1
|
Wang S, Luo Z, Luo H, Li Z, Yuan Z, Tang J, Lin L, Du Z, Zhou JR. Effects of a calcium/vitamin D/Zinc combination on anti-osteoporosis in ovariectomized rats. J Trace Elem Med Biol 2023; 77:127138. [PMID: 36773556 DOI: 10.1016/j.jtemb.2023.127138] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/12/2023] [Accepted: 01/25/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Osteoporosis is a major health problem in postmenopausal women, and characterized by deteriorated bone mass and micro-architecture. There have been some clinical trials demonstrating the beneficial effects of vitamin-D and some trace elements on calcium absorption and attenuation of osteoporosis development. However, effects of the combination of vitamin-D and zinc on calcium absorption and osteoporosis have not been adequately investigated. METHODS Network pharmacology was first performed to explore possible correlations between calcium/vitamin D/zinc and osteoporosis. Forty-nine female Sprague-Dawley rats (6 months old, 250 ± 20 g) were randomized into 7 experimental groups with 7 animals per group for the in vivo study, including one sham surgery control group, one ovariectomizing (OVX) group, and 5 OVX plus treatment groups. At the end of animal experiment, animal tibia and femur leg bones and blood were collected for H&E staining, bone microstructure analysis by a micro-CT, measurement of bone and serum Ca, P and Zn concentrations, and immunohistochemical detection of macrophage-colony stimulating factor receptor (M-CSFR) and receptor activator of nuclear factor-kappa B ligand (RANKL). RESULTS The network pharmacology analysis identified 57 candidate targets that were related to the osteoporosis-Ca/VitD/Zn interconnections. Further pathway analysis suggested that the combined treatment of Ca, VitD and Zn attenuated osteoporosis via modulation of metabolic pathways. We found that a therapy with Ca/VitD-M/Zn-M (73 mg/kg/day Ca, 0.6 g/kg/day VitD3 and 0.6 mg/kg/day zinc citrate) could significantly suppress the progression of osteoporosis in rats. After the Ca/VitD-M/Zn-M treatment, the ratio of bone volume/tissue volume, trabecular number and the trabecular thickness were all significantly elevated while the extent of trabecular separation was significantly reduced. Additionally, both serum calcium and bone calcium levels were significantly upregulated by the Ca/VitD/Zn treatment in a dose-dependent manner. The combination of Ca/VitD-M/Zn-M was superiou to either Ca/VitD-L/Zn-L or Ca/VitD-H/Zn-H treatment for such an effect. Moreover, the osteoporosis-associated M-CSFR and RANKL factors were both significantly downregulated by the Ca/VitD-M/Zn-M treatment in bone tissues of OVX rats. CONCLUSIONS The combined supplement of VitD and Zn facilitates the Ca(2 +) absorption and attenuates the development of osteoporosis via down-regulation of osteoporosis-associated factors M-CSFR and RANKL, thus potentially constitutes an alternative therapy for the postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Shanshan Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006 Guangzhou, Guangdong, China
| | - Zhen Luo
- Infinitus (China) Co., Ltd. R & D Center, 510653 Guangzhou, Guangdong, China
| | - Hujie Luo
- Infinitus (China) Co., Ltd. R & D Center, 510653 Guangzhou, Guangdong, China
| | - Zhiwei Li
- Foshan Allan Conney Biotechnology Co., Ltd. 523281 Foshan, Guangdong, China
| | - Zhengqiang Yuan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006 Guangzhou, Guangdong, China
| | - Jian Tang
- Infinitus (China) Co., Ltd. R & D Center, 510653 Guangzhou, Guangdong, China
| | - Li Lin
- Foshan Allan Conney Biotechnology Co., Ltd. 523281 Foshan, Guangdong, China
| | - Zhiyun Du
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006 Guangzhou, Guangdong, China.
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
2
|
Chen J, Li P, Ye S, Li W, Li M, Ding Y. Systems pharmacology-based drug discovery and active mechanism of phlorotannins for type 2 diabetes mellitus by integrating network pharmacology and experimental evaluation. J Food Biochem 2022; 46:e14492. [PMID: 36385377 DOI: 10.1111/jfbc.14492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/29/2022] [Accepted: 10/17/2022] [Indexed: 11/18/2022]
Abstract
Phlorotannins, polyphenolic compounds that exist only in brown algae, have an effect on T2DM. However, the structure of phlorotannins is complex and diverse, and the complex role of therapeutic targets and active compounds has not been revealed. In this study, the potential targets and pharmacological effects of phlorotannins in the treatment of T2DM were identified based on network pharmacology and enzyme activity inhibition experiment. In total, 15 phlorotannins and 53 associated targets were yielded. Among them, SRC, ESR1, AKT1, HSP90AB1, and AR were defined as core targets. 527 GO biological processes items and 101 KEGG pathways were obtained, including EGFR tyrosine kinase inhibitor resistance, thyroid hormone signaling pathway, AGE-RAGE signaling pathway in diabetic complications, and VEGF signaling pathway. Phlorotannins could enable resistance against T2DM by inflammatory, survival, gene transcription, proliferation, apoptosis, and atherosclerosis. Finally, α-glucosidase inhibition assay and molecular docking proved the effect of selected phlorotannins on T2DM. PRACTICAL APPLICATIONS: Phlorotannins are a kind of polyphenol compounds that only exists in brown algae. Its structure is polymerized by aromatic precursors phloroglucinol (1,3,5-trihydroxybenzene). They have aroused great interest due to their excellent and valuable biological activities. However, the structure of phlorotannins is complex and diverse, and the complex role of therapeutic targets and active compounds has not been revealed. In this study, the potential targets and pharmacological effects of phlorotannins in the treatment of T2DM were determined basis on network pharmacology and enzyme activity inhibition experiment. In conclusion, the results showed the value of phlorotannins treating on T2DM. Moreover, this study has great significance for improving the medicinal value of phlorotannins and screening natural products for the treatment of T2DM.
Collapse
Affiliation(s)
- Jialiang Chen
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Ping Li
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Shuhong Ye
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Wei Li
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, Daegu, South Korea
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yan Ding
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
3
|
Gong W, Chen X, Shi T, Shao X, An X, Qin J, Chen X, Jiang Q, Guo B. Network Pharmacology-Based Strategy for the Investigation of the Anti-Osteoporosis Effects and Underlying Mechanism of Zhuangguguanjie Formulation. Front Pharmacol 2021; 12:727808. [PMID: 34658868 PMCID: PMC8517248 DOI: 10.3389/fphar.2021.727808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/12/2021] [Indexed: 11/18/2022] Open
Abstract
As the society is aging, the increasing prevalence of osteoporosis has generated huge social and economic impact, while the drug therapy for osteoporosis is limited due to multiple targets involved in this disease. Zhuangguguanjie formulation (ZG) is extensively used in the clinical treatment of bone and joint diseases, but the underlying mechanism has not been fully described. This study aimed to examine the therapeutic effect and potential mechanism of ZG on postmenopausal osteoporosis. The ovariectomized (OVX) mice were treated with normal saline or ZG for 4 weeks after ovariectomy following a series of analyses. The bone mass density (BMD) and trabecular parameters were examined by micro-CT. Bone remodeling was evaluated by the bone histomorphometry analysis and ELISA assay of bone turnover biomarkers in serum. The possible drug–disease common targets were analyzed by network pharmacology. To predict the potential biological processes and related pathways, GO/KEGG enrichment analysis was performed. The effects of ZG on the differentiation phenotype of osteoclasts and osteoblasts and the predicted pathway were verified in vitro. The results showed that ZG significantly improved the bone mass and micro-trabecular architecture in OVX mice compared with untreated OVX mice. ZG could promote bone formation and inhibit bone resorption to ameliorate ovariectomy-induced osteoporosis as evidenced by increased number of osteoblast (N.Ob/Tb.Pm) and decreased number of osteoclast (N.Oc/Tb.Pm) in treated group compared with untreated OVX mice. After identifying potential drug–disease common targets by network pharmacology, GO enrichment analysis predicted that ZG might affect various biological processes including osteoblastic differentiation and osteoclast differentiation. The KEGG enrichment analysis suggested that PI3K/Akt and mTOR signaling pathways could be the possible pathways. Furthermore, the experiments in vitro validated our findings. ZG significantly down-regulated the expression of osteoclast differentiation markers, reduced osteoclastic resorption, and inhibited the phosphorylation of PI3K/Akt, while ZG obviously up-regulated the expression of osteogenic biomarkers, promoted the formation of calcium nodules, and hampered the phosphorylation of 70S6K1/mTOR, which can be reversed by the corresponding pathway activator. Thus, our study suggested that ZG could inhibit the PI3K/Akt signaling pathway to reduce osteoclastic bone resorption as well as hamper the mTORC1/S6K1 signaling pathway to promote osteoblastic bone formation.
