1
|
Fis1 phosphorylation by Met promotes mitochondrial fission and hepatocellular carcinoma metastasis. Signal Transduct Target Ther 2021; 6:401. [PMID: 34848680 PMCID: PMC8632923 DOI: 10.1038/s41392-021-00790-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 12/24/2022] Open
Abstract
Met tyrosine kinase, a receptor for a hepatocyte growth factor (HGF), plays a critical role in tumor growth, metastasis, and drug resistance. Mitochondria are highly dynamic and undergo fission and fusion to maintain a functional mitochondrial network. Dysregulated mitochondrial dynamics are responsible for the progression and metastasis of many cancers. Here, using structured illumination microscopy (SIM) and high spatial and temporal resolution live cell imaging, we identified mitochondrial trafficking of receptor tyrosine kinase Met. The contacts between activated Met kinase and mitochondria formed dramatically, and an intact HGF/Met axis was necessary for dysregulated mitochondrial fission and cancer cell movements. Mechanically, we found that Met directly phosphorylated outer mitochondrial membrane protein Fis1 at Tyr38 (Fis1 pY38). Fis1 pY38 promoted mitochondrial fission by recruiting the mitochondrial fission GTPase dynamin-related protein-1 (Drp1) to mitochondria. Fragmented mitochondria fueled actin filament remodeling and lamellipodia or invadopodia formation to facilitate cell metastasis in hepatocellular carcinoma (HCC) cells both in vitro and in vivo. These findings reveal a novel and noncanonical pathway of Met receptor tyrosine kinase in the regulation of mitochondrial activities, which may provide a therapeutic target for metastatic HCC.
Collapse
|
2
|
Pan Y, Wang X, He Y, Lin S, Zhu M, Li Y, Wang J, Wang J, Ma X, Xu J, Yang L, Yang G, Huang J, Lu Y, Sheng J. Tumor suppressor ATP4B serve as a promising biomarker for worsening of gastric atrophy and poor differentiation. Gastric Cancer 2021; 24:314-326. [PMID: 33111209 DOI: 10.1007/s10120-020-01128-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hydrogen/potassium ATPase β (ATP4B) is a proton pump acting an essential role in gastric acid secretion. This study aimed to investigate the diagnostic performance of ATP4B and its biological role in tumor progression in gastric cancer. METHODS The correlations between ATP4B expression level and clinicopathologic parameters, as well as the relevance of ATP4B expression with overall survival were assessed. The functional roles of ATP4B in gastric cancer were verified by gain- and loss-of-function cell models and tumor xenograft models. The possible downstream effects of ATP4B were analyzed by iTRAQ-based quantitative proteomics analysis. RESULTS A dramatic decrease in ATP4B was associated with malignant transformation in gastric mucosa lesions and correlated with poor differentiation. Restoration of ATP4B expression in gastric cancer cells significantly suppressed cell proliferation, cell viability, migration, invasion, tumorigenicity and induced apoptosis, whereas ATP4B silencing exerted the opposite effects. Mechanistically, we found a quality control on mitochondrial metabolism and functions in ATP4B-overexpression GC cells. CONCLUSIONS Our data suggest that decreasing ATP4B is an indicator for gastric mucosa malignant transformation and GC aggressive phenotype and it plays an inhibitory role in gastric cancer as a tumor suppressor via regulating mitochondrial metabolism and apoptosis pathway.
Collapse
Affiliation(s)
- Yuanming Pan
- Department of Gastroenterology, the 7th Medical Center of Chinese PLA General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 100700, China.,Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital/Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Xin Wang
- Department of Gastroenterology, the 7th Medical Center of Chinese PLA General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 100700, China
| | - Yuqi He
- Department of Gastroenterology, the 7th Medical Center of Chinese PLA General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 100700, China.,The Second School of Clinical Medicine, Southern Medical University, 253 Middle Industrial Avenue, Guangzhou, 510282, Guangdong, China
| | - Shuye Lin
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital/Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China.,College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, 3 Shangyuan Residence, Haidian District, Beijing, 100044, China
| | - Min Zhu
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital/Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Yangjie Li
- Department of Gastroenterology, the 7th Medical Center of Chinese PLA General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 100700, China.,The Second School of Clinical Medicine, Southern Medical University, 253 Middle Industrial Avenue, Guangzhou, 510282, Guangdong, China
| | - Jianxun Wang
- School of Basic Medical Sciences, Qingdao University, Qingdao, China.,Center for Regenerative Medicine, Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Jiheng Wang
- Department of Gastroenterology, the 7th Medical Center of Chinese PLA General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 100700, China
| | - Xianzong Ma
- Department of Gastroenterology, the 7th Medical Center of Chinese PLA General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 100700, China
| | - Junfeng Xu
- Department of Gastroenterology, the 7th Medical Center of Chinese PLA General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 100700, China
| | - Lang Yang
- Department of Gastroenterology, the 7th Medical Center of Chinese PLA General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 100700, China
| | - Guibin Yang
- Department of Gastroenterology, Aerospace Clinic Medical College of Peking University, No. 15 Yuanquan Road, Haidian District, Beijing, 100049, China
| | - Jiaqiang Huang
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, 3 Shangyuan Residence, Haidian District, Beijing, 100044, China. .,Cancer and Inflammation Program (CIP), Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, MD, USA.
