1
|
Wang J, Zhang HM, Zhu GH, Zhao LL, Shi J, Dai ZT, Li JP, Li XR, Sun F, Wu Y, Chen SY, Li HN, Liao XH, Xiang Y. STT3-mediated aberrant N-glycosylation of CD24 inhibits paclitaxel sensitivity in triple-negative breast cancer. Acta Pharmacol Sin 2025; 46:1097-1110. [PMID: 39668180 PMCID: PMC11950364 DOI: 10.1038/s41401-024-01419-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/27/2024] [Indexed: 12/14/2024]
Abstract
Paclitaxel is one of the main chemotherapic medicines against triple-negative breast cancer (TNBC) in clinic. However, it has been perplexed by paclitaxel resistance in TNBC patients, resulting in a poor prognosis. Abnormal protein glycosylation is closely related to the occurrence and progression of tumors and malignant phenotypes such as chemotherapy resistance. CD24 is a highly glycosylated membrane protein that is highly expressed in TNBC, leading to tumorigenesis and poor prognosis. In this study we investigated the relationship between abnormal glycosylation of CD24 and paclitaxel susceptibility in TNBC and the molecular mechanisms. We showed that CD24 protein levels were significantly up-regulated in both TNBC tissues and cells, and CD24 protein was highly glycosylated. Genetic and pharmacological inhibition of N-glycosylation of CD24 enhances the anticancer activity of paclitaxel in vitro and tumor xenograft models. We revealed that the molecular mechanism of N-glycosylation of CD24 in paclitaxel resistance involved inhibition of ferroptosis, a new form that regulates cell death. Inhibition of N-glycosylation of CD24 increased glutathione consumption, iron content, and lipid peroxidation, resulting in paclitaxel-induced ferroptosis. We demonstrated that endoplasmic reticulum (ER)-associated glycosyltransferase STT3 isoforms (including both STT3A and STT3B isoforms) enable N-glycosylation of the L-asparagine (N) site. Knockout of the endogenous STT3 isoform in TNBC cells partially reduced the glycosylation status of CD24. Our results demonstrate the critical role of N-glycosylation of CD24 in weakening drug sensitivity by inhibiting ferroptosis, highlighting new insights that targeting N-glycosylation of CD24 has great potential to promote chemotherapy sensitivity and efficacy.
Collapse
Affiliation(s)
- Jun Wang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, 430081, China
- Department of Materials Science, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Hui-Min Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, 430081, China
- Department of Human Anatomy&Histoembryology, School of Basic Medical sciences, Xinxiang Medical University, Xinxiang, 453000, China
| | - Guan-Hua Zhu
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Li-Li Zhao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Ji Shi
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Zhou-Tong Dai
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, 430081, China
- Department of Gynaecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia-Peng Li
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Xing-Rui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fan Sun
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Yuan Wu
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China
| | - Shao-Yong Chen
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, 430081, China.
| | - Han-Ning Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xing-Hua Liao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, 430081, China.
| | - Yuan Xiang
- Department of Medical Laboratory, Tongji Medical College, Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430014, China.