Collapse
Affiliation(s)
- Wang Gong
- Department of Sports Medicine and Adult Reconstructive Surgery, The Affiliated Hospital of Nanjing University Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Xingren Chen
- Department of Sports Medicine and Adult Reconstructive Surgery, The Affiliated Hospital of Nanjing University Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Tianshu Shi
- Department of Sports Medicine and Adult Reconstructive Surgery, The Affiliated Hospital of Nanjing University Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Xiaoyan Shao
- Department of Sports Medicine and Adult Reconstructive Surgery, The Affiliated Hospital of Nanjing University Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Xueying An
- Department of Sports Medicine and Adult Reconstructive Surgery, The Affiliated Hospital of Nanjing University Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Jianghui Qin
- Department of Sports Medicine and Adult Reconstructive Surgery, The Affiliated Hospital of Nanjing University Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Xiang Chen
- Department of Sports Medicine and Adult Reconstructive Surgery, The Affiliated Hospital of Nanjing University Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, The Affiliated Hospital of Nanjing University Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China.,Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Medical School, Nanjing University, Nanjing, China
| | - Baosheng Guo
- Department of Sports Medicine and Adult Reconstructive Surgery, The Affiliated Hospital of Nanjing University Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
4
|
Wang H, Tan H, Zhan W, Song L, Zhang D, Chen X, Lin Z, Wang W, Yang Y, Wang L, Bei W, Guo J. Molecular mechanism of Fufang Zhenzhu Tiaozhi capsule in the treatment of type 2 diabetes mellitus with nonalcoholic fatty liver disease based on network pharmacology and validation in minipigs. JOURNAL OF ETHNOPHARMACOLOGY 2021; 274:114056. [PMID: 33771638 DOI: 10.1016/j.jep.2021.114056] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fufang Zhenzhu Tiaozhi formula (FTZ) of which a patented preparation of Chinese herbal medicine has been well documented with significant clinical curative effect for hyperglycemia and hyperlipidemia. Because of the complexity of the chemical constituents of Chinese herbal formulas, the holistic pharmacological mechanism of FTZ acting on type 2 diabetes mellitus (T2DM) and nonalcoholic fatty liver disease (NAFLD) remains unclear. AIM OF THE STUDY To investigate the pharmacological efficacy and mechanism of FTZ in the treatment of T2DM accompanied by NAFLD. MATERIALS AND METHODS Network pharmacology and validation in minipigs were used in this study. First, potential bioactive compounds of FTZ were identified by the traditional Chinese medicine system pharmacology technology platform (TCMSP). Then, targets of compounds were gathered using DrugBank, SwissTargetPrediction and TCMSP, while targets for T2DM and NAFLD were collected from CTD (compounds-targets-diseases network) and GeneCards. Common targets were defined as direct therapeutic targets acting on T2DM with NAFLD. In addition, crucial targets were chosen by the protein-protein interaction (PPI) network and contribution to compound-therapeutic targets in T2DM with the NAFLD network. Furthermore, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to analyze the metabolism-related signaling pathways affected by FTZ. Candidate patterns selected by network pharmacology were tested in the minipigs model of T2DM with NAFLD. Measurements of triglycerides (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), fasting insulin (FINS) and fasting blood glucose (FBG) in the blood and the expression levels of proteins, including PI3K-AKT and HIF-1α, in the livers of the minipigs were followed by the administration of FTZ. RESULTS A total of 116 active compounds and 82 potential targets related to T2DM and NAFLD were found. Pathway and functional enrichment analysis showed that FTZ mainly regulates metabolism-related pathways, including PI3K-AKT, HIF-1α, TNFα and MAPK. Animal experiments showed that FTZ treatment significantly reduced the serum levels of TG, TC, LDL-C and FBG, increased serum levels of HDL-C, ameliorated systemic insulin resistance (IR), and attenuated liver damage in minipigs with T2DM and NAFLD. FTZ treatment has an obviously favorable influence on hepatic steatosis and liver lipid accumulation in the histopathologic features of HE, Oil red O staining, and electron microscopy. Mechanistically, FTZ improved liver metabolism by increasing the phosphorylation of PI3K-AKT and decreasing the expression of HIF-1α. CONCLUSION Network pharmacology was supported by experimental studies, which indicated that FTZ has demonstrated therapeutic benefits in T2DM and NAFLD by regulating the PI3K-AKT and HIF-1α signaling pathways.
Collapse
MESH Headings
- Animals
- Blood Glucose/drug effects
- Capsules
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Hypoglycemic Agents/chemistry
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Hypolipidemic Agents/chemistry
- Hypolipidemic Agents/pharmacology
- Hypolipidemic Agents/therapeutic use
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Insulin/blood
- Lipid Metabolism/drug effects
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Male
- Metabolic Networks and Pathways/drug effects
- Non-alcoholic Fatty Liver Disease/drug therapy
- Non-alcoholic Fatty Liver Disease/metabolism
- Non-alcoholic Fatty Liver Disease/pathology
- Pharmacology/methods
- Phosphatidylinositol 3-Kinases/metabolism
- Phytochemicals/analysis
- Phytochemicals/pharmacology
- Phytochemicals/therapeutic use
- Proto-Oncogene Proteins c-akt/metabolism
- Reproducibility of Results
- Swine
- Swine, Miniature
Collapse
Affiliation(s)
- Hong Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| | - Haibo Tan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| | - Wenjing Zhan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| | - Lixia Song
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| | - Dongxing Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| | - Xu Chen
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| | - Ziyang Lin
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| | - Weixuan Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| | - Yiqi Yang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| | - Lexun Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| | - Weijian Bei
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, China.
| |
Collapse
|
5
|
The crosstalk between platelets and body fat: A reverse translational study. Clin Nutr 2020; 40:2025-2034. [PMID: 33008652 DOI: 10.1016/j.clnu.2020.09.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 08/18/2020] [Accepted: 09/13/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Our previous study found that platelet counts were positively associated with body fat percentage in human. In the present study, we conducted a reverse translational study to explore the role of platelets in modulating pre-adipocyte proliferation in mice. METHODS Mouse pre-adipocyte cell line (3T3-L1) and human pre-adipocytes harvested from female subcutaneous fat were used. Pre-adipocytes were co-cultured with platelets or platelet releasate, which were isolated from mice or humans. The cell viability and proliferative ability of the pre-adipocytes were examined by MTT and flow cytometry assays. Western blotting analysis was used to determine the phosphorylation levels of proteins in the mTOR pathway. RESULTS The number of platelets in the adipose tissues from obese mice was significantly higher than that from lean mice. Platelets and collagen-activated platelet releasate stimulated the proliferation of human pre-adipocytes and 3T3-L1 cells in vitro. Besides, platelets from obese mice were more potent in stimulating pre-adipocyte proliferation than those from lean control mice. Mechanistically, platelets enhanced pre-adipocyte proliferation through the acceleration of cell cycle progression from G0/G1 to S phase cell cycle progression. At the molecular level, platelets promoted pre-adipocyte proliferation through mTOR pathway-mediated upregulation of cyclin D1 expression. CONCLUSION In conclusion, platelets and platelet releasate play an important role in the proliferation of pre-adipocytes. Our study may provide new clues and the molecular mechanism of the causal pathways between platelets and body fat to explain the finding we observed in population study.
Collapse
|
6
|
Gao PF, Huang D, Wen JY, Liu W, Zhang HW. Advances in the role of exosomal non-coding RNA in the development, diagnosis, and treatment of gastric cancer (Review). Mol Clin Oncol 2020; 13:101-108. [PMID: 32714531 DOI: 10.3892/mco.2020.2068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 05/15/2020] [Indexed: 02/06/2023] Open
Abstract
Exosomes are small vesicles secreted by a variety of cells that contain vrious biological macromolecules, including RNA, non-coding RNA and protein. An increasing number of studies have demonstrated that exosomes and particularly the non-coding RNAs they contain, serve important roles in many cellular processes, including the transmission of information. It is well established that the occurrence and development of gastric cancer, one of the four most common malignant tumors worldwide, involves the transmission of information. Based on the urgent need for the elucidation of the mechanism involved in this process, as well as advances in the diagnosis and treatment of gastric cancer, numerous reports have assessed the association between non-coding RNAs in exosomes and gastric cancer. The purpose of the present review was to summarize recent evidence on certain non-coding RNAs associated with the development, diagnosis and treatment of gastric cancer.