| | - Youyong Lu
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital/Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| | - Jianqiu Sheng
- Department of Gastroenterology, the 7th Medical Center of Chinese PLA General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 100700, China.
| |
Collapse
|
3
|
Wang J, Li J, Xiao Y, Fu B, Qin Z. TPP-based mitocans: a potent strategy for anticancer drug design. RSC Med Chem 2020; 11:858-875. [PMID: 33479681 PMCID: PMC7489259 DOI: 10.1039/c9md00572b] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 05/11/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the most important problems that endanger human health. The number of cancer patients is increasing rapidly worldwide. Compared with normal cells, cancer cells exhibit abnormal metabolism (abnormal glycolysis and oxidative phosphorylation, high levels of reactive oxygen species, anti-apoptosis, high mitochondrial membrane potential, and so on), and specific targeting of these metabolic abnormalities would be a promising drug design direction. These physiological characteristics are closely related to tumorigenesis and development, which are mainly regulated by mitochondria. Therefore, mitochondria have become important anticancer drug targets, attracting much attention in recent years. In this review, we systematically summarize various mitochondrial anticancer drugs developed, especially mitocans based on triphenylphosphonium (TPP), and discuss the advantages of TPP in endowing mitochondrial targeting function.
Collapse
Affiliation(s)
- Jiayao Wang
- College of science , China Agriculture University , Haidian District 100089 , China . ; Tel: +86 130 0199 1198
| | - Jiaqi Li
- College of science , China Agriculture University , Haidian District 100089 , China . ; Tel: +86 130 0199 1198
| | - Yumei Xiao
- College of science , China Agriculture University , Haidian District 100089 , China . ; Tel: +86 130 0199 1198
| | - Bin Fu
- College of science , China Agriculture University , Haidian District 100089 , China . ; Tel: +86 130 0199 1198
| | - Zhaohai Qin
- College of science , China Agriculture University , Haidian District 100089 , China . ; Tel: +86 130 0199 1198
| |
Collapse
|
4
|
Rodríguez-Hernández MA, de la Cruz-Ojeda P, López-Grueso MJ, Navarro-Villarán E, Requejo-Aguilar R, Castejón-Vega B, Negrete M, Gallego P, Vega-Ochoa Á, Victor VM, Cordero MD, Del Campo JA, Bárcena JA, Padilla CA, Muntané J. Integrated molecular signaling involving mitochondrial dysfunction and alteration of cell metabolism induced by tyrosine kinase inhibitors in cancer. Redox Biol 2020; 36:101510. [PMID: 32593127 PMCID: PMC7322178 DOI: 10.1016/j.redox.2020.101510] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/16/2020] [Indexed: 12/21/2022] Open
Abstract
Cancer cells have unlimited replicative potential, insensitivity to growth-inhibitory signals, evasion of apoptosis, cellular stress, and sustained angiogenesis, invasiveness and metastatic potential. Cancer cells adequately adapt cell metabolism and integrate several intracellular and redox signaling to promote cell survival in an inflammatory and hypoxic microenvironment in order to maintain/expand tumor phenotype. The administration of tyrosine kinase inhibitor (TKI) constitutes the recommended therapeutic strategy in different malignancies at advanced stages. There are important interrelationships between cell stress, redox status, mitochondrial function, metabolism and cellular signaling pathways leading to cell survival/death. The induction of apoptosis and cell cycle arrest widely related to the antitumoral properties of TKIs result from tightly controlled events involving different cellular compartments and signaling pathways. The aim of the present review is to update the most relevant studies dealing with the impact of TKI treatment on cell function. The induction of endoplasmic reticulum (ER) stress and Ca2+ disturbances, leading to alteration of mitochondrial function, redox status and phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt)-mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) signaling pathways that involve cell metabolism reprogramming in cancer cells will be covered. Emphasis will be given to studies that identify key components of the integrated molecular pattern including receptor tyrosine kinase (RTK) downstream signaling, cell death and mitochondria-related events that appear to be involved in the resistance of cancer cells to TKI treatments.
Collapse
Affiliation(s)
- María A Rodríguez-Hernández
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - P de la Cruz-Ojeda
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Mª José López-Grueso
- Department of Biochemistry and Molecular Biology, University of Cordoba, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Elena Navarro-Villarán
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Raquel Requejo-Aguilar
- Department of Biochemistry and Molecular Biology, University of Cordoba, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Beatriz Castejón-Vega
- Research Laboratory, Oral Medicine Department, University of Seville, Seville, Spain
| | - María Negrete
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Paloma Gallego
- Unit for the Clinical Management of Digestive Diseases, Hospital University "Nuestra Señora de Valme", Sevilla, Spain
| | - Álvaro Vega-Ochoa
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Victor M Victor
- Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Service of Endocrinology and Nutrition, Hospital University "Doctor Peset", Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; Department of Physiology, University of Valencia, Valencia, Spain
| | - Mario D Cordero
- Research Laboratory, Oral Medicine Department, University of Seville, Seville, Spain; Department of Physiology, Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center (CIBM), University of Granada, Armilla, Spain
| | - José A Del Campo
- Unit for the Clinical Management of Digestive Diseases, Hospital University "Nuestra Señora de Valme", Sevilla, Spain
| | - J Antonio Bárcena
- Department of Biochemistry and Molecular Biology, University of Cordoba, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - C Alicia Padilla
- Department of Biochemistry and Molecular Biology, University of Cordoba, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBiS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Department of General Surgery, Hospital University "Virgen del Rocío"/IBiS/CSIC/University of Seville, Seville, Spain.