| |
Collapse
|
2
|
Chen L, Zhao X, Sheng R, Lazarovici P, Zheng W. Artemisinin alleviates astrocyte overactivation and neuroinflammation by modulating the IRE1/NF-κB signaling pathway in in vitro and in vivo Alzheimer's disease models. Free Radic Biol Med 2025; 229:96-110. [PMID: 39826816 DOI: 10.1016/j.freeradbiomed.2025.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/25/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Recent studies have shown that neuroinflammation and heightened glial activity, particularly astrocyte overactivation, are associated with Alzheimer's disease (AD). Abnormal accumulation of amyloid-beta (Aβ) induces endoplasmic reticulum (ER) stress and activates astrocytes. Artemisinin (ART), a frontline anti-malarial drug, has been found to have neuroprotective properties. However, its impact on astrocytes remains unclear. In this study, we used Aβ1-42 induced astrocyte cultures and 3 × Tg-AD mice as in vitro and in vivo models, respectively, to investigate the effects of ART on AD related astrocyte overactivation and its underlying mechanisms. ART attenuated Aβ1-42-induced astrocyte activation, ER stress, and inflammatory responses in astrocyte cultures by inhibiting IRE1 phosphorylation and the NF-κB pathway, as evidenced by the overexpression of IRE1 WT and IRE1-K599A (kinase activity invalidated), along with application of activators and inhibitors related to ER stress. Furthermore, ART alleviated the detrimental effects and restored neurotrophic function of astrocytes on co-cultured neurons, preventing neuronal apoptosis during Aβ1-42 treatment. In 3 × Tg-AD mice, ART treatment improved cognitive function and reduced astrocyte overactivation, neuroinflammation, ER stress, and neuronal apoptosis. Moreover, ART attenuated the upregulation of IRE1/NF-κB pathway activity in AD mice. Astrocyte-specific overexpression of IRE1 via adeno-associated virus in AD mice reversed the ameliorating effects of ART. Our findings suggest that ART inhibits astrocyte overactivation and neuroinflammation in both in vitro and in vivo AD models by modulating the IRE1/NF-κB signaling pathway, thereby enhancing neuronal functions. This study underscores the therapeutic potential of ART in AD and highlights the significance of modulating the ER stress-inflammatory cycle and normalizing astrocyte-neuron communication.
Collapse
Affiliation(s)
- Lei Chen
- Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Macau, Taipa, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, China
| | - Xia Zhao
- Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Macau, Taipa, China
| | - Rui Sheng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, China.
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112002, Israel
| | - Wenhua Zheng
- Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Macau, Taipa, China.
| |
Collapse
|
3
|
Goswami N, Kinkpe L, Hua L, Zhuo Y, Fang Z, Che L, Lin Y, Xu S, Jiang X, Feng B, Wu D. Farnesol Improves Endoplasmic Reticulum Stress and Hepatic Metabolic Dysfunction Induced by Tunicamycin in Mice. BIOLOGY 2025; 14:213. [PMID: 40001981 PMCID: PMC11851907 DOI: 10.3390/biology14020213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025]
Abstract
Endoplasmic reticulum (ER) stress significantly affects liver metabolism, often leading to disorders such as hepatic steatosis. Tunicamycin (TM), a known ER stress inducer, is frequently used to model metabolic stress, but its specific effects on liver energy homeostasis remain unclear. This study investigates how farnesol (FOH), a natural compound with antioxidant and anti-inflammatory properties, counteracts TM-induced ER stress and its associated metabolic disruptions in the liver. Using both primary hepatocytes and a mouse model, this study demonstrates that TM treatment caused upregulation of ER stress markers, including ATF4, and disrupted genes related to lipid metabolism and gluconeogenesis. Co-treatment with FOH reduced these stress markers and restored the expression of metabolic genes. In vivo, FOH treatment alleviated oxidative stress, reduced lipid accumulation, and restored normal glycogen and lipid metabolism. Histological analysis further confirmed that FOH preserved liver architecture and minimized cellular damage. FOH also stabilized serum lipid profiles and modulated key metabolic biomarkers, suggesting its protective role against TM-induced liver injury. These findings suggest that FOH has therapeutic potential in mitigating ER stress-related metabolic dysfunctions, offering promising insights for the treatment of liver diseases linked to metabolic stress.
Collapse
Affiliation(s)
- Naqash Goswami
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (N.G.); (L.H.); (Y.Z.); (Z.F.); (L.C.); (Y.L.); (S.X.); (X.J.)