Collapse
Affiliation(s)
- Peng-Fei Gao
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Da Huang
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jun-Yan Wen
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wei Liu
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Hong-Wu Zhang
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
7
|
Phan TK, Bindra GK, Williams SA, Poon IK, Hulett MD. Combating Human Pathogens and Cancer by Targeting Phosphoinositides and Their Metabolism. Trends Pharmacol Sci 2019; 40:866-882. [DOI: 10.1016/j.tips.2019.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2022]
|
8
|
Zhang X, Wang S, Wang H, Cao J, Huang X, Chen Z, Xu P, Sun G, Xu J, Lv J, Xu Z. Circular RNA circNRIP1 acts as a microRNA-149-5p sponge to promote gastric cancer progression via the AKT1/mTOR pathway. Mol Cancer 2019; 18:20. [PMID: 30717751 PMCID: PMC6360801 DOI: 10.1186/s12943-018-0935-5] [Citation(s) in RCA: 592] [Impact Index Per Article: 98.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 12/27/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND CircRNA has emerged as a new non-coding RNA that plays crucial roles in tumour initiation and development. 'MiRNA sponge' is the most reported role played by circRNAs in many tumours. The AKT/mTOR axis is a classic signalling pathway in cancers that sustains energy homeostasis through energy production activities, such as the Warburg effect, and blocks catabolic activities, such as autophagy. Additionally, the AKT/mTOR axis exerts a positive effect on EMT, which promotes tumour metastasis. METHODS We detected higher circNRIP1 expression in gastric cancer by performing RNA-seq analysis. We verified the tumour promotor role of circNRIP1 in gastric cancer cells through a series of biological function assays. We then used a pull-down assay and dual-luciferase reporter assay to identify the downstream miR-149-5p of circNRIP1. Western blot analysis and immunofluorescence assays were performed to demonstrate that the circNRIP1-miR-149-5p-AKT1/mTOR axis is responsible for the altered metabolism in GC cells and promotes GC development. We then adopted a co-culture system to trace circNRIP1 transmission via exosomal communication and RIP experiments to determine that quaking regulates circNRIP1 expression. Finally, we confirmed the tumour suppressor role of microRNA-133a-3p in vivo in PDX mouse models. RESULTS We discovered that knockdown of circNRIP1 successfully blocked proliferation, migration, invasion and the expression level of AKT1 in GC cells. MiR-149-5p inhibition phenocopied the overexpression of circNRIP1 in GC cells, and overexpression of miR-149-5p blocked the malignant behaviours of circNRIP1. Moreover, it was proven that circNRIP1 can be transmitted by exosomal communication between GC cells, and exosomal circNRIP1 promoted tumour metastasis in vivo. We also demonstrated that quaking can promote circNRIP1 transcription. In the final step, the tumour promotor role of circNRIP1 was verified in PDX models. CONCLUSIONS We proved that circNRIP1 sponges miR-149-5p to affect the expression level of AKT1 and eventually acts as a tumour promotor in GC.
Collapse
Affiliation(s)
- Xing Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Sen Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Haixiao Wang
- Department of General Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu, China
| | - Jiacheng Cao
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Xiaoxu Huang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Zheng Chen
- Department of Surgical Oncology, University of Miami, Miami, USA
| | - Penghui Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Guangli Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Jianghao Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Jialun Lv
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China.
- Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
9
|
Suman S, Das TP, Sirimulla S, Alatassi H, Ankem MK, Damodaran C. Withaferin-A suppress AKT induced tumor growth in colorectal cancer cells. Oncotarget 2017; 7:13854-64. [PMID: 26883103 PMCID: PMC4924683 DOI: 10.18632/oncotarget.7351] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/29/2016] [Indexed: 12/12/2022] Open
Abstract
The oncogenic activation of AKT gene has emerged as a key determinant of the aggressiveness of colorectal cancer (CRC); hence, research has focused on targeting AKT signaling for the treatment of advanced stages of CRC. In this study, we explored the anti-tumorigenic effects of withaferin A (WA) on CRC cells overexpressing AKT in preclinical (in vitro and in vivo) models. Our results indicated that WA, a natural compound, resulted in significant inhibition of AKT activity and led to the inhibition of cell proliferation, migration and invasion by downregulating the epithelial to mesenchymal transition (EMT) markers in CRC cells overexpressing AKT. The oral administration of WA significantly suppressed AKT-induced aggressive tumor growth in a xenograft model. Molecular analysis revealed that the decreased expression of AKT and its downstream pro-survival signaling molecules may be responsible for tumor inhibition. Further, significant inhibition of some important EMT markers, i.e., Snail, Slug, β-catenin and vimentin, was observed in WA-treated human CRC cells overexpressing AKT. Significant inhibition of micro-vessel formation and the length of vessels were evident in WA-treated tumors, which correlated with a low expression of the angiogenic marker RETIC. In conclusion, the present study emphasizes the crucial role of AKT activation in inducing cell proliferation, angiogenesis and EMT in CRC cells and suggests that WA may overcome AKT-induced cell proliferation and tumor growth in CRC.
Collapse
Affiliation(s)
- Suman Suman
- Department of Urology, University of Louisville, Louisville, KY 40202, USA
| | - Trinath P Das
- Department of Urology, University of Louisville, Louisville, KY 40202, USA
| | - Suman Sirimulla
- Department of Basic Sciences, St. Louis College of Pharmacy, St. Louis, MO 63110, USA
| | - Houda Alatassi
- Department of Pathology, University of Louisville, Louisville, KY 40202, USA
| | - Murali K Ankem
- Department of Urology, University of Louisville, Louisville, KY 40202, USA
| | - Chendil Damodaran
- Department of Urology, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
10
|
Szigeti B, Roth YD, Sekar JAP, Goldberg AP, Pochiraju SC, Karr JR. A blueprint for human whole-cell modeling. ACTA ACUST UNITED AC 2017; 7:8-15. [PMID: 29806041 DOI: 10.1016/j.coisb.2017.10.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Whole-cell dynamical models of human cells are a central goal of systems biology. Such models could help researchers understand cell biology and help physicians treat disease. Despite significant challenges, we believe that human whole-cell models are rapidly becoming feasible. To develop a plan for achieving human whole-cell models, we analyzed the existing models of individual cellular pathways, surveyed the biomodeling community, and reflected on our experience developing whole-cell models of bacteria. Based on these analyses, we propose a plan for a project, termed the Human Whole-Cell Modeling Project, to achieve human whole-cell models. The foundations of the plan include technology development, standards development, and interdisciplinary collaboration.
Collapse
Affiliation(s)
- Balázs Szigeti
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Yosef D Roth
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - John A P Sekar
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Arthur P Goldberg
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Saahith C Pochiraju
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Jonathan R Karr
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| |
Collapse
|
11
|
Xu M, Tang X, Guo J, Sun W, Tang F. Reversal effect of adenovirus-mediated human interleukin 24 transfection on the cisplatin resistance of A549/DDP lung cancer cells. Oncol Rep 2017; 38:2843-2851. [PMID: 29048638 PMCID: PMC5780038 DOI: 10.3892/or.2017.6002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 09/18/2017] [Indexed: 01/11/2023] Open
Abstract
Interleukin-24 (IL-24) is a tumor-suppressor gene that has been documented in human melanoma cells. IL-24 has marked antitumor activities on various types of human cancer, but its underlying mechanism remains unclear. In the present, we investigated the effects of human IL-24 (hIL-24) on the chemotherapy resistance of lung cancer cells. The cisplatin (DDP)-resistant lung carcinoma cell line A549/DDP was subjected to adenovirus-mediated transfection with the human IL-24 gene (Ad-hIL-24). The growth-inhibitory and apoptotic effects of Ad-hIL-24 on A549/DDP cells were observed, and the expression levels of AKT, phosphorylated-AKT (p-AKT) and P-glycoprotein (P-gp) were detected. Ad-hIL-24 significantly decreased the levels of p-AKT and P-gp, and effectively inhibited A549/DDP cell growth. Furthermore, A549/DDP cells exhibited a significantly increased rate of apoptosis, as well as G2/M-phase arrest, following transfection with Ad-hIL-24, and these effects were increased in cells treated with Ad-IL-24 combined with DDP when compared with those treated with Ad-hIL-24 or DDP alone. These results suggest that hIL-24 can reverse the DDP resistance of lung cancer cells, and that the associated mechanism involves the induction of apoptosis and G2/M-phase arrest through the phosphoinositide3-kinase (PI3K)/AKT signaling pathway, as well as a decrease in drug resistance through P-gp expression.