| |
Collapse
|
5
|
EGFL9 promotes breast cancer metastasis by inducing cMET activation and metabolic reprogramming. Nat Commun 2019; 10:5033. [PMID: 31695034 PMCID: PMC6834558 DOI: 10.1038/s41467-019-13034-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/14/2019] [Indexed: 12/13/2022] Open
Abstract
The molecular mechanisms driving metastatic progression in triple-negative breast cancer (TNBC) patients are poorly understood. In this study, we demonstrate that epidermal growth factor-like 9 (EGFL9) is significantly upregulated in basal-like breast cancer cells and associated with metastatic progression in breast tumor samples. Functionally, EGFL9 is both necessary and sufficient to enhance cancer cell migration and invasion, as well as distant metastasis. Mechanistically, we demonstrate that EGFL9 binds cMET, activating cMET-mediated downstream signaling. EGFL9 and cMET co-localize at both the cell membrane and within the mitochondria. We further identify an interaction between EGFL9 and the cytochrome c oxidase (COX) assembly factor COA3. Consequently, EGFL9 regulates COX activity and modulates cell metabolism, promoting a Warburg-like metabolic phenotype. Finally, we show that combined pharmacological inhibition of cMET and glycolysis reverses EGFL9-driven stemness. Our results identify EGFL9 as a therapeutic target for combating metastatic progression in TNBC. Triple-negative breast cancer is an aggressive form of the disease. Here, the authors identify EGFL9 as a mediator of metastasis in TNBC through interactions with cMET.
Collapse
|
6
|
Poliaková M, Felser A, Pierzchala K, Nuoffer JM, Aebersold DM, Zimmer Y, Zamboni N, Medová M. Metabolomics reveals tepotinib-related mitochondrial dysfunction in MET-activating mutations-driven models. FEBS J 2019; 286:2692-2710. [PMID: 30993872 DOI: 10.1111/febs.14852] [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: 08/14/2018] [Revised: 02/27/2019] [Accepted: 04/12/2019] [Indexed: 11/26/2022]
Abstract
Genetic aberrations in the hepatocyte growth factor receptor tyrosine kinase MET induce oncogenic addiction in various types of human cancers, advocating MET as a viable anticancer target. Here, we report that MET signaling plays an important role in conferring a unique metabolic phenotype to cellular models expressing MET-activating mutated variants that are either sensitive or resistant toward MET small molecule inhibitors. MET phosphorylation downregulated by the specific MET inhibitor tepotinib resulted in markedly decreased viability and increased apoptosis in tepotinib-sensitive cells. Moreover, prior to the induction of MET inhibition-dependent cell death, tepotinib also led to an altered metabolic signature, characterized by a prominent reduction of metabolite ions related to amino sugar metabolism, gluconeogenesis, glycine and serine metabolism, and of numerous TCA cycle-related metabolites such as succinate, malate, and citrate. Functionally, a decrease in oxygen consumption rate, a reduced citrate synthase activity, a drop in membrane potential, and an associated misbalanced mitochondrial function were observed exclusively in MET inhibitor-sensitive cells. These data imply that interference with metabolic state can be considered an early indicator of efficient MET inhibition and particular changes reported here could be explored in the future as markers of efficacy of anti-MET therapies.