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling 712100, China;
| | - Lionel Kinkpe
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling 712100, China;
| | - Lun Hua
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (N.G.); (L.H.); (Y.Z.); (Z.F.); (L.C.); (Y.L.); (S.X.); (X.J.)
| | - Yong Zhuo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (N.G.); (L.H.); (Y.Z.); (Z.F.); (L.C.); (Y.L.); (S.X.); (X.J.)
| | - Zhengfeng Fang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (N.G.); (L.H.); (Y.Z.); (Z.F.); (L.C.); (Y.L.); (S.X.); (X.J.)
| | - Lianqiang Che
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (N.G.); (L.H.); (Y.Z.); (Z.F.); (L.C.); (Y.L.); (S.X.); (X.J.)
| | - Yan Lin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (N.G.); (L.H.); (Y.Z.); (Z.F.); (L.C.); (Y.L.); (S.X.); (X.J.)
| | - Shengyu Xu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (N.G.); (L.H.); (Y.Z.); (Z.F.); (L.C.); (Y.L.); (S.X.); (X.J.)
| | - Xuemei Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (N.G.); (L.H.); (Y.Z.); (Z.F.); (L.C.); (Y.L.); (S.X.); (X.J.)
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (N.G.); (L.H.); (Y.Z.); (Z.F.); (L.C.); (Y.L.); (S.X.); (X.J.)
| | - De Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (N.G.); (L.H.); (Y.Z.); (Z.F.); (L.C.); (Y.L.); (S.X.); (X.J.)
| |
Collapse
|
4
|
Wang L, Shi R, Wang S, Duan Y, Wang Z, Zheng P, Sun X, Chen X, Ji G, Shen Y, Dong B, Lin Y, Wen T, Tian Q, Guo Z, Hou Y, Wu S, Xiao L, Li M, Xiao L, Wu Q, Meng Y, Liu G, Duan S, Bai X, Liu T, Zhang Z, Zhan P, Lu Z, Xu D. ADSL promotes autophagy and tumor growth through fumarate-mediated Beclin1 dimethylation. Nat Chem Biol 2025:10.1038/s41589-024-01825-9. [PMID: 39881212 DOI: 10.1038/s41589-024-01825-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 12/11/2024] [Indexed: 01/31/2025]
Abstract
As an enzyme with a critical role in de novo purine synthesis, adenylosuccinate lyase (ADSL) expression is upregulated in various malignancies. However, whether ADSL possesses noncanonical functions that contribute to cancer progression remains poorly understood. Here, we demonstrate that protein kinase R-like endoplasmic reticulum kinase (PERK) activated by lipid deprivation or ER stress phosphorylates ADSL at S140, leading to an enhanced association between ADSL and Beclin1. Beclin1-associated ADSL produces fumarate, which in turn inhibits lysine demethylase 8-mediated Beclin1 demethylation, resulting in enhanced Beclin1 K117me2, subsequent disruption of the binding of BCL-2 to Beclin1 and elevated autophagy. Blocking the ADSL-Beclin1 axis by knock-in mutation or a cell-penetrating peptide inhibits autophagy induced by lipid deprivation and ER stress and blunts liver tumor growth in mice. Additionally, ADSL pS140-upregulated Beclin1 K117me2 levels are positively correlated with autophagy levels in human hepatocellular carcinoma specimens and poor patient prognosis. These findings uncover the function of ADSL in autophagy regulation and liver tumor development.
Collapse
Affiliation(s)
- Lei Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Runze Shi
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuo Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yuran Duan
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Zheng Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Peixiang Zheng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Xue Sun
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaohan Chen
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guimei Ji
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Yuli Shen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Bofei Dong
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Yanni Lin
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Ting Wen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Qi Tian
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Zhanpeng Guo
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Yueru Hou
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Shiqi Wu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Ling Xiao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Min Li
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Liwei Xiao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Qingang Wu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Ying Meng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Guijun Liu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Sofie Duan
- Canyon Crest Academy, San Diego, CA, USA
| | - Xueli Bai
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Tong Liu
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
- Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Daqian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China.
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China.