Collapse
Affiliation(s)
- Mingju Xu
- Department of Clinical Laboratory of Zhuhai Hospital, Jinan University and Zhuhai People's Hospital, Zhuhai, Guangdong 519000, P.R. China
| | - Xioawei Tang
- Metallurgical Science and Engineering, Central South University, Changsha, Hunan 410083, P.R. China
| | - Jinjin Guo
- Zhuhai Campus, Zunyi Medical College, Zhuhai, Guangdong 519041, P.R. China
| | - Wangbang Sun
- Zhuhai Campus, Zunyi Medical College, Zhuhai, Guangdong 519041, P.R. China
| | - Faqing Tang
- Department of Clinical Laboratory of Zhuhai Hospital, Jinan University and Zhuhai People's Hospital, Zhuhai, Guangdong 519000, P.R. China
| |
Collapse
|
12
|
Mediani L, Gibellini F, Bertacchini J, Frasson C, Bosco R, Accordi B, Basso G, Bonora M, Calabrò ML, Mattiolo A, Sgarbi G, Baracca A, Pinton P, Riva G, Rampazzo E, Petrizza L, Prodi L, Milani D, Luppi M, Potenza L, De Pol A, Cocco L, Capitani S, Marmiroli S. Reversal of the glycolytic phenotype of primary effusion lymphoma cells by combined targeting of cellular metabolism and PI3K/Akt/ mTOR signaling. Oncotarget 2016; 7:5521-37. [PMID: 26575168 PMCID: PMC4868703 DOI: 10.18632/oncotarget.6315] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/27/2015] [Indexed: 12/12/2022] Open
Abstract
PEL is a B-cell non-Hodgkin lymphoma, occurring predominantly as a lymphomatous effusion in body cavities, characterized by aggressive clinical course, with no standard therapy. Based on previous reports that PEL cells display a Warburg phenotype, we hypothesized that the highly hypoxic environment in which they grow in vivo makes them more reliant on glycolysis, and more vulnerable to drugs targeting this pathway. We established here that indeed PEL cells in hypoxia are more sensitive to glycolysis inhibition. Furthermore, since PI3K/Akt/mTOR has been proposed as a drug target in PEL, we ascertained that pathway-specific inhibitors, namely the dual PI3K and mTOR inhibitor, PF-04691502, and the Akt inhibitor, Akti 1/2, display improved cytotoxicity to PEL cells in hypoxic conditions. Unexpectedly, we found that these drugs reduce lactate production/extracellular acidification rate, and, in combination with the glycolysis inhibitor 2-deoxyglucose (2-DG), they shift PEL cells metabolism from aerobic glycolysis towards oxidative respiration. Moreover, the associations possess strong synergistic cytotoxicity towards PEL cells, and thus may reduce adverse reaction in vivo, while displaying very low toxicity to normal lymphocytes. Finally, we showed that the association of 2-DG and PF-04691502 maintains its cytotoxic and proapoptotic effect also in PEL cells co-cultured with human primary mesothelial cells, a condition known to mimic the in vivo environment and to exert a protective and pro-survival action. All together, these results provide a compelling rationale for the clinical development of new therapies for the treatment of PEL, based on combined targeting of glycolytic metabolism and constitutively activated signaling pathways.
Collapse
Affiliation(s)
- Laura Mediani
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Federica Gibellini
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Jessika Bertacchini
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy.,Department of Morphology, Surgery and Experimental Medicine, Section of Anatomy and Histology and LTTA Center, University of Ferrara, Ferrara, Italy
| | - Chiara Frasson
- Department of Woman's and Child's Health and Institute of Pediatric Research - Città della Speranza Foundation, University of Padova, Padova, Italy
| | - Raffaella Bosco
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Benedetta Accordi
- Department of Woman's and Child's Health and Institute of Pediatric Research - Città della Speranza Foundation, University of Padova, Padova, Italy
| | - Giuseppe Basso
- Department of Woman's and Child's Health and Institute of Pediatric Research - Città della Speranza Foundation, University of Padova, Padova, Italy
| | - Massimo Bonora
- Department of Morphology, Surgery and Experimental Medicine Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy
| | - Maria Luisa Calabrò
- Immunology and Molecular Oncology, Veneto Institute of Oncology, IOV IRCCS, Padova, Italy
| | - Adriana Mattiolo
- Immunology and Molecular Oncology, Veneto Institute of Oncology, IOV IRCCS, Padova, Italy
| | - Gianluca Sgarbi
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandra Baracca
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy
| | - Giovanni Riva
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, AOU Policlinico, Modena, Italy
| | - Enrico Rampazzo
- Department of Chemistry, University of Bologna, Bologna, Italy
| | - Luca Petrizza
- Department of Chemistry, University of Bologna, Bologna, Italy
| | - Luca Prodi
- Department of Chemistry, University of Bologna, Bologna, Italy
| | - Daniela Milani
- Department of Morphology, Surgery and Experimental Medicine, Section of Anatomy and Histology and LTTA Center, University of Ferrara, Ferrara, Italy
| | - Mario Luppi
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, AOU Policlinico, Modena, Italy
| | - Leonardo Potenza
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, AOU Policlinico, Modena, Italy
| | - Anto De Pol
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucio Cocco
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Silvano Capitani
- Department of Morphology, Surgery and Experimental Medicine, Section of Anatomy and Histology and LTTA Center, University of Ferrara, Ferrara, Italy
| | - Sandra Marmiroli
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
13
|
Staphylococcus aureus Infection Reduces Nutrition Uptake and Nucleotide Biosynthesis in a Human Airway Epithelial Cell Line. Metabolites 2016; 6:metabo6040041. [PMID: 27834866 PMCID: PMC5192447 DOI: 10.3390/metabo6040041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 10/28/2016] [Accepted: 11/02/2016] [Indexed: 11/16/2022] Open
Abstract
The Gram positive opportunistic human pathogen Staphylococcus aureus induces a variety of diseases including pneumonia. S. aureus is the second most isolated pathogen in cystic fibrosis patients and accounts for a large proportion of nosocomial pneumonia. Inside the lung, the human airway epithelium is the first line in defence with regard to microbial recognition and clearance as well as regulation of the immune response. The metabolic host response is, however, yet unknown. To address the question of whether the infection alters the metabolome and metabolic activity of airway epithelial cells, we used a metabolomics approach. The nutrition uptake by the human airway epithelial cell line A549 was monitored over time by proton magnetic resonance spectroscopy (1H-NMR) and the intracellular metabolic fingerprints were investigated by gas chromatography and high performance liquid chromatography (GC-MS) and (HPLC-MS). To test the metabolic activity of the host cells, glutamine analogues and labelled precursors were applied after the infection. We found that A549 cells restrict uptake of essential nutrients from the medium after S. aureus infection. Moreover, the infection led to a shutdown of the purine and pyrimidine synthesis in the A549 host cell, whereas other metabolic routes such as the hexosamine biosynthesis pathway remained active. In summary, our data show that the infection with S. aureus negatively affects growth, alters the metabolic composition and specifically impacts the de novo nucleotide biosynthesis in this human airway epithelial cell model.
Collapse
|
14
|
Politz O, Siegel F, Bärfacker L, Bömer U, Hägebarth A, Scott WJ, Michels M, Ince S, Neuhaus R, Meyer K, Fernández-Montalván AE, Liu N, von Nussbaum F, Mumberg D, Ziegelbauer K. BAY 1125976, a selective allosteric AKT1/2 inhibitor, exhibits high efficacy on AKT signaling-dependent tumor growth in mouse models. Int J Cancer 2016; 140:449-459. [PMID: 27699769 DOI: 10.1002/ijc.30457] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 08/24/2016] [Indexed: 12/26/2022]
Abstract
The PI3K-AKT-mTOR signaling cascade is activated in the majority of human cancers, and its activation also plays a key role in resistance to chemo and targeted therapeutics. In particular, in both breast and prostate cancer, increased AKT pathway activity is associated with cancer progression, treatment resistance and poor disease outcome. Here, we evaluated the activity of a novel allosteric AKT1/2 inhibitor, BAY 1125976, in biochemical, cellular mechanistic, functional and in vivo efficacy studies in a variety of tumor models. In in vitro kinase activity assays, BAY 1125976 potently and selectively inhibited the activity of full-length AKT1 and AKT2 by binding into an allosteric binding pocket formed by kinase and PH domain. In accordance with this proposed allosteric binding mode, BAY 1125976 bound to inactive AKT1 and inhibited T308 phosphorylation by PDK1, while the activity of truncated AKT proteins lacking the pleckstrin homology domain was not inhibited. In vitro, BAY 1125976 inhibited cell proliferation in a broad panel of human cancer cell lines. Particularly high activity was observed in breast and prostate cancer cell lines expressing estrogen or androgen receptors. Furthermore, BAY 1125976 exhibited strong in vivo efficacy in both cell line and patient-derived xenograft models such as the KPL4 breast cancer model (PIK3CAH1074R mutant), the MCF7 and HBCx-2 breast cancer models and the AKTE17K mutant driven prostate cancer (LAPC-4) and anal cancer (AXF 984) models. These findings indicate that BAY 1125976 is a potent and highly selective allosteric AKT1/2 inhibitor that targets tumors displaying PI3K/AKT/mTOR pathway activation, providing opportunities for the clinical development of new, effective treatments.