Collapse
Affiliation(s)
- Michaela Poliaková
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland.,Department for BioMedical Research, Radiation Oncology, University of Bern, Switzerland
| | - Andrea Felser
- University Institute of Clinical Chemistry, Bern University Hospital, Switzerland
| | - Katarzyna Pierzchala
- Center for Biomedical Imaging (CIBM), EPFL SB CIBM - AIT/LIFMET, Lausanne, Switzerland
| | - Jean-Marc Nuoffer
- University Institute of Clinical Chemistry, Bern University Hospital, Switzerland
| | - Daniel Matthias Aebersold
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland.,Department for BioMedical Research, Radiation Oncology, University of Bern, Switzerland
| | - Yitzhak Zimmer
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland.,Department for BioMedical Research, Radiation Oncology, University of Bern, Switzerland
| | - Nicola Zamboni
- Department of Biology, Institute of Molecular Systems Biology, Eidgenössische Technische Hochschule Zürich, Switzerland
| | - Michaela Medová
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland.,Department for BioMedical Research, Radiation Oncology, University of Bern, Switzerland
| |
Collapse
|
7
|
Battogtokh G, Choi YS, Kang DS, Park SJ, Shim MS, Huh KM, Cho YY, Lee JY, Lee HS, Kang HC. Mitochondria-targeting drug conjugates for cytotoxic, anti-oxidizing and sensing purposes: current strategies and future perspectives. Acta Pharm Sin B 2018; 8:862-880. [PMID: 30505656 PMCID: PMC6251809 DOI: 10.1016/j.apsb.2018.05.006] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/04/2018] [Accepted: 04/18/2018] [Indexed: 12/26/2022] Open
Abstract
Mitochondrial targeting is a promising approach for solving current issues in clinical application of chemotherapy and diagnosis of several disorders. Here, we discuss direct conjugation of mitochondrial-targeting moieties to anticancer drugs, antioxidants and sensor molecules. Among them, the most widely applied mitochondrial targeting moiety is triphenylphosphonium (TPP), which is a delocalized cationic lipid that readily accumulates and penetrates through the mitochondrial membrane due to the highly negative mitochondrial membrane potential. Other moieties, including short peptides, dequalinium, guanidine, rhodamine, and F16, are also known to be promising mitochondrial targeting agents. Direct conjugation of mitochondrial targeting moieties to anticancer drugs, antioxidants and sensors results in increased cytotoxicity, anti-oxidizing activity and sensing activity, respectively, compared with their non-targeting counterparts, especially in drug-resistant cells. Although many mitochondria-targeted anticancer drug conjugates have been investigated in vitro and in vivo, further clinical studies are still needed. On the other hand, several mitochondria-targeting antioxidants have been analyzed in clinical phases I, II and III trials, and one conjugate has been approved for treating eye disease in Russia. There are numerous ongoing studies of mitochondria-targeted sensors.
Collapse
Key Words
- (Fx, r)3, (l-cyclohexyl alanine-d-arginine)3
- 4-AT, 4-amino-TEMPO
- 5-FU, 5-Fluorouracil
- AD, Alzheimer׳s disease
- AIE, aggregation-induced emission
- ATP, adenosine triphosphate
- Anticancer agents
- Antioxidants
- Arg, arginine
- Aβ, beta amyloid
- BODIPY, boron-dipyrromethene
- C-dots, carbon dots
- CAT, catalase
- COX, cytochrome c oxidase
- CZBI, carbazole and benzo[e]indolium
- CoA, coenzyme A
- DDS, drug delivery system
- DEPMPO, 5-(diethylphosphono)-5-methyl-1-pyrroline N-oxide
- DIPPMPO, 5-(diisopropoxyphosphoryl)-5-methyl-1-pyrroline-N-oxide
- DQA, dequalinium
- Direct conjugation
- Dmt, dimethyltyrosine
- EPR, enhanced permeability and retention
- F16, (E)-4-(1H-indol-3-ylvinyl)-N-methylpyridinium iodide
- GPX, glutathione peroxidase
- GS, gramicidin S
- HTPP, 5-(4-hydroxy-phenyl)-10,15,20-triphenylporphyrin
- IMM, inner mitochondrial membrane
- IMS, intermembrane space
- IOA, imidazole-substituted oleic acid
- LA, lipoic acid
- LAH2, dihydrolipoic acid
- Lys, lysine
- MET, mesenchymal-epithelial transition
- MLS, mitochondria localization sequences
- MPO, myeloperoxidase
- MPP, mitochondria-penetrating peptides
- MitoChlor, TPP-chlorambucil
- MitoE, TPP-vitamin E
- MitoLA, TPP-lipoic acid
- MitoQ, TPP-ubiquinone
- MitoVES, TPP-vitamin E succinate
- Mitochondria-targeting
- Nit, nitrooxy
- NitDOX, nitrooxy-DOX
- OMM, outer mitochondrial membrane
- OXPHOS, oxidative phosphorylation
- PD, Parkinson׳s disease
- PDT, photodynamic therapy
- PET, photoinduced electron transfer
- PS, photosensitizer
- PTPC, permeability transition pore complex
- Phe, phenylalanine
- RNS, reactive nitrogen species
- ROS, reactive oxygen species
- SOD, superoxide dismutase
- SS peptide, Szeto-Schiller peptides
- Sensing agents
- SkQ1, Skulachev ion-quinone
- TEMPOL, 4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl
- TPEY-TEMPO, [2-(1-oxyl-2,2,6,6-tetramethylpiperidin-4-ylimino)-ethyl]-triphenyl-phosphonium
- TPP, triphenylphosphonium
- Tyr, tyrosine
- VDAC/ANT, voltage-dependent anion channel/adenine nucleotide translocase
- VES, vitamin E succinate
- XO, xanthine oxidase
- mitoTEMPO, (2-(2,2,6,6-tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium)
- mtCbl, (Fx,r)3-chlorambucil
- mtDNA, mitochondrial DNA
- mtPt, mitochondria-targeting (Fx,r)3-platinum(II)
- nDNA, nuclear DNA
- αTOS, alpha-tocopheryl succinate.
