chromatin leakage. The resulting cGAS-STING activation may further enhance these killing effects.
Collapse
Affiliation(s)
- Sheng-Chieh Hsu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Chia-Lin Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Mei-Ling Cheng
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Clinical Metabolomics Core Laboratory, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
| | - Cheng-Ying Chu
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- CRISPR Gene Targeting Core Lab, Taipei Medical University, Taipei 110, Taiwan
| | - Chun A. Changou
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
- The PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan
| | - Yen-Ling Yu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Shauh-Der Yeh
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Urology, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Tse-Chun Kuo
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Cheng-Chin Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Chien-Feng Li
- Department of Pathology, Chi Mei Medical Center, Tainan City 73657, Taiwan
- Institute of Cancer Research, National Health Research Institutes, Miaoli County 35053, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Lu-Hai Wang
- Chinese Medicine Research Center and Institute of Integrated Medicine, China Medical University, Taichung 40402, Taiwan
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Yun Yen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - David K. Ann
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Hung-Jung Wang
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan
- Doctoral Degree Program in Translational Medicine, Tzu Chi University and Academia Sinica, Hualien 97004, Taiwan
| | - Hsing-Jien Kung
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County 35053, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
25
|
Guo Y, Lv B, Liu R, Dai Z, Zhang F, Liang Y, Yu B, Zeng D, Lv XB, Zhang Z. Role of LncRNAs in regulating cancer amino acid metabolism. Cancer Cell Int 2021; 21:209. [PMID: 33849550 PMCID: PMC8045299 DOI: 10.1186/s12935-021-01926-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/07/2021] [Indexed: 12/17/2022] Open
Abstract
The metabolic change of tumor cells is an extremely complicated process that involves the intersection and integration of various signal pathways. Compared with normal tissues, cancer cells show distinguished metabolic characteristics called metabolic reprogramming, which has been considered as a sign of cancer occurrence. With the deepening of tumor research in recent years, people gradually found that amino acid metabolism played crucial roles in cancer progression. Long non-coding RNAs (lncRNAs), which are implicated in many important biological processes, were firstly discovered dysregulating in cancer tissues and participating in extensive regulation of tumorigenesis. This review focuses on the reprogramming of amino acid metabolism in cancers and how lncRNAs participate in the regulatory network by interacting with other macromolecular substances. Understanding the functions of lncRNA in amino acid reprogramming in tumors might provide a new vision on the mechanisms of tumorigenesis and the development of new approaches for cancer therapy.
Collapse
Affiliation(s)
- Yuhong Guo
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Nanchang Key Laboratory of Orthopaedics, The Third Affiliated Hospital of Nanchang University, Nanchang, China.,Medical Department of Graduate School, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bin Lv
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Nanchang Key Laboratory of Orthopaedics, The Third Affiliated Hospital of Nanchang University, Nanchang, China.,Medical Department of Graduate School, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Renfeng Liu
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Nanchang Key Laboratory of Orthopaedics, The Third Affiliated Hospital of Nanchang University, Nanchang, China.,Medical Department of Graduate School, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zhengzai Dai
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Nanchang Key Laboratory of Orthopaedics, The Third Affiliated Hospital of Nanchang University, Nanchang, China.,Medical Department of Graduate School, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Feifei Zhang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Yiping Liang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Bo Yu
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Nanchang Key Laboratory of Orthopaedics, The Third Affiliated Hospital of Nanchang University, Nanchang, China.,Medical Department of Graduate School, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Duo Zeng
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Nanchang Key Laboratory of Orthopaedics, The Third Affiliated Hospital of Nanchang University, Nanchang, China.,Medical Department of Graduate School, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiao-Bin Lv
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.
| | - Zhiping Zhang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China. .,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, Northern 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China. .,Nanchang Key Laboratory of Orthopaedics, The Third Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
26
|
Cerrato A, Bedia C, Capriotti AL, Cavaliere C, Gentile V, Maggi M, Montone CM, Piovesana S, Sciarra A, Tauler R, Laganà A. Untargeted metabolomics of prostate cancer zwitterionic and positively charged compounds in urine. Anal Chim Acta 2021; 1158:338381. [PMID: 33863412 DOI: 10.1016/j.aca.2021.338381] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 02/07/2023]
Abstract
Prostate cancer, a leading cause of cancer-related deaths worldwide, principally occurs in over 50-year-old men. Nowadays there is urgency to discover biomarkers alternative to prostate-specific antigen, as it cannot discriminate patients with benign prostatic hyperplasia from clinically significant forms of prostatic cancer. In the present paper, 32 benign prostatic hyperplasia and 41 prostatic cancer urine samples were collected and analyzed. Polar and positively charged metabolites were therein investigated using an analytical platform comprising an up to 40-fold analyte enrichment step by graphitized carbon black solid-phase extraction, HILIC separation, and untargeted high-resolution mass spectrometry analysis. These classes of compounds are often neglected in common metabolomics experiments even though previous studies reported their significance in cancer biomarker discovery. The complex metabolomics big datasets, generated by the UHPLC-HRMS, were analyzed with the ROIMCR procedure, based on the selection of the MS regions of interest data and their analysis by the Multivariate Curve-Resolution Alternating Least Squares chemometrics method. This approach allowed the resolution and tentative identification of the metabolites differentially expressed by the two data sets. Among these, amino acids and carnitine derivatives were tentatively identified highlighting the importance of the proposed methodology for cancer biomarker research.