Collapse
Affiliation(s)
- Oliver Politz
- Bayer AG, Global Drug Discovery, 13342 Berlin, Germany
| | | | - Lars Bärfacker
- Bayer AG, Global Drug Discovery, 42096 Wuppertal, Germany
| | - Ulf Bömer
- Bayer AG, Global Drug Discovery, 13342 Berlin, Germany
| | | | - William J Scott
- Bayer HealthCare Pharmaceuticals Inc, Whippany, NJ 07981-0915
| | - Martin Michels
- Bayer AG, Global Drug Discovery, 42096 Wuppertal, Germany
| | - Stuart Ince
- Bayer HealthCare Pharmaceuticals Inc, Whippany, NJ 07981-0915
| | | | - Kirstin Meyer
- Bayer AG, Global Drug Discovery, 13342 Berlin, Germany
| | | | - Ningshu Liu
- Bayer AG, Global Drug Discovery, 13342 Berlin, Germany
| | - Franz von Nussbaum
- Bayer S.A.S./Bayer CropScience, Small Molecules Research, Disease Control Chemistry, 69263 Lyon, France
| | | | | |
Collapse
|
15
|
Silveira CGT, Marschner G, Canny GO, Klocke S, Hunold P, Köster F, Ahrens T, Rody A, Hornung D. Disrupting Y-Box-Binding Protein 1 Function Using OSU-03012 Prevents Endometriosis Progression in In Vitro and In Vivo Models. Reprod Sci 2016; 24:67-76. [PMID: 27217374 DOI: 10.1177/1933719116649695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The objective of the present study was to test the ability of OSU-03012 (2-amino-N-[4-[5-phenanthren-2-yl-3-(trifluoromethyl)pyrazol-1-yl]phenyl]acetamide), a novel and potent celecoxib-derivative, to impair endometriosis progression in in vitro and in vivo models based on its ability to indirectly block Y-box-binding protein 1 (YB-1) function. 12Z human endometriotic epithelial cells and sexually mature female C57BL/6J mice were treated with OSU-03012. Cellular proliferation was quantified by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromid assay. Expression of YB-1 and phosphorylated YB-1 in 12Z cells and endometriotic lesions was evaluated by Western blotting and immunohistochemistry (IHC). The IHC for proliferating cell nuclear antigen was performed. OSU-03012 treatment resulted in decreased YB-1 and its phosphorylated form in both in vitro and in vivo models. Endometriotic lesion size was significantly reduced in OSU-03012-treated mice (27.6 ± 4.0 mm3) compared to those from the control group (50.5 ± 6.9 mm3, P < .0001). A significant reduction in endometriotic epithelial cell proliferation was observed in endometriotic lesions exposed to OSU-03012 treatment ( P = .0346). In conclusion, targeting YB-1 via OSU-03012 showed a potent antiproliferative effect on endometriotic epithelial cells in vitro and in a mouse model of disease.
Collapse
Affiliation(s)
- Cássia G T Silveira
- 1 Department of Obstetrics and Gynecology, University of Lübeck, Lübeck, Germany
| | - Gabriele Marschner
- 1 Department of Obstetrics and Gynecology, University of Lübeck, Lübeck, Germany
| | | | - Silke Klocke
- 1 Department of Obstetrics and Gynecology, University of Lübeck, Lübeck, Germany
| | - Peter Hunold
- 3 Clinic for Radiology and Nuclear Medicine, University of Lübeck, Lübeck, Germany
| | - Frank Köster
- 1 Department of Obstetrics and Gynecology, University of Lübeck, Lübeck, Germany
| | - Thorben Ahrens
- 1 Department of Obstetrics and Gynecology, University of Lübeck, Lübeck, Germany
| | - Achim Rody
- 1 Department of Obstetrics and Gynecology, University of Lübeck, Lübeck, Germany
| | - Daniela Hornung
- 1 Department of Obstetrics and Gynecology, University of Lübeck, Lübeck, Germany.,4 Department of Gynecology and Obstetrics, Diakonissenkrankenhaus Karlsruhe Rüppurr, Karlsruhe, Germany
| |
Collapse
|
16
|
Jiao Y, Zhang J, Lu L, Xu J, Qin L. The Fto Gene Regulates the Proliferation and Differentiation of Pre-Adipocytes in Vitro. Nutrients 2016; 8:102. [PMID: 26907332 PMCID: PMC4772064 DOI: 10.3390/nu8020102] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/22/2016] [Accepted: 02/15/2016] [Indexed: 12/17/2022] Open
Abstract
The highly regulated differentiation and proliferation of pre-adipocytes play a key role in the initiation of obesity. Fat mass and obesity associated (FTO) is a novel gene strongly associated with the risk of obesity. A deficiency of FTO may cause growth retardation in addition to fat mass and adipocyte size reduction in vivo. To investigate the potential role of Fto gene on the proliferation and differentiation of pre-adipocytes, we generated Fto-knockdown and overexpressed 3T3-L1 cells. Using numerous proliferation assays our results suggest that Fto knockdown leads to suppression of proliferation, lower mitochondrial membrane potential, less cellular ATP, and decreased and smaller intracellular lipid droplets compared with controls (p < 0.05). Western blot analysis demonstrated that Fto knockdown can significantly suppress peroxisome proliferator-activated receptor gamma (PPARγ) and glucose transporter type 4 (GLUT4) expression and inhibit Akt phosphorylation. By contrast, overexpression of Fto had the opposing effect on proliferation, mitochondrial membrane potential, ATP generation, in vitro differentiation, Akt phosphorylation, and PPARγ and GLUT4 expression. Moreover, we demonstrated that Wortmannin, a phosphoinositide 3-kinase (PI3K) inhibitor, could inhibit phospho-Akt in Fto overexpressed 3T3-L1 cells. Taken together, the results suggest that Fto regulates the proliferation and differentiation of 3T3-L1 cells via multiple mechanisms, including PPARγ and PI3K/Akt signaling.
Collapse
Affiliation(s)
- Yang Jiao
- Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou 215123, China.
- Department of Radiation Genetics, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.
| | - Jingying Zhang
- Department of Radiation Genetics, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.
| | - Lunjie Lu
- Department of Radiation Genetics, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.
| | - Jiaying Xu
- Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou 215123, China.
| | - Liqiang Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Medical College of Soochow University, Suzhou 215123, China.
| |
Collapse
|
17
|
Allegretti M, Ricciardi MR, Licchetta R, Mirabilii S, Orecchioni S, Reggiani F, Talarico G, Foà R, Bertolini F, Amadori S, Torrisi MR, Tafuri A. The pan-class I phosphatidyl-inositol-3 kinase inhibitor NVP-BKM120 demonstrates anti-leukemic activity in acute myeloid leukemia. Sci Rep 2015; 5:18137. [PMID: 26674543 PMCID: PMC4682184 DOI: 10.1038/srep18137] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/16/2015] [Indexed: 12/14/2022] Open
Abstract
Aberrant activation of the PI3K/Akt/mTOR pathway is a common feature of acute myeloid leukemia (AML) patients contributing to chemoresistance, disease progression and unfavourable outcome. Therefore, inhibition of this pathway may represent a potential therapeutic approach in AML. The aim of this study was to evaluate the pre-clinical activity of NVP-BKM120 (BKM120), a selective pan-class I PI3K inhibitor, on AML cell lines and primary samples. Our results demonstrate that BKM120 abrogates the activity of the PI3K/Akt/mTOR signaling, promoting cell growth arrest and significant apoptosis in a dose- and time-dependent manner in AML cells but not in the normal counterpart. BKM120-induced cytotoxicity is associated with a profound modulation of metabolic behaviour in both cell lines and primary samples. In addition, BKM120 synergizes with the glycolitic inhibitor dichloroacetate enhancing apoptosis induction at lower doses. Finally, in vivo administration of BKM120 to a xenotransplant mouse model of AML significantly inhibited leukemia progression and improved the overall survival of treated mice. Taken together, our findings indicate that BKM120, alone or in combination with other drugs, has a significant anti-leukemic activity supporting its clinical development as a novel therapeutic agent in AML.
Collapse
MESH Headings
- Adult
- Aged
- Aminopyridines/pharmacology
- Animals
- Blotting, Western
- Cell Line, Tumor
- Cell Survival/drug effects
- Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors
- Class I Phosphatidylinositol 3-Kinases/metabolism
- Dose-Response Relationship, Drug
- Female
- HL-60 Cells
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Middle Aged
- Morpholines/pharmacology
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction/drug effects
- TOR Serine-Threonine Kinases/metabolism
- Time Factors
- U937 Cells
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Matteo Allegretti
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Maria Rosaria Ricciardi
- Department of Clinical and Molecular Medicine, “Sant’Andrea” Hospital, Sapienza University of Rome, Rome, Italy
| | - Roberto Licchetta
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Simone Mirabilii
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Stefania Orecchioni
- Division of Clinical Haematology-Oncology, European Institute of Oncology, Milan, Italy
| | - Francesca Reggiani
- Division of Clinical Haematology-Oncology, European Institute of Oncology, Milan, Italy
| | - Giovanna Talarico
- Division of Clinical Haematology-Oncology, European Institute of Oncology, Milan, Italy
| | - Roberto Foà
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Francesco Bertolini
- Division of Clinical Haematology-Oncology, European Institute of Oncology, Milan, Italy
| | - Sergio Amadori
- Department of Hematology, Tor Vergata University Hospital, Rome, Italy
| | - Maria Rosaria Torrisi
- Department of Clinical and Molecular Medicine, “Sant’Andrea” Hospital, Sapienza University of Rome, Rome, Italy
| | - Agostino Tafuri
- Department of Clinical and Molecular Medicine, “Sant’Andrea” Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
18
|
Abeyrathna P, Su Y. The critical role of Akt in cardiovascular function. Vascul Pharmacol 2015; 74:38-48. [PMID: 26025205 PMCID: PMC4659756 DOI: 10.1016/j.vph.2015.05.008] [Citation(s) in RCA: 317] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/07/2015] [Accepted: 05/16/2015] [Indexed: 12/30/2022]
Abstract
Akt kinase, a member of AGC kinases, is important in many cellular functions including proliferation, migration, cell growth and metabolism. There are three known Akt isoforms which play critical and diverse roles in the cardiovascular system. Akt activity is regulated by its upstream regulatory pathways at transcriptional and post-translational levels. Beta-catenin/Tcf-4, GLI1 and Stat-3 are some of few known transcriptional regulators of AKT gene. Threonine 308 and serine 473 are the two critical phosphorylation sites of Akt1. Translocation of Akt to the cell membrane facilitates PDK1 phosphorylation of the threonine site. The serine site is phosphorylated by mTORC2. Ack1, Src, PTK6, TBK1, IKBKE and IKKε are some of the non-canonical pathways which affect the Akt activity. Protein-protein interactions of Akt to actin and Hsp90 increase the Akt activity while Akt binding to other proteins such as CTMP and TRB3 reduces the Akt activity. The action of Akt on its downstream targets determines its function in cardiovascular processes such as cell survival, growth, proliferation, angiogenesis, vasorelaxation, and cell metabolism. Akt promotes cell survival via caspase-9, YAP, Bcl-2, and Bcl-x activities. Inhibition of FoxO proteins by Akt also increases cell survival by transcriptional mechanisms. Akt stimulates cell growth and proliferation through mTORC1. Akt also increases VEGF secretion and mediates eNOS phosphorylation, vasorelaxation and angiogenesis. Akt can increase cellular metabolism through its downstream targets GSK3 and GLUT4. The alterations of Akt signaling play an important role in many cardiovascular pathological processes such as atherosclerosis, cardiac hypertrophy, and vascular remodeling. Several Akt inhibitors have been developed and tested as anti-tumor agents. They could be potential novel therapeutics for the cardiovascular diseases.