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China.
| |
Collapse
|
5
|
Miao C, Huang Y, Zhang C, Wang X, Wang B, Zhou X, Song Y, Wu P, Chen ZS, Feng Y. Post-translational modifications in drug resistance. Drug Resist Updat 2025; 78:101173. [PMID: 39612546 DOI: 10.1016/j.drup.2024.101173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/24/2024] [Accepted: 11/16/2024] [Indexed: 12/01/2024]
Abstract
Resistance to antitumor drugs, antimicrobial drugs, and antiviral drugs severely limits treatment effectiveness and cure rate of diseases. Protein post-translational modifications (PTMs) represented by glycosylation, ubiquitination, SUMOylation, acetylation, phosphorylation, palmitoylation, and lactylation are closely related to drug resistance. PTMs are typically achieved by adding sugar chains (glycosylation), small proteins (ubiquitination), lipids (palmitoylation), or functional groups (lactylation) to amino acid residues. These covalent additions are usually the results of signaling cascades and could be reversible, with the triggering mechanisms depending on the type of modifications. PTMs are involved in antitumor drug resistance, not only as inducers of drug resistance but also as targets for reversing drug resistance. Bacteria exhibit multiple PTMs-mediated antimicrobial drug resistance. PTMs allow viral proteins and host cell proteins to form complex interaction networks, inducing complex antiviral drug resistance. This review summarizes the important roles of PTMs in drug resistance, providing new ideas for exploring drug resistance mechanisms, developing new drug targets, and guiding treatment plans.
Collapse
Affiliation(s)
- Chenggui Miao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei 230012, China; Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yurong Huang
- Department of Respiratory Medicine, Center of Infectious Diseases and Pathogen Biology, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, The First Hospital, Jilin University, Changchun 130021, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Xiao Wang
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Bing Wang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Xinyue Zhou
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yingqiu Song
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Peng Wu
- Department of Anatomy, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Zhe-Sheng Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong.
| |
Collapse
|
6
|
Zhong H, Zhou Z, Wang H, Wang R, Shen K, Huang R, Wang Z. The Biological Roles and Clinical Applications of the PI3K/AKT Pathway in Targeted Therapy Resistance in HER2-Positive Breast Cancer: A Comprehensive Review. Int J Mol Sci 2024; 25:13376. [PMID: 39769140 PMCID: PMC11677710 DOI: 10.3390/ijms252413376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Epidermal growth factor receptor 2-positive breast cancer (HER2+ BC) is a highly invasive and malignant type of tumor. Due to its resistance to HER2-targeted therapy, HER2+ BC has a poor prognosis and a tendency for metastasis. Understanding the mechanisms underlying this resistance and developing effective treatments for HER2+ BC are major research challenges. The phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT) pathway, which is frequently altered in cancers, plays a critical role in cellular proliferation and drug resistance. This signaling pathway activates various downstream pathways and exhibits complex interactions with other signaling networks. Given the significance of the PI3K/AKT pathway in HER2+ BC, several targeted drugs are currently in development. Multiple drugs have entered clinical trials or gained market approval, bringing new hope for HER2+ BC therapy. However, new drugs and therapies raise concerns related to safety, regulation, and ethics. Populations of different races and disease statuses exhibit varying responses to treatments. Therefore, in this review, we summarize current knowledge on the alteration and biological roles of the PI3K/AKT pathway, as well as its clinical applications and perspectives, providing new insights for advancing targeted therapies in HER2+ BC.
Collapse
Affiliation(s)
| | | | | | | | | | - Renhong Huang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (H.Z.); (Z.Z.); (H.W.); (R.W.); (K.S.)
| | - Zheng Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (H.Z.); (Z.Z.); (H.W.); (R.W.); (K.S.)