Collapse
Affiliation(s)
- Andrea Cerrato
- Dipartimento di Chimica, Sapienza Università di Roma, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Carmen Bedia
- Institute of Environmental Assessment and Water Research (IDAEA-CSIC), Barcelona, Spain
| | - Anna Laura Capriotti
- Dipartimento di Chimica, Sapienza Università di Roma, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Chiara Cavaliere
- Dipartimento di Chimica, Sapienza Università di Roma, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Vincenzo Gentile
- Dipartimento di Scienze Ginecologio-ostetriche e Scienze Urologiche, Sapienza Università, di Roma, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Martina Maggi
- Dipartimento di Scienze Ginecologio-ostetriche e Scienze Urologiche, Sapienza Università, di Roma, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Carmela Maria Montone
- Dipartimento di Chimica, Sapienza Università di Roma, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Susy Piovesana
- Dipartimento di Chimica, Sapienza Università di Roma, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Alessandro Sciarra
- Dipartimento di Scienze Ginecologio-ostetriche e Scienze Urologiche, Sapienza Università, di Roma, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Roma Tauler
- Institute of Environmental Assessment and Water Research (IDAEA-CSIC), Barcelona, Spain
| | - Aldo Laganà
- Dipartimento di Chimica, Sapienza Università di Roma, Piazzale Aldo Moro 5, 00185, Rome, Italy; CNR NANOTEC, Campus Ecotekne, University of Salento, Via Monteroni, 73100, Lecce, Italy
| |
Collapse
|
27
|
Metabolomic characterisation of progression and spontaneous regression of melanoma in the melanoma-bearing Libechov minipig model. Melanoma Res 2021; 31:140-151. [PMID: 33625100 DOI: 10.1097/cmr.0000000000000722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Melanoma-bearing Libechov minipig (MeLiM) represents a large animal model for melanoma research. This model shows a high incidence of complete spontaneous regression of melanoma - a phenomenon uncommon in humans. Here, we present the first metabolomic characterisation of the MeLiM model comparing animals with progressing and spontaneously regressing melanomas. Plasma samples of 19 minipigs with progression and 27 minipigs with evidence of regression were analysed by a targeted metabolomic assay based on mass spectrometry detection. Differences in plasma metabolomics patterns were investigated by univariate and multivariate statistical analyses. Overall, 185 metabolites were quantified in each plasma sample. Significantly altered metabolomic profile was found, and 42 features were differentially regulated in plasma. Besides, the machine learning approach was used to create a predictive model utilising Arg/Orn and Arg/ADMA ratios to discriminate minipigs with progressive disease development from minipigs with regression evidence. Our results suggest that progression of melanoma in the MeLiM model is associated with alteration of arginine, glycerophospholipid and acylcarnitines metabolism. Moreover, this study provides targeted metabolomics characterisation of an animal model of melanoma with progression and spontaneous regression of tumours.
Collapse
|
28
|
El-Mais N, Fakhoury I, Abdellatef S, Abi-Habib R, El-Sibai M. Human recombinant arginase I [HuArgI (Co)-PEG5000]-induced arginine depletion inhibits ovarian cancer cell adhesion and migration through autophagy-mediated inhibition of RhoA. J Ovarian Res 2021; 14:13. [PMID: 33423701 PMCID: PMC7798344 DOI: 10.1186/s13048-021-00767-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 01/04/2021] [Indexed: 11/10/2022] Open
Abstract
Ovarian carcinoma is the second most common malignancy of the female reproductive system and the leading cause of death from female reproductive system malignancies. Cancer cells have increased proliferation rate and thus require high amounts of amino acids, including arginine. L-arginine is a non-essential amino acid synthesized from L-citrulline by the Arginosuccinate synthetase (ASS1) enzyme. We have previously shown that the ovarian cancer cells, SKOV3, are auxotrophic to arginine, and that arginine deprivation by treatment with the genetically engineered human arginase I (HuArgI (Co)-PEG5000) triggers the death of SKOV3 cells by autophagy. In this study we examine the effect of HuArgI (Co)-PEG5000 on ovarian cancer cell migration and we dissect the mechanism involved. Wound healing assays, 2D random cell migration assays and cell adhesion analysis indicate that arginine deprivation decreases SKOV3 cell migration and adhesion. This effect was mimicked when autophagy was induced through rapamycin and reversed with the autophagy inhibitor chloroquine when autophagy was inhibited. This proved that arginine deprivation leads to the inhibition of cancer cell migration through autophagy, in addition to cell death. In addition, we were able to establish through pull-down assays and reversal experiments, that arginine deprivation-mediated autophagy inhibits cell migration through a direct inhibition of RhoA, member of the Rho family of GTPases. In conclusion, here we identify, for the first time, an autophagy-mediated inhibition of RhoA that plays an important role in regulating ovarian cancer cells motility and adhesion in response to arginine depletion.
Collapse
Affiliation(s)
- Nour El-Mais
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, Beirut, 1102 2801, Lebanon
| | - Isabelle Fakhoury
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, Beirut, 1102 2801, Lebanon
| | - Sandra Abdellatef
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, Beirut, 1102 2801, Lebanon
| | - Ralph Abi-Habib
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, Beirut, 1102 2801, Lebanon
| | - Mirvat El-Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, Beirut, 1102 2801, Lebanon.
| |
Collapse
|
29
|
Targeting Metabolic Pathways in Kidney Cancer: Rationale and Therapeutic Opportunities. ACTA ACUST UNITED AC 2020; 26:407-418. [PMID: 32947309 DOI: 10.1097/ppo.0000000000000472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alterations in cellular sugar, amino acid and nucleic acid, and lipid metabolism, as well as in mitochondrial function, are a hallmark of renal cell carcinoma (RCC). The activation of oncogenes such as hypoxia-inducible factor and loss of the von Hippel-Lindau function and other tumor suppressors frequently occur early on during tumorigenesis and are the drivers for these changes, collectively known as "metabolic reprogramming," which promotes cellular growth, proliferation, and stress resilience. However, tumor cells can become addicted to reprogrammed metabolism. Here, we review the current knowledge of metabolic addictions in clear cell RCC, the most common form of RCC, and to what extent this has created therapeutic opportunities to interfere with such altered metabolic pathways to selectively target tumor cells. We highlight preclinical and emerging clinical data on novel therapeutics targeting metabolic traits in clear cell RCC to provide a comprehensive overview on current strategies to exploit metabolic reprogramming clinically.