Collapse
Affiliation(s)
- Prasanna Abeyrathna
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Yunchao Su
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
19
|
Pseudolaric acid B exerts antitumor activity via suppression of the Akt signaling pathway in HeLa cervical cancer cells. Mol Med Rep 2015; 12:2021-6. [DOI: 10.3892/mmr.2015.3615] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 03/18/2015] [Indexed: 11/05/2022] Open
|
20
|
Mulukutla BC, Yongky A, Grimm S, Daoutidis P, Hu WS. Multiplicity of steady states in glycolysis and shift of metabolic state in cultured mammalian cells. PLoS One 2015; 10:e0121561. [PMID: 25806512 PMCID: PMC4373774 DOI: 10.1371/journal.pone.0121561] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 02/11/2015] [Indexed: 01/23/2023] Open
Abstract
Cultured mammalian cells exhibit elevated glycolysis flux and high lactate production. In the industrial bioprocesses for biotherapeutic protein production, glucose is supplemented to the culture medium to sustain continued cell growth resulting in the accumulation of lactate to high levels. In such fed-batch cultures, sometimes a metabolic shift from a state of high glycolysis flux and high lactate production to a state of low glycolysis flux and low lactate production or even lactate consumption is observed. While in other cases with very similar culture conditions, the same cell line and medium, cells continue to produce lactate. A metabolic shift to lactate consumption has been correlated to the productivity of the process. Cultures that exhibited the metabolic shift to lactate consumption had higher titers than those which didn't. However, the cues that trigger the metabolic shift to lactate consumption state (or low lactate production state) are yet to be identified. Metabolic control of cells is tightly linked to growth control through signaling pathways such as the AKT pathway. We have previously shown that the glycolysis of proliferating cells can exhibit bistability with well-segregated high flux and low flux states. Low lactate production (or lactate consumption) is possible only at a low glycolysis flux state. In this study, we use mathematical modeling to demonstrate that lactate inhibition together with AKT regulation on glycolysis enzymes can profoundly influence the bistable behavior, resulting in a complex steady-state topology. The transition from the high flux state to the low flux state can only occur in certain regions of the steady state topology, and therefore the metabolic fate of the cells depends on their metabolic trajectory encountering the region that allows such a metabolic state switch. Insights from such switch behavior present us with new means to control the metabolism of mammalian cells in fed-batch cultures.
Collapse
Affiliation(s)
- Bhanu Chandra Mulukutla
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Andrew Yongky
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Simon Grimm
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Prodromos Daoutidis
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Wei-Shou Hu
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
21
|
Castellanos-Martín A, Castillo-Lluva S, Sáez-Freire MDM, Blanco-Gómez A, Hontecillas-Prieto L, Patino-Alonso C, Galindo-Villardon P, Pérez del Villar L, Martín-Seisdedos C, Isidoro-Garcia M, Abad-Hernández MDM, Cruz-Hernández JJ, Rodríguez-Sánchez CA, González-Sarmiento R, Alonso-López D, De Las Rivas J, García-Cenador B, García-Criado J, Lee DY, Bowen B, Reindl W, Northen T, Mao JH, Pérez-Losada J. Unraveling heterogeneous susceptibility and the evolution of breast cancer using a systems biology approach. Genome Biol 2015; 16:40. [PMID: 25853295 PMCID: PMC4389302 DOI: 10.1186/s13059-015-0599-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 01/27/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND An essential question in cancer is why individuals with the same disease have different clinical outcomes. Progress toward a more personalized medicine in cancer patients requires taking into account the underlying heterogeneity at different molecular levels. RESULTS Here, we present a model in which there are complex interactions at different cellular and systemic levels that account for the heterogeneity of susceptibility to and evolution of ERBB2-positive breast cancers. Our model is based on our analyses of a cohort of mice that are characterized by heterogeneous susceptibility to ERBB2-positive breast cancers. Our analysis reveals that there are similarities between ERBB2 tumors in humans and those of backcross mice at clinical, genomic, expression, and signaling levels. We also show that mice that have tumors with intrinsically high levels of active AKT and ERK are more resistant to tumor metastasis. Our findings suggest for the first time that a site-specific phosphorylation at the serine 473 residue of AKT1 modifies the capacity for tumors to disseminate. Finally, we present two predictive models that can explain the heterogeneous behavior of the disease in the mouse population when we consider simultaneously certain genetic markers, liver cell signaling and serum biomarkers that are identified before the onset of the disease. CONCLUSIONS Considering simultaneously tumor pathophenotypes and several molecular levels, we show the heterogeneous behavior of ERBB2-positive breast cancer in terms of disease progression. This and similar studies should help to better understand disease variability in patient populations.
Collapse
Affiliation(s)
- Andrés Castellanos-Martín
- />Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca/CSIC, Salamanca, 37007 Spain
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Current address: Institute for Research in Biomedicine (IRB Barcelona), C/Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Sonia Castillo-Lluva
- />Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca/CSIC, Salamanca, 37007 Spain
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
| | - María del Mar Sáez-Freire
- />Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca/CSIC, Salamanca, 37007 Spain
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca, 37007 Spain
| | - Adrián Blanco-Gómez
- />Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca/CSIC, Salamanca, 37007 Spain
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
| | - Lourdes Hontecillas-Prieto
- />Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca/CSIC, Salamanca, 37007 Spain
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
| | - Carmen Patino-Alonso
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Departamento de Estadística, Universidad de Salamanca, Salamanca, 37007 Spain
| | - Purificación Galindo-Villardon
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Departamento de Estadística, Universidad de Salamanca, Salamanca, 37007 Spain
| | - Luis Pérez del Villar
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Departamento de Parasitología CIETUS, Universidad de Salamanca, Salamanca, 37007 Spain
| | - Carmen Martín-Seisdedos
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Servicio de Bioquímica Clínica, Hospital Universitario de Salamanca, Salamanca, 37007 Spain
| | - María Isidoro-Garcia
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Servicio de Bioquímica Clínica, Hospital Universitario de Salamanca, Salamanca, 37007 Spain
| | - María del Mar Abad-Hernández
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Departamento de Anatomía Patológica, Facultad de Medicina Universidad de Salamanca, Salamanca, 37007 Spain
| | - Juan Jesús Cruz-Hernández
- />Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca/CSIC, Salamanca, 37007 Spain
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Departamento de Medicina, Universidad de Salamanca, Salamanca, 37007 Spain
- />Servicio de Oncología, Hospital Universitario de Salamanca, Salamanca, 37007 Spain
| | - César Augusto Rodríguez-Sánchez
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Servicio de Oncología, Hospital Universitario de Salamanca, Salamanca, 37007 Spain
| | - Rogelio González-Sarmiento
- />Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca/CSIC, Salamanca, 37007 Spain
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Departamento de Medicina, Universidad de Salamanca, Salamanca, 37007 Spain
| | - Diego Alonso-López
- />Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca/CSIC, Salamanca, 37007 Spain
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Unidad de Bioinformática, CIC-IBMCC, Salamanca, 37007 Spain
| | - Javier De Las Rivas
- />Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca/CSIC, Salamanca, 37007 Spain
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Unidad de Bioinformática, CIC-IBMCC, Salamanca, 37007 Spain
| | - Begoña García-Cenador
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Departamento de Cirugía, Universidad de Salamanca, Salamanca, 37007 Spain
| | - Javier García-Criado
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
- />Departamento de Cirugía, Universidad de Salamanca, Salamanca, 37007 Spain
| | - Do Yup Lee
- />Advanced Fermentation Fusion Science and Technology, Kookmin University, Seoul, 136-702 Korea
| | - Benjamin Bowen
- />Department of Bioenergy/GTL & Structural Biology, Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA
| | - Wolfgang Reindl
- />Department of Bioenergy/GTL & Structural Biology, Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA
| | - Trent Northen
- />Department of Bioenergy/GTL & Structural Biology, Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA
| | - Jian-Hua Mao
- />Life Sciences Division, Lawrence Berkeley National Laboratory (LBNL), University of California, Berkeley, CA 94720 USA
| | - Jesús Pérez-Losada
- />Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca/CSIC, Salamanca, 37007 Spain
- />Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, 37007 Spain
| |
Collapse
|
22
|
Xiaoji Decoction (消积饮) inhibited cell proliferation and induced apoptosis through Akt signaling pathway in human lung cancer A549 cells. Chin J Integr Med 2014; 20:701-5. [DOI: 10.1007/s11655-014-1772-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Indexed: 12/13/2022]
|
23
|
Marín-Hernández A, López-Ramírez SY, Gallardo-Pérez JC, Rodríguez-Enríquez S, Moreno-Sánchez R, Saavedra E. Systems Biology Approaches to Cancer Energy Metabolism. SYSTEMS BIOLOGY OF METABOLIC AND SIGNALING NETWORKS 2014. [DOI: 10.1007/978-3-642-38505-6_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
24
|
Cloud infrastructures for in silico drug discovery: economic and practical aspects. BIOMED RESEARCH INTERNATIONAL 2013; 2013:138012. [PMID: 24106693 PMCID: PMC3782806 DOI: 10.1155/2013/138012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 06/26/2013] [Accepted: 06/27/2013] [Indexed: 11/17/2022]
Abstract
Cloud computing opens new perspectives for small-medium biotechnology laboratories that need to perform bioinformatics analysis in a flexible and effective way. This seems particularly true for hybrid clouds that couple the scalability offered by general-purpose public clouds with the greater control and ad hoc customizations supplied by the private ones. A hybrid cloud broker, acting as an intermediary between users and public providers, can support customers in the selection of the most suitable offers, optionally adding the provisioning of dedicated services with higher levels of quality. This paper analyses some economic and practical aspects of exploiting cloud computing in a real research scenario for the in silico drug discovery in terms of requirements, costs, and computational load based on the number of expected users. In particular, our work is aimed at supporting both the researchers and the cloud broker delivering an IaaS cloud infrastructure for biotechnology laboratories exposing different levels of nonfunctional requirements.