| |
Collapse
|
7
|
Han X, Qin H, Lu Y, Chen H, Yuan Z, Zhang Y, Yang X, Zheng L, Yan S. Post-translational modifications: The potential ways for killing cancer stem cells. Heliyon 2024; 10:e34015. [PMID: 39092260 PMCID: PMC11292267 DOI: 10.1016/j.heliyon.2024.e34015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/31/2024] Open
Abstract
While strides in cancer treatment continue to advance, the enduring challenges posed by cancer metastasis and recurrence persist as formidable contributors to the elevated mortality rates observed in cancer patients. Among the multifaceted factors implicated in tumor recurrence and metastasis, cancer stem cells (CSCs) emerge as noteworthy entities due to their inherent resistance to conventional therapies and heightened invasive capacities. Characterized by their notable abilities for self-renewal, differentiation, and initiation of tumorigenesis, the eradication of CSCs emerges as a paramount objective. Recent investigations increasingly emphasize the pivotal role of post-translational protein modifications (PTMs) in governing the self-renewal and replication capabilities of CSCs. This review accentuates the critical significance of several prevalent PTMs and the intricate interplay of PTM crosstalk in regulating CSC behavior. Furthermore, it posits that the manipulation of PTMs may offer a novel avenue for targeting and eliminating CSC populations, presenting a compelling perspective on cancer therapeutics with substantial potential for future applications.
Collapse
Affiliation(s)
- Xuedan Han
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Hai Qin
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, No. 206, Sixian Street, Baiyun District, Guiyang City, 550014, Guizhou Province, China
| | - Yu Lu
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Haitao Chen
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Zhengdong Yuan
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Yiwen Zhang
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Xuena Yang
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Simin Yan
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
8
|
Bharathiraja P, Yadav P, Sajid A, Ambudkar SV, Prasad NR. Natural medicinal compounds target signal transduction pathways to overcome ABC drug efflux transporter-mediated multidrug resistance in cancer. Drug Resist Updat 2023; 71:101004. [PMID: 37660590 PMCID: PMC10840887 DOI: 10.1016/j.drup.2023.101004] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/11/2023] [Accepted: 08/19/2023] [Indexed: 09/05/2023]
Abstract
ATP-binding cassette (ABC) transporters such as ABCB1, ABCG2, and ABCC1 are the major players in drug efflux-mediated multidrug resistance (MDR), which severely affects the efficacy of chemotherapy. Several synthetic compounds block the drug transport by ABC transporters; however, they exhibit a narrow therapeutic window, and produce side effects in non-target normal tissues. Conversely, the downregulation of the expression of ABC drug transporters seems to be a promising strategy to reverse MDR in cancer cells. Several signaling pathways, such as NF-κB, STAT3, Gli, NICD, YAP/TAZ, and Nrf2 upregulate the expression of ABC drug transporters in drug-resistant cancers. Recently, natural medicinal compounds have gained importance to overcome the ABC drug-efflux pump-mediated MDR in cancer. These compounds target transcription factors and the associated signal transduction pathways, thereby downregulating the expression of ABC transporters in drug-resistant cancer cells. Several potent natural compounds have been identified as lead candidates to synergistically enhance chemotherapeutic efficacy, and a few of them are already in clinical trials. Therefore, modulation of signal transduction pathways using natural medicinal compounds for the reversal of ABC drug transporter-mediated MDR in cancer is a novel approach for improving the efficiency of the existing chemotherapeutics. In this review, we discuss the modulatory role of natural medicinal compounds on cellular signaling pathways that regulate the expression of ABC transporters in drug-resistant cancer cells.
Collapse
Affiliation(s)
- Pradhapsingh Bharathiraja
- Department of Biochemistry & Biotechnology, Annamalai University, Annamalai Nagar 608 002, Tamil Nadu, India
| | - Priya Yadav
- Department of Biochemistry & Biotechnology, Annamalai University, Annamalai Nagar 608 002, Tamil Nadu, India
| | - Andaleeb Sajid
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD 20892-4256, USA
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD 20892-4256, USA.
| | - N Rajendra Prasad
- Department of Biochemistry & Biotechnology, Annamalai University, Annamalai Nagar 608 002, Tamil Nadu, India.