Collapse
|
30
|
Zhang L, Wang P, Shi M, Fang Z, Ji J, Liao X, Hu X, Chen F. The modulation of Luffa cylindrica (L.) Roem supplementation on gene expression and amino acid profiles in liver for alleviating hepatic steatosis via gut microbiota in high-fat diet-fed mice: insight from hepatic transcriptome analysis. J Nutr Biochem 2020; 80:108365. [DOI: 10.1016/j.jnutbio.2020.108365] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/03/2020] [Accepted: 02/21/2020] [Indexed: 12/13/2022]
|
31
|
Al-Koussa H, El Mais N, Maalouf H, Abi-Habib R, El-Sibai M. Arginine deprivation: a potential therapeutic for cancer cell metastasis? A review. Cancer Cell Int 2020; 20:150. [PMID: 32390765 PMCID: PMC7201942 DOI: 10.1186/s12935-020-01232-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/23/2020] [Indexed: 01/04/2023] Open
Abstract
Arginine is a semi essential amino acid that is used in protein biosynthesis. It can be obtained from daily food intake or synthesized in the body through the urea cycle using l-citrulline as a substrate. Arginine has a versatile role in the body because it helps in cell division, wound healing, ammonia disposal, immune system, and hormone biosynthesis. It is noteworthy that l-arginine is the precursor for the biosynthesis of nitric oxide (NO) and polyamines. In the case of cancer cells, arginine de novo synthesis is not enough to compensate for their high nutritional needs, forcing them to rely on extracellular supply of arginine. In this review, we will go through the importance of arginine deprivation as a novel targeting therapy by discussing the different arginine deprivation agents and their mechanism of action. We will also focus on the factors that affect cell migration and on the influence of arginine on metastases through polyamine and NO.
Collapse
Affiliation(s)
- Houssam Al-Koussa
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, 1102 2801 Beirut, Lebanon
| | - Nour El Mais
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, 1102 2801 Beirut, Lebanon
| | - Hiba Maalouf
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, 1102 2801 Beirut, Lebanon
| | - Ralph Abi-Habib
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, 1102 2801 Beirut, Lebanon
| | - Mirvat El-Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, P.O. Box: 13-5053, Chouran, 1102 2801 Beirut, Lebanon
| |
Collapse
|
32
|
Yang XH, Jing Y, Wang S, Ding F, Zhang XX, Chen S, Zhang L, Hu QG, Ni YH. Integrated Non-targeted and Targeted Metabolomics Uncovers Amino Acid Markers of Oral Squamous Cell Carcinoma. Front Oncol 2020; 10:426. [PMID: 32351881 PMCID: PMC7174902 DOI: 10.3389/fonc.2020.00426] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/10/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose: It is very important to develop potential molecular associated with oral squamous cell carcinoma (OSCC) malignant transformation and progression. Thus, the aim of our study was to determine the amino acid metabolic characteristics of OSCC patients and test their diagnostic value. Experimental Design: Eight pairs of matched tumor and normal samples were collected for gas chromatography–mass spectrometry (GC-MS) high-throughput untargeted analysis. Another 20 cases (each case including tumor and normal tissues) were also enrolled for ultrahigh-performance liquid chromatography–tandem mass spectrometer (UHPLC-MS/MS) amino acid quantitative analysis. T-test and receiver operating characteristic (ROC) curve analysis were used to determine candidate markers. Principal component analysis, partial least squares discriminant analysis, and heat map analysis were used to verify the ability of candidate markers to distinguish tumors from normal tissues. Results: A total of 10 amino acids biomarker were selected as OSCC candidate diagnostic biomarkers by GC-MS high-throughput untargeted metabolomics analyses [area under the curve (AUC) >0.80]. We further measured the specific concentration of these candidate amino acids biomarkers in another batch of 20 cases by UHPLC-MS/MS quantitative analysis. The result validated that nine amino acids had been detected, which had statistically significant difference (t-test, p < 0.05). Moreover, three of nine amino acid markers (glutamate, aspartic acid, and proline) displayed high sensitivity and specificity (AUC >0.90) by ROC curve analysis and obtained optimal sensitivity and specificity by binary logistic regression in the Glmnet package (AUC = 0.942). Conclusions: In conclusion, a panel including three amino acids (glutamate, aspartic acid, and proline) was identified as potential diagnostic biomarkers of OSCC by a combination of non-targeted and targeted metabolomics methods.
Collapse
Affiliation(s)
- Xi-Hu Yang
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yue Jing
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Shuai Wang
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Feng Ding
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiao-Xin Zhang
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Sheng Chen
- Department of Oral Pathology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lei Zhang
- Department of Oral Pathology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qin-Gang Hu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yan-Hong Ni
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
33
|
Fakhri S, Moradi SZ, Farzaei MH, Bishayee A. Modulation of dysregulated cancer metabolism by plant secondary metabolites: A mechanistic review. Semin Cancer Biol 2020; 80:276-305. [PMID: 32081639 DOI: 10.1016/j.semcancer.2020.02.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/08/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022]
Abstract
Several signaling pathways and basic metabolites are responsible for the control of metabolism in both normal and cancer cells. As emerging hallmarks of cancer metabolism, the abnormal activities of these pathways are of the most noticeable events in cancer. This altered metabolism expedites the survival and proliferation of cancer cells, which have attracted a substantial amount of interest in cancer metabolism. Nowadays, targeting metabolism and cross-linked signaling pathways in cancer has been a hot topic to investigate novel drugs against cancer. Despite the efficiency of conventional drugs in cancer therapy, their associated toxicity, resistance, and high-cost cause limitations in their application. Besides, considering the numerous signaling pathways cross-linked with cancer metabolism, discovery, and development of multi-targeted and safe natural compounds has been a high priority. Natural secondary metabolites have exhibited promising anticancer effects by targeting dysregulated signaling pathways linked to cancer metabolism. The present review reveals the metabolism and cross-linked dysregulated signaling pathways in cancer. The promising therapeutic targets in cancer, as well as the critical role of natural secondary metabolites for significant anticancer enhancements, have also been highlighted to find novel/potential therapeutic agents for cancer treatment.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran.