Collapse
|
25
|
Suman S, Kurisetty V, Das TP, Vadodkar A, Ramos G, Lakshmanaswamy R, Damodaran C. Activation of AKT signaling promotes epithelial-mesenchymal transition and tumor growth in colorectal cancer cells. Mol Carcinog 2013; 53 Suppl 1:E151-60. [PMID: 24000138 DOI: 10.1002/mc.22076] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 07/11/2013] [Accepted: 07/15/2013] [Indexed: 02/01/2023]
Affiliation(s)
- Suman Suman
- Department of Biomedical Sciences; Paul L Foster School of Medicine; Texas Tech University Health Sciences Center; El Paso Texas
| | - Vittal Kurisetty
- Department of Biomedical Sciences; Paul L Foster School of Medicine; Texas Tech University Health Sciences Center; El Paso Texas
| | - Trinath P. Das
- Department of Biomedical Sciences; Paul L Foster School of Medicine; Texas Tech University Health Sciences Center; El Paso Texas
| | - Aditi Vadodkar
- Department of Biomedical Sciences; Paul L Foster School of Medicine; Texas Tech University Health Sciences Center; El Paso Texas
| | - Gabriel Ramos
- Department of Biomedical Sciences; Paul L Foster School of Medicine; Texas Tech University Health Sciences Center; El Paso Texas
| | - Rajkumar Lakshmanaswamy
- Department of Biomedical Sciences; Paul L Foster School of Medicine; Texas Tech University Health Sciences Center; El Paso Texas
| | - Chendil Damodaran
- Department of Biomedical Sciences; Paul L Foster School of Medicine; Texas Tech University Health Sciences Center; El Paso Texas
| |
Collapse
|
26
|
Zhang M, Peng L, Qiao ZB, He HT, Zhou Y, Xu Z. Inhibition of the PI3K/Akt signaling pathway inhibits cell proliferation and induces apoptosis in hepatocellular carcinoma cell line HepG2. Shijie Huaren Xiaohua Zazhi 2013; 21:2250-2257. [DOI: 10.11569/wcjd.v21.i23.2250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of the PI3K/Akt signaling pathway inhibitor wortmannin on cell proliferation, apoptosis and expression of pAkt, Skp2 and P27kip1 in human hepatocellular carcinoma cell line HepG2.
METHODS: After treatment with different concentrations of wortmannin (0, 10, 50, 100, 200 nmol/L) for different durations (3, 10, 24 h), proliferation of HepG2 cells was analyzed by MTT assay, cell cycle and apoptosis were detected by flow cytometry, expression of pAkt, Skp2 and P27kip1 proteins was detected by Western blot, and the mRNA expression of Skp2 and P27kip1 was detected by reverse transcription-polymerase chain reaction.
RESULTS: Wortmannin inhibited the proliferation of HepG2 cells in a dose- and time-dependent manner. The apoptosis rates of HepG2 cells significantly increased from 8.46% ± 1.17% to 28.03% ± 2.67% after treatment with wortmannin (P < 0.05). Wortmannin induced an increase in the percentage of cells in G0/G1 phase and a decrease in the percentage of cells in S phase cells (both P < 0.05). After treatment with wortmannin, the expression of pAkt and Skp2 proteins was down-regulated, while that of P27kip1 protein was up-regulated (all P < 0.05). In addition, Skp2 mRNA expression in HepG2 cells was significantly down-regulated (P < 0.05), although the expression of P27kip1 mRNA was not changed.
CONCLUSION: Wortmannin inhibits cell proliferation and induces apoptosis in human hepatocellular carcinoma cell line HepG2 possibly by regulating the expression of Skp2 and P27kip1.
Collapse
|
27
|
Eisenreich W, Heesemann J, Rudel T, Goebel W. Metabolic host responses to infection by intracellular bacterial pathogens. Front Cell Infect Microbiol 2013; 3:24. [PMID: 23847769 PMCID: PMC3705551 DOI: 10.3389/fcimb.2013.00024] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/11/2013] [Indexed: 12/12/2022] Open
Abstract
The interaction of bacterial pathogens with mammalian hosts leads to a variety of physiological responses of the interacting partners aimed at an adaptation to the new situation. These responses include multiple metabolic changes in the affected host cells which are most obvious when the pathogen replicates within host cells as in case of intracellular bacterial pathogens. While the pathogen tries to deprive nutrients from the host cell, the host cell in return takes various metabolic countermeasures against the nutrient theft. During this conflicting interaction, the pathogen triggers metabolic host cell responses by means of common cell envelope components and specific virulence-associated factors. These host reactions generally promote replication of the pathogen. There is growing evidence that pathogen-specific factors may interfere in different ways with the complex regulatory network that controls the carbon and nitrogen metabolism of mammalian cells. The host cell defense answers include general metabolic reactions, like the generation of oxygen- and/or nitrogen-reactive species, and more specific measures aimed to prevent access to essential nutrients for the respective pathogen. Accurate results on metabolic host cell responses are often hampered by the use of cancer cell lines that already exhibit various de-regulated reactions in the primary carbon metabolism. Hence, there is an urgent need for cellular models that more closely reflect the in vivo infection conditions. The exact knowledge of the metabolic host cell responses may provide new interesting concepts for antibacterial therapies.