| |
Collapse
|
9
|
Scheich S, Chen J, Liu J, Schnütgen F, Enssle JC, Ceribelli M, Thomas CJ, Choi J, Morris V, Hsiao T, Nguyen H, Wang B, Bolomsky A, Phelan JD, Corcoran S, Urlaub H, Young RM, Häupl B, Wright GW, Huang DW, Ji Y, Yu X, Xu W, Yang Y, Zhao H, Muppidi J, Pan KT, Oellerich T, Staudt LM. Targeting N-linked Glycosylation for the Therapy of Aggressive Lymphomas. Cancer Discov 2023; 13:1862-1883. [PMID: 37141112 PMCID: PMC10524254 DOI: 10.1158/2159-8290.cd-22-1401] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/21/2023] [Accepted: 05/02/2023] [Indexed: 05/05/2023]
Abstract
Diffuse large B-cell lymphoma (DLBCL) can be subdivided into the activated B-cell (ABC) and germinal center B cell-like (GCB) subtypes. Self-antigen engagement of B-cell receptors (BCR) in ABC tumors induces their clustering, thereby initiating chronic active signaling and activation of NF-κB and PI3 kinase. Constitutive BCR signaling is essential in some GCB tumors but primarily activates PI3 kinase. We devised genome-wide CRISPR-Cas9 screens to identify regulators of IRF4, a direct transcriptional target of NF-κB and an indicator of proximal BCR signaling in ABC DLBCL. Unexpectedly, inactivation of N-linked protein glycosylation by the oligosaccharyltransferase-B (OST-B) complex reduced IRF4 expression. OST-B inhibition of BCR glycosylation reduced BCR clustering and internalization while promoting its association with CD22, which attenuated PI3 kinase and NF-κB activation. By directly interfering with proximal BCR signaling, OST-B inactivation killed models of ABC and GCB DLBCL, supporting the development of selective OST-B inhibitors for the treatment of these aggressive cancers. SIGNIFICANCE DLBCL depends on constitutive BCR activation and signaling. There are currently no therapeutics that target the BCR directly and attenuate its pathologic signaling. Here, we unraveled a therapeutically exploitable, OST-B-dependent glycosylation pathway that drives BCR organization and proximal BCR signaling. This article is highlighted in the In This Issue feature, p. 1749.
Collapse
Affiliation(s)
- Sebastian Scheich
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jiji Chen
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD, USA
| | - Jiamin Liu
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD, USA
| | - Frank Schnütgen
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Julius C. Enssle
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Michele Ceribelli
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Craig J. Thomas
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Jaewoo Choi
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vivian Morris
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Johns Hopkins University, Department of Biology, Baltimore, MD, 21218, USA
| | - Tony Hsiao
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hang Nguyen
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Boya Wang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arnold Bolomsky
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James D. Phelan
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sean Corcoran
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
- Bioanalytics, Institute of Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Ryan M. Young
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Björn Häupl
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - George W. Wright
- Biometric Research Branch, Division of Cancer Diagnosis and Treatment, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Da Wei Huang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yanlong Ji
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Xin Yu
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Weihong Xu
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yandan Yang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hong Zhao
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jagan Muppidi
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kuan-Ting Pan
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Thomas Oellerich
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Louis M. Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
10
|