Collapse
Affiliation(s)
- Gantumur Battogtokh
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Gyeonggi-do 14662, Republic of Korea
| | - Yeon Su Choi
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Gyeonggi-do 14662, Republic of Korea
| | - Dong Seop Kang
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Gyeonggi-do 14662, Republic of Korea
| | - Sang Jun Park
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Gyeonggi-do 14662, Republic of Korea
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Kang Moo Huh
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Yong-Yeon Cho
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Gyeonggi-do 14662, Republic of Korea
| | - Joo Young Lee
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Gyeonggi-do 14662, Republic of Korea
| | - Hye Suk Lee
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Gyeonggi-do 14662, Republic of Korea
| | - Han Chang Kang
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Gyeonggi-do 14662, Republic of Korea
| |
Collapse
|
8
|
Cheng J, Wu LM, Deng XS, Wu J, Lv Z, Zhao HF, Yang Z, Ni Y. MicroRNA-449a suppresses hepatocellular carcinoma cell growth via G1 phase arrest and the HGF/MET c-Met pathway. Hepatobiliary Pancreat Dis Int 2018; 17:336-344. [PMID: 30108016 DOI: 10.1016/j.hbpd.2018.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 07/16/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Accumulating evidence demonstrates that microRNAs (miRNAs) play essential roles in tumorigenesis and cancer progression of hepatocellular carcinoma (HCC). Average targets of a miRNA were more than 100. And one miRNA may act in tumor via regulating several targets. The present study aimed to explore more potential targets of miR-449a by proteomics technology and further uncover the role of miR-449a in HCC tumorigenesis. METHODS Technologies such as iTRAQ-based quantitative proteomic were used to investigate the effect of miR-449a on HCC. The expression of c-Met and miR-449a was detected by qRT-PCR in HCC samples. Gain- and loss-of-function experiments were performed to identify the function and potential target of miR-449a in HCC cells. RESULTS In HCC, miR-449a was significantly downregulated, while c-Met was upregulated concurrently. Quantitative proteomics and luciferase reporter assay identified c-Met as a direct target of miR-449a. Moreover, miR-449a inhibited HCC growth not only by targeting CDK6 but also by suppressing c-Met/Ras/Raf/ERK signaling pathway. Furthermore, the inhibition of c-Met expression with a specific siRNA significantly inhibited cells growth and deregulated the ERK pathway in HCC. CONCLUSION The tumor suppressor miR-449a suppresses HCC tumorigenesis by repressing the c-Met/ERK pathway.
Collapse
Affiliation(s)
- Jun Cheng
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Li-Ming Wu
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou 310003, China
| | - Xue-Song Deng
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Jian Wu
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou 310003, China
| | - Zhen Lv
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou 310003, China
| | - Hang-Fen Zhao
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Zhang Yang
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; School of Life Science, Fudan University, Shanghai 200433, China
| | - Yong Ni
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China.
| |
Collapse
|
9
|
Lu M, Zhan X. The crucial role of multiomic approach in cancer research and clinically relevant outcomes. EPMA J 2018; 9:77-102. [PMID: 29515689 PMCID: PMC5833337 DOI: 10.1007/s13167-018-0128-8] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/29/2018] [Indexed: 02/06/2023]
Abstract
Cancer with heavily economic and social burden is the hot point in the field of medical research. Some remarkable achievements have been made; however, the exact mechanisms of tumor initiation and development remain unclear. Cancer is a complex, whole-body disease that involves multiple abnormalities in the levels of DNA, RNA, protein, metabolite and medical imaging. Biological omics including genomics, transcriptomics, proteomics, metabolomics and radiomics aims to systematically understand carcinogenesis in different biological levels, which is driving the shift of cancer research paradigm from single parameter model to multi-parameter systematical model. The rapid development of various omics technologies is driving one to conveniently get multi-omics data, which accelerates predictive, preventive and personalized medicine (PPPM) practice allowing prediction of response with substantially increased accuracy, stratification of particular patients and eventual personalization of medicine. This review article describes the methodology, advances, and clinically relevant outcomes of different "omics" technologies in cancer research, and especially emphasizes the importance and scientific merit of integrating multi-omics in cancer research and clinically relevant outcomes.
Collapse
Affiliation(s)
- Miaolong Lu
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| | - Xianquan Zhan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
- The State Key Laboratory of Medical Genetics, Central South University, 88 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| |
Collapse
|
10
|
Poliaková M, Aebersold DM, Zimmer Y, Medová M. The relevance of tyrosine kinase inhibitors for global metabolic pathways in cancer. Mol Cancer 2018; 17:27. [PMID: 29455660 PMCID: PMC5817809 DOI: 10.1186/s12943-018-0798-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/01/2018] [Indexed: 12/11/2022] Open
Abstract
Tumor metabolism is a thrilling discipline that focuses on mechanisms used by cancer cells to earn crucial building blocks and energy to preserve growth and overcome resistance to various treatment modalities. At the same time, therapies directed specifically against aberrant signalling pathways driven by protein tyrosine kinases (TKs) involved in proliferation, metastasis and growth count for several years to promising anti-cancer approaches. In this respect, small molecule inhibitors are the most widely used clinically relevant means for targeted therapy, with a rising number of approvals for TKs inhibitors. In this review, we discuss recent observations related to TKs-associated metabolism and to metabolic feedback that is initialized as cellular response to particular TK-targeted therapies. These observations provide collective evidence that therapeutic responses are primarily linked to such pathways as regulation of lipid and amino acid metabolism, TCA cycle and glycolysis, advocating therefore the development of further effective targeted therapies against a broader spectrum of TKs to treat patients whose tumors display deregulated signalling driven by these proteins.