Collapse
Affiliation(s)
- Wolfgang Eisenreich
- Lehrstuhl für Biochemie, Center of Isotopologue Profiling, Technische Universität München Garching, Germany
| | | | | | | |
Collapse
|
28
|
Simões AES, Pereira DM, Amaral JD, Nunes AF, Gomes SE, Rodrigues PM, Lo AC, D'Hooge R, Steer CJ, Thibodeau SN, Borralho PM, Rodrigues CMP. Efficient recovery of proteins from multiple source samples after TRIzol(®) or TRIzol(®)LS RNA extraction and long-term storage. BMC Genomics 2013; 14:181. [PMID: 23496794 PMCID: PMC3620933 DOI: 10.1186/1471-2164-14-181] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Accepted: 03/06/2013] [Indexed: 11/10/2022] Open
Abstract
Background Simultaneous isolation of nucleic acids and proteins from a single biological sample facilitates meaningful data interpretation and reduces time, cost and sampling errors. This is particularly relevant for rare human and animal specimens, often scarce, and/or irreplaceable. TRIzol® and TRIzol®LS are suitable for simultaneous isolation of RNA, DNA and proteins from the same biological sample. These reagents are widely used for RNA and/or DNA isolation, while reports on their use for protein extraction are limited, attributable to technical difficulties in protein solubilisation. Results TRIzol®LS was used for RNA isolation from 284 human colon cancer samples, including normal colon mucosa, tubulovillous adenomas, and colon carcinomas with proficient and deficient mismatch repair system. TRIzol® was used for RNA isolation from human colon cancer cells, from brains of transgenic Alzheimer’s disease mice model, and from cultured mouse cortical neurons. Following RNA extraction, the TRIzol®-chloroform fractions from human colon cancer samples and from mouse hippocampus and frontal cortex were stored for 2 years and 3 months, respectively, at −80°C until used for protein isolation. Simple modifications to the TRIzol® manufacturer’s protocol, including Urea:SDS solubilization and sonication, allowed improved protein recovery yield compared to the TRIzol® manufacturer’s protocol. Following SDS-PAGE and Ponceau and Coomassie staining, recovered proteins displayed wide molecular weight range and staining pattern comparable to those obtainable with commonly used protein extraction protocols. We also show that nuclear and cytosolic proteins can be easily extracted and detected by immunoblotting, and that posttranslational modifications, such as protein phosphorylation, are detectable in proteins recovered from TRIzol®-chloroform fractions stored for up to 2 years at −80°C. Conclusions We provide a novel approach to improve protein recovery from samples processed for nucleic acid extraction with TRIzol® and TRIzol®LS compared to the manufacturer`s protocol, allowing downstream immunoblotting and evaluation of steady-state relative protein expression levels. The method was validated in large sets of samples from multiple sources, including human colon cancer and brains of transgenic Alzheimer’s disease mice model, stored in TRIzol®-chloroform for up to two years. Collectively, we provide a faster and cheaper alternative to the TRIzol® manufacturer`s protein extraction protocol, illustrating the high relevance, and wide applicability, of the present protein isolation method for the immunoblot evaluation of steady-state relative protein expression levels in samples from multiple sources, and following prolonged storage.
Collapse
Affiliation(s)
- André E S Simões
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
MAJ CARLO, MOSCA ETTORE, MERELLI IVAN, MAURI GIANCARLO, MILANESI LUCIANO. SENSITIVITY ANALYSIS FOR STUDYING THE RELATION BETWEEN BIOCHEMICAL REACTIONS AND METABOLIC PHENOTYPES. J Bioinform Comput Biol 2013; 11:1340002. [DOI: 10.1142/s0219720013400027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Metabolic models are the most widespread type of models in systems biology and are currently used in several applications, including metabolic engineering and investigations of pathological states in which metabolic disorders play a relevant role. Once a model has been defined and corroborated, sensitivity analysis techniques can be used to study the model behavior in relation to perturbations of the model parameters. Here, we describe how it is possible to combine regionalized sensitivity analysis and response surface methodology to screen and quantitatively characterize the relation between metabolic phenotypes and biochemical reactions rates. By means of this approach, we identified the most important reactions for the citric acid efflux from mitochondria, one of the key metabolic traits of cancer cells.
Collapse
Affiliation(s)
- CARLO MAJ
- Istituto Tecnologie Biomediche – Consiglio Nazionale Ricerche, Via F.lli Cervi 93, 20090 Segrate, Italy
- Dipartimento di Informatica, Sistemistica e Comunicazione, Università Degli Studi di Milano-Bicocca, Viale Sarca 336, Milano 20126, Italy
| | - ETTORE MOSCA
- Istituto Tecnologie Biomediche – Consiglio Nazionale Ricerche, Via F.lli Cervi 93, 20090 Segrate, Italy
| | - IVAN MERELLI
- Istituto Tecnologie Biomediche – Consiglio Nazionale Ricerche, Via F.lli Cervi 93, 20090 Segrate, Italy
| | - GIANCARLO MAURI
- Dipartimento di Informatica, Sistemistica e Comunicazione, Università Degli Studi di Milano-Bicocca, Viale Sarca 336, Milano 20126, Italy
| | - LUCIANO MILANESI
- Istituto Tecnologie Biomediche – Consiglio Nazionale Ricerche, Via F.lli Cervi 93, 20090 Segrate, Italy
| |
Collapse
|
30
|
Mosca E, Alfieri R, Maj C, Bevilacqua A, Canti G, Milanesi L. Computational modeling of the metabolic States regulated by the kinase akt. Front Physiol 2012. [PMID: 23181020 PMCID: PMC3502886 DOI: 10.3389/fphys.2012.00418] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Signal transduction and gene regulation determine a major reorganization of metabolic activities in order to support cell proliferation. Protein Kinase B (PKB), also known as Akt, participates in the PI3K/Akt/mTOR pathway, a master regulator of aerobic glycolysis and cellular biosynthesis, two activities shown by both normal and cancer proliferating cells. Not surprisingly considering its relevance for cellular metabolism, Akt/PKB is often found hyperactive in cancer cells. In the last decade, many efforts have been made to improve the understanding of the control of glucose metabolism and the identification of a therapeutic window between proliferating cancer cells and proliferating normal cells. In this context, we have modeled the link between the PI3K/Akt/mTOR pathway, glycolysis, lactic acid production, and nucleotide biosynthesis. We used a computational model to compare two metabolic states generated by two different levels of signaling through the PI3K/Akt/mTOR pathway: one of the two states represents the metabolism of a growing cancer cell characterized by aerobic glycolysis and cellular biosynthesis, while the other state represents the same metabolic network with a reduced glycolytic rate and a higher mitochondrial pyruvate metabolism. Biochemical reactions that link glycolysis and pentose phosphate pathway revealed their importance for controlling the dynamics of cancer glucose metabolism.
Collapse
Affiliation(s)
- Ettore Mosca
- Institute for Biomedical Technologies, Consiglio Nazionale delle Ricerche Segrate Milano, Italy
| | | | | | | | | | | |
Collapse
|
31
|
Jang S, Park JW, Cha HR, Jung SY, Lee JE, Jung SS, Kim JO, Kim SY, Lee CS, Park HS. Silver nanoparticles modify VEGF signaling pathway and mucus hypersecretion in allergic airway inflammation. Int J Nanomedicine 2012; 7:1329-43. [PMID: 22457593 PMCID: PMC3310409 DOI: 10.2147/ijn.s27159] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The anti-inflammatory action of silver nanoparticles (NPs) has been reported in a murine model of asthma in a previous study. But more specific mechanisms of silver NPs in an attenuation of allergic airway inflammation have not yet been established. Vascular and mucous changes are believed to contribute largely in pathophysiology in asthma. Among various factors related to vascular changes, vascular endothelial growth factor (VEGF) plays a pivotal role in vascular changes in asthma. Mucin proteins MUC5AC and MUC5B have been implicated as markers of goblet cell metaplasia in lung pathologies. The aim of this study was to investigate the effects of silver NPs on VEGF signaling pathways and mucus hypersecretion. Ovalbumin (OVA)-inhaled female BALBc mice were used to evaluate the role of silver NPs and the related molecular mechanisms in allergic airway disease. In this study, with an OVA-induced murine model of allergic airway disease, it was found that the increased levels of hypoxia-inducible factor (HIF)-1α, VEGF, phosphatidylinositol-3 kinase (PI3K) and phosphorylated-Akt levels, and mucous glycoprotein expression (Muc5ac) in lung tissues were substantially decreased by the administration of silver NPs. In summary, silver NPs substantially suppressed mucus hypersecretion and PI3K/HIF-1α/VEGF signaling pathway in an allergic airway inflammation.
Collapse
Affiliation(s)
- Sunhyae Jang
- Division of Pulmonology, Allergy and Critical Care Medicine, Department of Internal Medicine, Chungnam National University Medical School, Daesadong, Daejeon, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Xia Y, Liu Y, Rocchi P, Wang M, Fan Y, Qu F, Iovanna JL, Peng L. Targeting heat shock factor 1 with a triazole nucleoside analog to elicit potent anticancer activity on drug-resistant pancreatic cancer. Cancer Lett 2012; 318:145-53. [PMID: 22266185 DOI: 10.1016/j.canlet.2011.09.043] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Revised: 09/28/2011] [Accepted: 09/29/2011] [Indexed: 11/26/2022]
Abstract
Issued from a lead optimization process, we have identified a novel triazole nucleoside analog which elicits potent anticancer activity on drug-resistant pancreatic cancer. Most importantly, this compound targets heat shock response pathways by down-regulation of heat shock transcription factor 1 and consequential down-regulation of multiple heat shock proteins HSP27, HSP70 and HSP90. Down-regulation of these proteins caused the shut-down of several oncogenic pathways and caspase-dependent apoptosis resulting in a potent anticancer effect in vitro and in vivo. These results demonstrate the potential rewards gained in searching for anticancer candidates with multimodal actions on heat shock response pathways via HSF1 down-regulation.
Collapse
Affiliation(s)
- Yi Xia
- Aix-Marseille Université, Centre Interdisciplinaire de Nanoscience de Marseille, Marseille, France
| | | | | | | | | | | | | | | |
Collapse
|