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| |
Collapse
|
34
|
Abstract
Macroautophagy (referred to here as autophagy) degrades and recycles cytoplasmic constituents to sustain cellular and mammalian metabolism and survival during starvation. Deregulation of autophagy is involved in numerous diseases, such as cancer. Cancers up-regulate autophagy and depend on it for survival, growth, and malignancy in a tumor cell-autonomous fashion. Recently, it has become apparent that autophagy in host tissues as well as the tumor cells themselves contribute to tumor growth. Understanding how autophagy regulates metabolism and tumor growth has revealed new essential tumor nutrients, where they come from, and how they are supplied and used, which can now be targeted for cancer therapy.
Collapse
Affiliation(s)
- Laura Poillet-Perez
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
35
|
Liu Z, Tu MJ, Zhang C, Jilek JL, Zhang QY, Yu AM. A reliable LC-MS/MS method for the quantification of natural amino acids in mouse plasma: Method validation and application to a study on amino acid dynamics during hepatocellular carcinoma progression. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1124:72-81. [PMID: 31177050 DOI: 10.1016/j.jchromb.2019.05.039] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/17/2019] [Accepted: 05/31/2019] [Indexed: 01/15/2023]
Abstract
A simple and fast LC-MS/MS method was developed and validated for simultaneous quantification of 20 proteinogenic l-amino acids (AAs) in a small volume (5 μL) of mouse plasma. Chromatographic separation was achieved on an Intrada Amino Acid column within 13 min via gradient elution with an aqueous solution containing 100 mM ammonium formate and an organic mobile phase containing acetonitrile, water and formic acid (v:v:v = 95:5:0.3), at the flow rate of 0.6 mL/min. Individual AAs and corresponding stable-isotope-labeled AAs internal standards were analyzed by multiple reaction monitoring (MRM) in positive ion mode under optimized conditions. Method validation consisted of linearity, sensitivity, accuracy and precision, recovery, matrix effect, and stability, and the results demonstrated this LC-MS/MS method as a specific, accurate, and reliable assay. This LC-MS/MS method was thus utilized to compare the dynamics of individual plasma AAs between healthy and orthotopic hepatocellular carcinoma (HCC) xenograft mice housed under identical conditions. Our results revealed that, 5 weeks after HCC tumor progression, plasma l-arginine concentrations were significantly decreased in HCC mice while l-alanine and l-threonine levels were sharply increased. These findings support the utilities of this LC-MS/MS method and the promise of specific AAs as possible biomarkers for HCC.
Collapse
Affiliation(s)
- Zhenzhen Liu
- Department of Medical Function, Health Science Center, Yangtze University, Jingzhou, Hubei 434000, China; Department of Biochemistry & Molecular Medicine, School of Medicine, UC Davis, Sacramento, CA 95817, USA
| | - Mei-Juan Tu
- Department of Biochemistry & Molecular Medicine, School of Medicine, UC Davis, Sacramento, CA 95817, USA
| | - Chao Zhang
- Department of Biochemistry & Molecular Medicine, School of Medicine, UC Davis, Sacramento, CA 95817, USA
| | - Joseph L Jilek
- Department of Biochemistry & Molecular Medicine, School of Medicine, UC Davis, Sacramento, CA 95817, USA
| | - Qian-Yu Zhang
- Department of Biochemistry & Molecular Medicine, School of Medicine, UC Davis, Sacramento, CA 95817, USA
| | - Ai-Ming Yu
- Department of Biochemistry & Molecular Medicine, School of Medicine, UC Davis, Sacramento, CA 95817, USA.
| |
Collapse
|
36
|
Ocaña MC, Martínez-Poveda B, Quesada AR, Medina MÁ. Metabolism within the tumor microenvironment and its implication on cancer progression: An ongoing therapeutic target. Med Res Rev 2019; 39:70-113. [PMID: 29785785 DOI: 10.1002/med.21511] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/30/2018] [Accepted: 05/01/2018] [Indexed: 01/03/2025]
Abstract
Since reprogramming energy metabolism is considered a new hallmark of cancer, tumor metabolism is again in the spotlight of cancer research. Many studies have been carried out and many possible therapies have been developed in the last years. However, tumor cells are not alone. A series of extracellular components and stromal cells, such as endothelial cells, cancer-associated fibroblasts, tumor-associated macrophages, and tumor-infiltrating T cells, surround tumor cells in the so-called tumor microenvironment (TME). Metabolic features of these cells are being studied in deep in order to find relationships between metabolism within the TME and tumor progression. Moreover, it cannot be forgotten that tumor growth is able to modulate host metabolism and homeostasis, so that TME is not the whole story. Importantly, the metabolic switch in cancer is just a consequence of the flexibility and adaptability of metabolism and should not be surprising. Treatments of cancer patients with combined therapies including antitumor agents with those targeting stromal cell metabolism, antiangiogenic drugs, and/or immunotherapy are being developed as promising therapeutics.
Collapse
Affiliation(s)
- Ma Carmen Ocaña
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, and IBIMA (Biomedical Research Institute of Málaga), Andalucía Tech, Universidad de Málaga, Málaga, Spain
| | - Beatriz Martínez-Poveda
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, and IBIMA (Biomedical Research Institute of Málaga), Andalucía Tech, Universidad de Málaga, Málaga, Spain
| | - Ana R Quesada
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, and IBIMA (Biomedical Research Institute of Málaga), Andalucía Tech, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), Málaga, Spain
| | - Miguel Ángel Medina
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, and IBIMA (Biomedical Research Institute of Málaga), Andalucía Tech, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), Málaga, Spain
| |
Collapse
|
37
|
Brin E, Wu K, Dagostino E, Meng-Chiang Kuo M, He Y, Shia WJ, Chen LC, Stempniak M, Hickey R, Almassy R, Showalter R, Thomson J. TRAIL stabilization and cancer cell sensitization to its pro-apoptotic activity achieved through genetic fusion with arginine deiminase. Oncotarget 2018; 9:36914-36928. [PMID: 30651925 PMCID: PMC6319333 DOI: 10.18632/oncotarget.26398] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 11/16/2018] [Indexed: 02/06/2023] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) binds to death receptors and induces apoptosis in various cancer cell lines while sparing normal cells. Recombinant TRAIL has shown good safety and efficacy profiles in preclinical cancer models. However, clinical success has been limited due to poor PK and development of resistance to death receptor-induced apoptosis. We have addressed these issues by creating a fusion protein of TRAIL and arginine deiminase (ADI). The fusion protein benefits from structural and functional synergies between its two components and has an extended half-life in vivo. ADI downregulates survivin, upregulates DR5 receptor and sensitizes cancer cells to TRAIL induced apoptosis. ADI-TRAIL fusion protein was efficacious in a number of cell lines and synergized with some standard of care drugs. In an HCT116 xenograft model ADI-TRAIL localized to the tumor and induced dose-dependent tumor regression, the fusion protein was superior to rhTRAIL administered at the same molar amounts.