Collapse
Affiliation(s)
- Michaela Poliaková
- Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Daniel M Aebersold
- Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Yitzhak Zimmer
- Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.,Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Michaela Medová
- Department of Radiation Oncology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland. .,Department for BioMedical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.
| |
Collapse
|
11
|
Way L, Faktor J, Dvorakova P, Nicholson J, Vojtesek B, Graham D, Ball KL, Hupp T. Rearrangement of mitochondrial pyruvate dehydrogenase subunit dihydrolipoamide dehydrogenase protein-protein interactions by the MDM2 ligand nutlin-3. Proteomics 2017; 16:2327-44. [PMID: 27273042 PMCID: PMC5026170 DOI: 10.1002/pmic.201500501] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 04/27/2016] [Accepted: 06/03/2016] [Indexed: 12/22/2022]
Abstract
Drugs targeting MDM2's hydrophobic pocket activate p53. However, these agents act allosterically and have agonist effects on MDM2's protein interaction landscape. Dominant p53‐independent MDM2‐drug responsive‐binding proteins have not been stratified. We used as a variable the differential expression of MDM2 protein as a function of cell density to identify Nutlin‐3 responsive MDM2‐binding proteins that are perturbed independent of cell density using SWATH‐MS. Dihydrolipoamide dehydrogenase, the E3 subunit of the mitochondrial pyruvate dehydrogenase complex, was one of two Nutlin‐3 perturbed proteins identified fours hour posttreatment at two cell densities. Immunoblotting confirmed that dihydrolipoamide dehydrogenase was induced by Nutlin‐3. Depletion of MDM2 using siRNA also elevated dihydrolipoamide dehydrogenase in Nutlin‐3 treated cells. Mitotracker confirmed that Nutlin‐3 inhibits mitochondrial activity. Enrichment of mitochondria using TOM22+ immunobeads and TMT labeling defined key changes in the mitochondrial proteome after Nutlin‐3 treatment. Proximity ligation identified rearrangements of cellular protein–protein complexes in situ. In response to Nutlin‐3, a reduction of dihydrolipoamide dehydrogenase/dihydrolipoamide acetyltransferase protein complexes highlighted a disruption of the pyruvate dehydrogenase complex. This coincides with an increase in MDM2/dihydrolipoamide dehydrogenase complexes in the nucleus that was further enhanced by the nuclear export inhibitor Leptomycin B. The data suggest one therapeutic impact of MDM2 drugs might be on the early perturbation of specific protein–protein interactions within the mitochondria. This methodology forms a blueprint for biomarker discovery that can identify rearrangements of MDM2 protein–protein complexes in drug‐treated cells.
Collapse
Affiliation(s)
- Luke Way
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Jakub Faktor
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Petra Dvorakova
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Judith Nicholson
- CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Borek Vojtesek
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Duncan Graham
- Centre for Molecular Nanometrology, WestCHEM, Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Kathryn L Ball
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK.,Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Ted Hupp
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK. .,Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
| |
Collapse
|
12
|
Yang T, Ng WH, Chen H, Chomchopbun K, Huynh TH, Go ML, Kon OL. Mitochondrial-Targeting MET Kinase Inhibitor Kills Erlotinib-Resistant Lung Cancer Cells. ACS Med Chem Lett 2016; 7:807-12. [PMID: 27563407 DOI: 10.1021/acsmedchemlett.6b00223] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 06/23/2016] [Indexed: 12/13/2022] Open
Abstract
Lung cancer cells harboring activating EGFR mutations acquire resistance to EGFR tyrosine kinase inhibitors (TKIs) by activating several bypass mechanisms, including MET amplification and overexpression. We show that a significant proportion of activated MET protein in EGFR TKI-resistant HCC827 lung cancer cells resides within the mitochondria. Targeting the total complement of MET in the plasma membrane and mitochondria should render these cells more susceptible to cell death and hence provide a means of circumventing drug resistance. Herein, the mitochondrial targeting triphenylphosphonium (TPP) moiety was introduced to the selective MET kinase inhibitor PHA665752. The resulting TPP analogue rapidly localized to the mitochondria of MET-overexpressing erlotinib-resistant HCC827 cells, partially suppressed the phosphorylation (Y1234/Y1235) of MET in the mitochondrial inner membrane and was as cytotoxic and apoptogenic as the parent compound. These findings provide support for the targeting of mitochondrial MET with a TPP-TKI conjugate as a means of restoring responsiveness to chemotherapy.