Collapse
Affiliation(s)
- Elena Brin
- Polaris Pharmaceuticals, San Diego, CA, USA
| | | | | | | | - Yudou He
- Polaris Pharmaceuticals, San Diego, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Fouda AY, Xu Z, Shosha E, Lemtalsi T, Chen J, Toque HA, Tritz R, Cui X, Stansfield BK, Huo Y, Rodriguez PC, Smith SB, Caldwell RW, Narayanan SP, Caldwell RB. Arginase 1 promotes retinal neurovascular protection from ischemia through suppression of macrophage inflammatory responses. Cell Death Dis 2018; 9:1001. [PMID: 30254218 PMCID: PMC6156564 DOI: 10.1038/s41419-018-1051-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/24/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022]
Abstract
The lack of effective therapies to limit neurovascular injury in ischemic retinopathy is a major clinical problem. This study aimed to examine the role of ureohydrolase enzyme, arginase 1 (A1), in retinal ischemia-reperfusion (IR) injury. A1 competes with nitric oxide synthase (NOS) for their common substrate l-arginine. A1-mediated l-arginine depletion reduces nitric oxide (NO) formation by NOS leading to vascular dysfunction when endothelial NOS is involved but prevents inflammatory injury when inducible NOS is involved. Studies were performed using wild-type (WT) mice, global A1+/− knockout (KO), endothelial-specific A1 KO, and myeloid-specific A1 KO mice subjected to retinal IR injury. Global as well as myeloid-specific A1 KO mice showed worsened IR-induced neuronal loss and retinal thinning. Deletion of A1 in endothelial cells had no effect, while treatment with PEGylated (PEG) A1 improved neuronal survival in WT mice. In addition, A1+/− KO mice showed worsened vascular injury manifested by increased acellular capillaries. Western blotting analysis of retinal tissue showed increased inflammatory and necroptotic markers with A1 deletion. In vitro experiments showed that macrophages lacking A1 exhibit increased inflammatory response upon LPS stimulation. PEG-A1 treatment dampened this inflammatory response and decreased the LPS-induced metabolic reprogramming. Moreover, intravitreal injection of A1 KO macrophages or systemic macrophage depletion with clodronate liposomes increased neuronal loss after IR injury. These results demonstrate that A1 reduces IR injury-induced retinal neurovascular degeneration via dampening macrophage inflammatory responses. Increasing A1 offers a novel strategy for limiting neurovascular injury and promoting macrophage-mediated repair.
Collapse
Affiliation(s)
- Abdelrahman Y Fouda
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Zhimin Xu
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Esraa Shosha
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Tahira Lemtalsi
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Jijun Chen
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Haroldo A Toque
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Rebekah Tritz
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Xuezhi Cui
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Brian K Stansfield
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Department of Pediatrics, Augusta University, Augusta, GA, USA
| | - Yuqing Huo
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA
| | | | - Sylvia B Smith
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA.,Department of Ophthalmology, Augusta University, Augusta, GA, USA
| | - R William Caldwell
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - S Priya Narayanan
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, USA
| | - Ruth B Caldwell
- Charlie Norwood VA Medical Center, Augusta, GA, USA. .,Vascular Biology Center, Augusta University, Augusta, GA, USA. .,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA. .,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA. .,Department of Ophthalmology, Augusta University, Augusta, GA, USA.
| |
Collapse
|
39
|
Jahani M, Noroznezhad F, Mansouri K. Arginine: Challenges and opportunities of this two-faced molecule in cancer therapy. Biomed Pharmacother 2018; 102:594-601. [DOI: 10.1016/j.biopha.2018.02.109] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/21/2018] [Accepted: 02/23/2018] [Indexed: 12/15/2022] Open
|
40
|
Long Y, Tsai WB, Chang JT, Estecio M, Wangpaichitr M, Savaraj N, Feun LG, Chen HHW, Kuo MT. Cisplatin-induced synthetic lethality to arginine-starvation therapy by transcriptional suppression of ASS1 is regulated by DEC1, HIF-1α, and c-Myc transcription network and is independent of ASS1 promoter DNA methylation. Oncotarget 2018; 7:82658-82670. [PMID: 27765932 PMCID: PMC5347722 DOI: 10.18632/oncotarget.12308] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 09/19/2016] [Indexed: 12/31/2022] Open
Abstract
Many human tumors require extracellular arginine (Arg) for growth because the key enzyme for de novo biosynthesis of Arg, argininosuccinate synthetase 1 (ASS1), is silenced. These tumors are sensitive to Arg-starvation therapy using pegylated arginine deiminase (ADI-PEG20) which digests extracellular Arg. Many previous studies reported that ASS1 silencing is due to epigenetic inactivation of ASS1 expression by DNA methylation, and that the demethylation agent 5-aza-deoxycytidine (Aza-dC) can induce ASS1 expression. Moreover, it was reported that cisplatin suppresses ASS1 expression through ASS1 promoter methylation, leading to synthetic lethality to ADI-PEG20 treatment. We report here that cisplatin supppresses ASS1 expression is due to upregulation of HIF-1α and downregulation of c-Myc, which function as negative and positive regulators of ASS1 expression, respectively, by reciprocal bindings to the ASS1 promoter. In contrast, we found that Aza-dC induces ASS1 expression by downregulation of HIF-1α but upregulation of c-Myc. We further demonstrated that the clock protein DEC1 is the master regulator of HIF-1α and c-Myc that regulate ASS1. cDDP upregulates DEC1, whereas Aza-dC suppresses its expression. Using two proteasomal inhibitors bortezomib and carfilzomib which induce HIF-1α accumulation, we further demonstrated that HIF-1α is involved in ASS1 silencing for the maintenance of Arg auxotrophy for targeted Arg-starvation therapy.
Collapse
Affiliation(s)
- Yan Long
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wen-Bin Tsai
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Marcos Estecio
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Medhi Wangpaichitr
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Naramol Savaraj
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Lynn G Feun
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Helen H W Chen
- Department of Radiation Oncology, National Cheng Kung University, National Cheng Kung University Hospital, College of Medicine, Tainan, Taiwan
| | - Macus Tien Kuo
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
41
|
Mörén L, Perryman R, Crook T, Langer JK, Oneill K, Syed N, Antti H. Metabolomic profiling identifies distinct phenotypes for ASS1 positive and negative GBM. BMC Cancer 2018; 18:167. [PMID: 29422017 PMCID: PMC5806242 DOI: 10.1186/s12885-018-4040-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 01/23/2018] [Indexed: 11/10/2022] Open
Abstract
Background Tumour cells have a high demand for arginine. However, a subset of glioblastomas has a defect in the arginine biosynthetic pathway due to epigenetic silencing of the rate limiting enzyme argininosuccinate synthetase (ASS1). These tumours are auxotrophic for arginine and susceptible to the arginine degrading enzyme, pegylated arginine deiminase (ADI-PEG20). Moreover, ASS1 deficient GBM have a worse prognosis compared to ASS1 positive tumours. Since altered tumour metabolism is one of the hallmarks of cancer we were interested to determine if these two subtypes exhibited different metabolic profiles that could allow for their non-invasive detection as well as unveil additional novel therapeutic opportunities. Methods We looked for basal metabolic differences using one and two-dimensional gas chromatography-time-of-flight mass spectrometry (1D/2D GC-TOFMS) followed by targeted analysis of 29 amino acids using liquid chromatography-time-of-flight mass spectrometry (LC-TOFMS). We also looked for differences upon arginine deprivation in a single ASS1 negative and positive cell line (SNB19 and U87 respectively). The acquired data was evaluated by chemometric based bioinformatic methods. Results Orthogonal partial least squares-discriminant analysis (OPLS-DA) of both the 1D and 2D GC-TOFMS data revealed significant systematic difference in metabolites between the two subgroups with ASS1 positive cells generally exhibiting an overall elevation of identified metabolites, including those involved in the arginine biosynthetic pathway. Pathway and network analysis of the metabolite profile show that ASS1 negative cells have altered arginine and citrulline metabolism as well as altered amino acid metabolism. As expected, we observed significant metabolite perturbations in ASS negative cells in response to ADI-PEG20 treatment. Conclusions This study has highlighted significant differences in the metabolome of ASS1 negative and positive GBM which warrants further study to determine their diagnostic and therapeutic potential for the treatment of this devastating disease.
Collapse
Affiliation(s)
- Lina Mörén
- Department of Chemistry, Umeå University, SE 901 87, Umeå, Sweden
| | - Richard Perryman
- John Fulcher Neuro-Oncology Laboratory, Imperial College London, London, UK
| | - Tim Crook
- St Luke's Cancer Centre, Royal Surrey County Hospital, Guildford, Surrey, UK
| | - Julia K Langer
- John Fulcher Neuro-Oncology Laboratory, Imperial College London, London, UK
| | - Kevin Oneill
- John Fulcher Neuro-Oncology Laboratory, Imperial College London, London, UK
| | - Nelofer Syed
- John Fulcher Neuro-Oncology Laboratory, Imperial College London, London, UK.
| | - Henrik Antti
- Department of Chemistry, Umeå University, SE 901 87, Umeå, Sweden.
| |
Collapse
|
42
|
Chromatin remodeling system p300-HDAC2-Sin3A is involved in Arginine Starvation-Induced HIF-1α Degradation at the ASS1 promoter for ASS1 Derepression. Sci Rep 2017; 7:10814. [PMID: 28883660 PMCID: PMC5589935 DOI: 10.1038/s41598-017-11445-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/17/2017] [Indexed: 12/17/2022] Open
Abstract
Argininosuccinate synthetase 1 (ASS1) is the key enzyme that controls biosynthesis of arginine (Arg). ASS1 is silenced in many human malignancies therefore, these tumors require extracellular Arg for growth. The Arg-degrading recombinant protein, pegylated arginine deiminase (ADI-PEG20), has been in clinical trials for targeting Arg auxotrophic tumors by Arg starvation therapy. Resistance to Arg starvation is often developed through reactivation of ASS1 expression. We previously demonstrated that ASS1 silencing is controlled by HIF-1α and Arg starvation-reactivated ASS1 is associated with HIF-1α downregulation. However, mechanisms underlying ASS1 repression and HIF-1α turnover are not known. Here, we demonstrate that interplay of p300-HDAC2-Sin3A in the chromatin remodeling system is involved in HIF-1α degradation at the ASS1 promoter. The histone acetyltransferase p300 is normally associated with the ASS1 promoter to maintain acetylated H3K14ac and H3K27ac for ASS1 silencing. Arg starvation induces p300 dissociation, allowing histone HDAC2 and cofactor Sin3A to deacetylate these histones at the ASS1 promoter, thereby facilitating HIF-1α-proteasomal complex, driven by PHD2, to degrade HIF-1α in situ. Arg starvation induces PHD2 and HDAC2 interaction which is sensitive to antioxidants. This is the first report describing epigenetic regulation of chromosomal HIF-1α turnover in gene activation that bears important implication in cancer therapy.