Collapse
Affiliation(s)
- Tianming Yang
- Department
of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543
| | | | - Huan Chen
- Department
of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543
| | - Kamon Chomchopbun
- Department
of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543
| | | | - Mei Lin Go
- Department
of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543
| | - Oi Lian Kon
- Department
of Biochemistry, National University of Singapore, 8 Medical
Drive, Singapore 117596
| |
Collapse
|
13
|
Farid SG, Morris-Stiff G. "OMICS" technologies and their role in foregut primary malignancies. Curr Probl Surg 2015; 52:409-41. [PMID: 26527526 DOI: 10.1067/j.cpsurg.2015.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 08/03/2015] [Indexed: 12/18/2022]
|
14
|
Zhang Y, Ng CK, Cohen Y, Cao B. Cell growth and protein expression of Shewanella oneidensis in biofilms and hydrogel-entrapped cultures. MOLECULAR BIOSYSTEMS 2014; 10:1035-42. [PMID: 24626808 DOI: 10.1039/c3mb70520j] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The performance of biofilm-based bioprocesses is difficult to predict and control because of the intrinsic heterogeneous and dynamic properties of microbial biofilms. Biofilm mimics, such as microbial cells entrapped in polymeric scaffolds that are permeable for nutrients, have been proposed to replace real biofilms to achieve long-term robust performance in engineering applications. However, the physiological differences between cells that are physically entrapped in a synthetic polymeric matrix and biofilm cells that are encased in a self-produced polymeric matrix remain unknown. In this study, using Shewanella oneidensis as a model organism and alginate hydrogel as a model synthetic matrix, we compared the cell growth and protein expression in entrapped cultures and biofilms. The hydrogel-entrapped cultures were found to exhibit a growth rate comparable with biofilms. There was no substantial difference in cell viability, surface charge, as well as hydrophobicity between the cells grown in alginate hydrogel and those grown in biofilms. However, the gel-entrapped cultures were found to be physiologically different from biofilms. The gel-entrapped cultures had a higher demand for metabolic energy. The siderophore-mediated iron uptake was repressed in the gel-entrapped cells. The presence of the hydrogel matrix decreased the expression of proteins involved in biofilm formation, while inducing the production of extracellular DNA (eDNA) in the gel-entrapped cultures. These results advance the fundamental understanding of the physiology of hydrogel-entrapped cells, which can lead to more efficient biofilm mimic-based applications.
Collapse
Affiliation(s)
- Yingdan Zhang
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore.
| | | | | | | |
Collapse
|
15
|
Kauffman EC, Ricketts CJ, Rais-Bahrami S, Yang Y, Merino MJ, Bottaro DP, Srinivasan R, Linehan WM. Molecular genetics and cellular features of TFE3 and TFEB fusion kidney cancers. Nat Rev Urol 2014; 11:465-75. [PMID: 25048860 DOI: 10.1038/nrurol.2014.162] [Citation(s) in RCA: 235] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Despite nearly two decades passing since the discovery of gene fusions involving TFE3 or TFEB in sporadic renal cell carcinoma (RCC), the molecular mechanisms underlying the renal-specific tumorigenesis of these genes remain largely unclear. The recently published findings of The Cancer Genome Atlas Network reported that five of the 416 surveyed clear cell RCC tumours (1.2%) harboured SFPQ-TFE3 fusions, providing further evidence for the importance of gene fusions. A total of five TFE3 gene fusions (PRCC-TFE3, ASPSCR1-TFE3, SFPQ-TFE3, NONO-TFE3, and CLTC-TFE3) and one TFEB gene fusion (MALAT1-TFEB) have been identified in RCC tumours and characterized at the mRNA transcript level. A multitude of molecular pathways well-described in carcinogenesis are regulated in part by TFE3 or TFEB proteins, including activation of TGFβ and ETS transcription factors, E-cadherin expression, CD40L-dependent lymphocyte activation, mTORC1 signalling, insulin-dependent metabolism regulation, folliculin signalling, and retinoblastoma-dependent cell cycle arrest. Determining which pathways are most important to RCC oncogenesis will be critical in discovering the most promising therapeutic targets for this disease.
Collapse
Affiliation(s)
- Eric C Kauffman
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - Christopher J Ricketts
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - Soroush Rais-Bahrami
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - Youfeng Yang
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - Maria J Merino
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - Donald P Bottaro
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - W Marston Linehan
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| |
Collapse
|
16
|
Adav SS, Chao LT, Sze SK. Protein abundance in multiplexed samples (PAMUS) for quantitation of Trichoderma reesei secretome. J Proteomics 2013; 83:180-96. [DOI: 10.1016/j.jprot.2013.03.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Revised: 03/20/2013] [Accepted: 03/23/2013] [Indexed: 11/27/2022]
|
17
|
Rodríguez-Suárez E, Whetton AD. The application of quantification techniques in proteomics for biomedical research. MASS SPECTROMETRY REVIEWS 2013; 32:1-26. [PMID: 22847841 DOI: 10.1002/mas.21347] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 02/09/2012] [Accepted: 02/10/2012] [Indexed: 06/01/2023]
Abstract
The systematic analysis of biological processes requires an understanding of the quantitative expression patterns of proteins, their interacting partners and their subcellular localization. This information was formerly difficult to accrue as the relative quantification of proteins relied on antibody-based methods and other approaches with low throughput. The advent of soft ionization techniques in mass spectrometry plus advances in separation technologies has aligned protein systems biology with messenger RNA, DNA, and microarray technologies to provide data on systems as opposed to singular protein entities. Another aspect of quantitative proteomics that increases its importance for the coming few years is the significant technical developments underway both for high pressure liquid chromatography and mass spectrum devices. Hence, robustness, reproducibility and mass accuracy are still improving with every new generation of instruments. Nonetheless, the methods employed require validation and comparison to design fit for purpose experiments in advanced protein analyses. This review considers the newly developed systematic protein investigation methods and their value from the standpoint that relative or absolute protein quantification is required de rigueur in biomedical research.