Collapse
|
43
|
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]
|
44
|
PEGylated arginine deiminase can modulate tumor immune microenvironment by affecting immune checkpoint expression, decreasing regulatory T cell accumulation and inducing tumor T cell infiltration. Oncotarget 2017; 8:58948-58963. [PMID: 28938609 PMCID: PMC5601705 DOI: 10.18632/oncotarget.19564] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 07/16/2017] [Indexed: 12/31/2022] Open
Abstract
PEGylated arginine deiminase (ADI-PEG 20) is being investigated in clinical studies in arginine auxotrophic cancers and is well-tolerated. The anti-tumor properties of ADI-PEG 20 have been extensively investigated - ADI-PEG 20 inhibits the growth of auxotrophic cancers in vitro and in vivo - however, its impact on immune cells is largely unknown. Here we report the potential impact of ADI-PEG 20 on the tumor immune microenvironment. ADI-PEG 20 induced immunosuppressive programmed death-ligand 1 expression on some cancer cells in vitro, but the magnitude of the increase was cell line dependent and in most relatively small. Using healthy donor human peripheral blood mononuclear cells (PBMCs) we discovered that when present during initiation of T cell activation (but not later on) ADI-PEG 20 can inhibit their differentiation after early activation stage manifested by the expression of CD69 marker. In vivo, ADI-PEG 20 induced tumor T-cell infiltration in a poorly immunogenic syngeneic mouse melanoma B16-F10 model and reduced its growth as a single agent or when combined with anti-PD-1 mAb. It was also effective by itself or in combination with anti-PD-L1 mAb in CT26 colon carcinoma syngeneic model.
Collapse
|
45
|
Thongkum A, Wu C, Li YY, Wangpaichitr M, Navasumrit P, Parnlob V, Sricharunrat T, Bhudhisawasdi V, Ruchirawat M, Savaraj N. The Combination of Arginine Deprivation and 5-Fluorouracil Improves Therapeutic Efficacy in Argininosuccinate Synthetase Negative Hepatocellular Carcinoma. Int J Mol Sci 2017; 18:ijms18061175. [PMID: 28587170 PMCID: PMC5485998 DOI: 10.3390/ijms18061175] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/18/2017] [Accepted: 05/26/2017] [Indexed: 12/12/2022] Open
Abstract
Argininosuccinate synthetase (ASS), a key enzyme to synthesize arginine is down regulated in many tumors including hepatocellular carcinoma (HCC). Similar to previous reports, we have found the decrease in ASS expression in poorly differentiated HCC. These ASS(-) tumors are auxotrophic for arginine. Pegylated arginine deiminase (ADI-PEG20), which degrades arginine, has shown activity in these tumors, but the antitumor effect is not robust and hence combination treatment is needed. Herein, we have elucidated the effectiveness of ADI-PEG20 combined with 5-Fluorouracil (5-FU) in ASS(-)HCC by targeting urea cycle and pyrimidine metabolism using four HCC cell lines as model. SNU398 and SNU387 express very low levels of ASS or ASS(-) while Huh-1, and HepG2 express high ASS similar to normal cells. Our results showed that the augmented cytotoxic effect of combination treatment only occurs in SNU398 and SNU387, and not in HepG2 and Huh-1 (ASS(+)) cells, and is partly due to reduced anti-apoptotic proteins X-linked inhibitor of apoptosis protein (XIAP), myeloid leukemia cell differentiation protein (Mcl-1) and B-cell lymphoma-2 (Bcl-2). Importantly, lack of ASS also influences essential enzymes in pyrimidine synthesis (carbamoyl-phosphate synthetase2, aspartate transcarbamylase and dihydrooratase (CAD) and thymidylate synthase (TS)) and malate dehydrogenase-1 (MDH-1) in TCA cycle. ADI-PEG20 treatment decreased these enzymes and made them more vulnerable to 5-FU. Transfection of ASS restored these enzymes and abolished the sensitivity to ADI-PEG20 and combination treatment. Overall, our data suggest that ASS influences multiple enzymes involved in 5-FU sensitivity. Combining ADI-PEG20 and 5-FU may be effective to treat ASS(-)hepatoma and warrants further clinical investigation.
Collapse
Affiliation(s)
- Angkana Thongkum
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Laksi, Bangkok 10210, Thailand.
- Chulabhorn Graduate Institute, Laksi, Bangkok 10210, Thailand.
| | - Chunjing Wu
- Division of Hematology/Oncology, Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA.
| | - Ying-Ying Li
- Division of Hematology/Oncology, Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Medhi Wangpaichitr
- Division of Hematology/Oncology, Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33125, USA.
| | - Panida Navasumrit
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Laksi, Bangkok 10210, Thailand.
- Chulabhorn Graduate Institute, Laksi, Bangkok 10210, Thailand.
- Center of Excellence on Environmental Health, Toxicology (EHT), Ministry of Education, Bangkok 10300, Thailand.
| | - Varabhorn Parnlob
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Laksi, Bangkok 10210, Thailand.
| | - Thaniya Sricharunrat
- Laboratory Unit of Pathology, Chulabhorn Hospital, Laksi, Bangkok 10210, Thailand.
| | - Vajarabhongsa Bhudhisawasdi
- Department of Surgery, Faculty of Medicine, Khonkaen University, Khonkaen 40000, Thailand.
- Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Laksi, Bangkok 10210, Thailand.
| | - Mathuros Ruchirawat
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Laksi, Bangkok 10210, Thailand.
- Center of Excellence on Environmental Health, Toxicology (EHT), Ministry of Education, Bangkok 10300, Thailand.
| | - Niramol Savaraj
- Division of Hematology/Oncology, Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
46
|
Construction and in vitro evaluation of enzyme nanoreactors based on carboxymethyl chitosan for arginine deprivation in cancer therapy. Carbohydr Polym 2017; 162:35-41. [DOI: 10.1016/j.carbpol.2017.01.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 01/02/2017] [Accepted: 01/05/2017] [Indexed: 11/22/2022]
|
47
|
Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S, Ishige N. Proliferation and motility of hepatocellular, pancreatic and gastric cancer cells grown in a medium without glucose and arginine, but with galactose and ornithine. Oncol Lett 2017; 13:1276-1280. [PMID: 28454246 PMCID: PMC5403308 DOI: 10.3892/ol.2017.5568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 11/10/2016] [Indexed: 12/14/2022] Open
Abstract
Human primary hepatocytes are able to survive in a medium without glucose and arginine, but supplemented with galactose and ornithine (hepatocyte selection medium; HSM). To address the possibility of the application of HSM in cancer therapy, hepatocellular carcinoma cells, pancreatic cancer cells and gastric cancer cells were cultured in HSM. Cell proliferation was analyzed using an MTS assay. Morphological changes were analyzed using hematoxylin and eosin staining. Apoptosis was analyzed using a TUNEL assay and cell motility was assessed with a scratch assay. Cell proliferation was significantly suppressed in cell lines grown in HSM (P<0.01 in all the cell lines). Hematoxylin and eosin staining revealed pyknotic nuclei, suggesting that these cells had undergone apoptosis. The number of TUNEL-positive cells was significantly increased in HSM. In the scratch assay, the distance between the growing edge and the scratched edge was significantly lower (P<0.01 in all the cell lines) in cells cultured in HSM, compared with those grown in Dulbecco's modified Eagle's medium or RPMI-1640. Therefore, the proliferation and motility of hepatocellular carcinoma cells, pancreatic cancer cells and gastric cancer cells was suppressed, and these cells subsequently underwent apoptosis in a medium without glucose and arginine, but containing galactose and ornithine.
Collapse
Affiliation(s)
- Minoru Tomizawa
- Department of Gastroenterology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Fuminobu Shinozaki
- Department of Radiology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Yasufumi Motoyoshi
- Department of Neurology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Takao Sugiyama
- Department of Rheumatology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Shigenori Yamamoto
- Department of Pediatrics, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Naoki Ishige
- Department of Neurosurgery, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| |
Collapse
|
48
|
Manig F, Kuhne K, von Neubeck C, Schwarzenbolz U, Yu Z, Kessler BM, Pietzsch J, Kunz-Schughart LA. The why and how of amino acid analytics in cancer diagnostics and therapy. J Biotechnol 2017; 242:30-54. [DOI: 10.1016/j.jbiotec.2016.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/28/2016] [Accepted: 12/01/2016] [Indexed: 12/11/2022]
|
49
|
Morris SM. Arginine Metabolism Revisited. J Nutr 2016; 146:2579S-2586S. [PMID: 27934648 DOI: 10.3945/jn.115.226621] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/22/2016] [Accepted: 02/05/2016] [Indexed: 01/20/2023] Open
Abstract
Mammalian arginine metabolism is complex due to the expression of multiple enzymes that utilize arginine as substrate and to interactions or competition between specific enzymes involved in arginine metabolism. Moreover, cells may contain multiple intracellular arginine pools that are not equally accessible to all arginine metabolic enzymes, thus presenting additional challenges to more fully understanding arginine metabolism. At the whole-body level, arginine metabolism ultimately results in the production of a biochemically diverse range of products, including nitric oxide, urea, creatine, polyamines, proline, glutamate, agmatine, and homoarginine. Included in this group of compounds are the methylated arginines (e.g., asymmetric dimethylarginine), which are released upon degradation of proteins containing methylated arginine residues. Changes in arginine concentration also can regulate cellular metabolism and function via a variety of arginine sensors. Although much is known about arginine metabolism, elucidation of the physiologic or pathophysiologic roles for all of the pathways and their metabolites remains an active area of investigation, as exemplified by current findings highlighted in this review.
Collapse
Affiliation(s)
- Sidney M Morris
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
50
|
Long Y, Tsai WB, Wang D, Hawke DH, Savaraj N, Feun LG, Hung MC, Chen HHW, Kuo MT. Argininosuccinate synthetase 1 (ASS1) is a common metabolic marker of chemosensitivity for targeted arginine- and glutamine-starvation therapy. Cancer Lett 2016; 388:54-63. [PMID: 27913198 DOI: 10.1016/j.canlet.2016.11.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/22/2016] [Accepted: 11/23/2016] [Indexed: 11/26/2022]
Abstract
Argininosuccinate synthetase 1 (ASS1) is the rate-limiting enzyme that catalyzes the biosynthesis of arginine (Arg). Many malignant human tumors are auxotrophic for Arg because ASS1 is silenced. ASS1 has been established as a sensor of Arg auxotrophic response and a chemosensitivity marker for Arg starvation therapy. Here, we report that ASS1 is also a sensor for glutamine (Gln)-deprivation response, and that upregulation of ASS1 expression is associated with resistance to Gln-starvation treatments. Knockdown of ASS1 expression resulted in increased sensitivity to both Arg- and Gln-starvation, whereas increased ASS1 expression by ectopic transfection is associated with resistance to both Arg- and Gln-starvation. The addition of permeable fumarate, a metabolite that bridges the tricarboxylic acid and urea cycles, resulted in downregulation of ASS1 expression and increased sensitivity to both Arg- and Gln-deprivation treatments. Mechanistically, the Gln-deprivation response, like the arginine-auxotrophic response, downregulates HIF-1α resulting in de-silencing of ASS1. Our results demonstrate that ASS1 is a common biosensor for Arg and Gln deprivation response and a shared target for Arg- and Gln-starvation therapies which have been in several current clinical trials.
Collapse
Affiliation(s)
- Yan Long
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wen-Bin Tsai
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dajuan Wang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China
| | - David H Hawke
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Niramol Savaraj
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lynn G Feun
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Helen H W Chen
- National Cheng Kung University, National Cheng Kung University Hospital, College of Medicine, Department of Radiation Oncology, Tainan, Taiwan
| | - Macus Tien Kuo
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|