Collapse
|
18
|
Guo T, Fan L, Ng WH, Zhu Y, Ho M, Wan WK, Lim KH, Ong WS, Lee SS, Huang S, Kon OL, Sze SK. Multidimensional Identification of Tissue Biomarkers of Gastric Cancer. J Proteome Res 2012; 11:3405-13. [DOI: 10.1021/pr300212g] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Tiannan Guo
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive,
Singapore 637551
| | - Lingling Fan
- Center for Stem Cell Research & Application, Union Hospital, Huazhong University of Science and Technology, Wuhan, P.R. China 430022
| | | | - Yi Zhu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive,
Singapore 637551
| | | | - Wei Keat Wan
- Pathology Department, Singapore General Hospital, Outram Road, Singapore
169608
| | - Kiat Hon Lim
- Pathology Department, Singapore General Hospital, Outram Road, Singapore
169608
| | | | | | - Shiang Huang
- Center for Stem Cell Research & Application, Union Hospital, Huazhong University of Science and Technology, Wuhan, P.R. China 430022
| | | | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive,
Singapore 637551
| |
Collapse
|
19
|
Lin LL, Huang HC, Juan HF. Discovery of biomarkers for gastric cancer: a proteomics approach. J Proteomics 2012; 75:3081-97. [PMID: 22498886 DOI: 10.1016/j.jprot.2012.03.046] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 03/10/2012] [Accepted: 03/25/2012] [Indexed: 01/06/2023]
Abstract
Gastric cancer is the second leading cause of cancer-related deaths worldwide. Although many treatment options exist for patients with gastric tumors, the incidence and mortality rate of gastric cancer are on the rise. The early stages of gastric cancer are non-symptomatic, and the treatment response is unpredictable. This situation is further aggravated by a lack of diagnostic biomarkers that can aid in the early detection and prognosis of gastric cancer and in the prediction of chemoresistance. Moreover, clinical surgical specimens are rarely obtained, and traditional biomarkers of gastric cancer are not very effective. Many studies in the field of proteomics have contributed to the discovery and establishment of powerful diagnostic tools (e.g., ProteinChip array) in the management of cancer. The evolution in proteomic technologies has not only enabled the screening of a large number of samples but also enabled the identification of pathologically significant proteins, such as phosphoproteins, and the quantitation of difference in protein expression under different conditions. Multiplexed assays are used widely to accurately fractionate various complex samples such as blood, tissue, cells, and Helicobacter pylori-infected specimens to identify differentially expressed proteins. Biomarker detection studies have substantially contributed to the areas of secretome, metabolome, and phosphoproteome. Here, we review the development of potential biomarkers in the natural history of gastric cancer, with specific emphasis on the characteristics of target protein convergence.
Collapse
Affiliation(s)
- Li-Ling Lin
- Institute of Molecular and Cellular Biology and Department of Life Science, National Taiwan University, Taipei, Taiwan
| | | | | |
Collapse
|
20
|
Hao P, Qian J, Ren Y, Sze SK. Electrostatic repulsion-hydrophilic interaction chromatography (ERLIC) versus strong cation exchange (SCX) for fractionation of iTRAQ-labeled peptides. J Proteome Res 2011; 10:5568-74. [PMID: 22014306 DOI: 10.1021/pr2007686] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The iTRAQ technique is popular for the comparative analysis of proteins in different complex samples. To increase the dynamic range and sensitivity of peptide identification in shotgun proteomics, SCX chromatography is generally used for the fractionation of iTRAQ-labeled peptides before LC-MS/MS analysis. However, SCX suffers from clustering of similarly charged peptides and the need to desalt fractions. In this report, SCX is compared with the alternative ERLIC method for fractionating iTRAQ-labeled peptides. The simultaneous effect of electrostatic repulsion and hydrophilic interaction in ERLIC results in peptide elution in order of decreasing pI and GRAVY values (increasing polarity). Volatile solvents can be used. We applied ERLIC to iTRAQ-labeled peptides from rat liver tissue, and 2745 proteins and 30,016 unique peptides were identified with high confidence from three technical replicates. This was 12.9 and 49.4% higher, respectively, than was obtained using SCX. In addition, ERLIC is appreciably better at the identification of highly hydrophobic peptides. The results indicate that ERLIC is a more convenient and more effective alternative to SCX for the fractionation of iTRAQ-labeled peptides. Quantification data show that both SCX and ERLIC fractionation have no significant effect on protein quantification by iTRAQ.
Collapse
Affiliation(s)
- Piliang Hao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore
| | | | | | | |
Collapse
|