Ma H, Chen X, Mo S, Zhang Y, Mao X, Chen J, Liu Y, Tong WM, Lu Z, Yu S, Chen J. Targeting N-glycosylation of 4F2hc mediated by glycosyltransferase B3GNT3 sensitizes ferroptosis of pancreatic ductal adenocarcinoma. Cell Death Differ 2023; 30:1988-2004. [PMID: 37479744 PMCID: PMC10406883 DOI: 10.1038/s41418-023-01188-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 06/09/2023] [Accepted: 06/26/2023] [Indexed: 07/23/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a highly fatal malignancy partially due to the acquired alterations related to aberrant protein glycosylation that pathologically remodel molecular biological processes and protect PDAC cells from death. Ferroptosis driven by lethal lipid peroxidation provides a targetable vulnerability for PDAC. However, the crosstalk between glycosylation and ferroptosis remains unclear. Here, we identified 4F2hc, a subunit of the glutamate-cystine antiporter system Xc-, and its asparagine (N)-glycosylation is involved in PDAC ferroptosis by N- and O-linked glycoproteomics. Knockdown of SLC3A2 (gene name of 4F2hc) or blocking the N-glycosylation of 4F2hc potentiates ferroptosis sensitization of PDAC cells by impairing the activity of system Xc- manifested by a marked decrease in intracellular glutathione. Mechanistically, we found that the glycosyltransferase B3GNT3 catalyzes the glycosylation of 4F2hc, stabilizes the 4F2hc protein, and enhances the interaction between 4F2hc and xCT. Knockout of B3GNT3 or deletion of enzymatically active B3GNT3 sensitizes PDAC cells to ferroptosis. Reconstitution of 4F2hc-deficient cells with wildtype 4F2hc restores ferroptosis resistance while glycosylation-mutated 4F2hc does not. Additionally, upon combination with a ferroptosis inducer, treatment with the classical N-glycosylation inhibitor tunicamycin (TM) markedly triggers the overactivation of lipid peroxidation and enhances the sensitivity of PDAC cells to ferroptosis. Notably, we confirmed that genetic perturbation of SLC3A2 or combination treatment with TM significantly augments ferroptosis-induced inhibition of orthotopic PDAC. Clinically, high expression of 4F2hc and B3GNT3 contributes to the progression and poor survival of PDAC patients. Collectively, our findings reveal a previously unappreciated function of N-glycosylation of 4F2hc in ferroptosis and suggest that dual targeting the vulnerabilities of N-glycosylation and ferroptosis may be an innovative therapeutic strategy for PDAC.
Collapse
Affiliation(s)
- Heng Ma
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Xianlong Chen
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Shengwei Mo
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Yue Zhang
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Xinxin Mao
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Jingci Chen
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Yilin Liu
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Wei-Min Tong
- Department of Pathology, Institute of Basic Medical Sciences, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Zhaohui Lu
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Shuangni Yu
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China.
| | - Jie Chen
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China.
| |
Collapse
|
11
|
Adamczyk-Grochala J, Bloniarz D, Zielinska K, Lewinska A, Wnuk M. DNMT2/TRDMT1 gene knockout compromises doxorubicin-induced unfolded protein response and sensitizes cancer cells to ER stress-induced apoptosis. Apoptosis 2023; 28:166-185. [PMID: 36273376 PMCID: PMC9950192 DOI: 10.1007/s10495-022-01779-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2022] [Indexed: 11/26/2022]
Abstract
The acidic, hypoxic and nutrient-deprived tumor microenvironment may induce endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) may exert an important cytoprotective role by promoting folding of newly synthesized proteins and cancer cell survival. The lack of DNMT2/TRDMT1 methyltransferase-mediated C38 tRNA methylation compromises translational fidelity that may result in the accumulation of misfolded and aggregated proteins leading to proteotoxic stress-related cell death. In the present study, DNMT2/TRDMT1 gene knockout-mediated effects were investigated during doxorubicin (DOX)-induced ER stress and PERK-, IRE1- and ATF6-orchestrated UPR in four genetically different cellular models of cancer (breast and cervical cancer, osteosarcoma and glioblastoma cells). Upon DOX stimulation, DNMT2/TRDMT1 gene knockout impaired PERK activation and modulated NSUN and 5-methylcytosine RNA-based responses and microRNA profiles. The lack of DNMT2/TRDMT1 gene in DOX-treated four cancer cell lines resulted in decreased levels of four microRNAs, namely, miR-23a-3p, miR-93-5p, miR-125a-5p and miR-191-5p involved in the regulation of several pathways such as ubiquitin-mediated proteolysis, amino acid degradation and translational misregulation in cancer. We conclude that DNMT2/TRDMT1 gene knockout, at least in selected cellular cancer models, affects adaptive responses associated with protein homeostasis networks that during prolonged ER stress may result in increased sensitivity to apoptotic cell death.
Collapse
Affiliation(s)
- Jagoda Adamczyk-Grochala
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland
| | - Dominika Bloniarz
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland
| | - Klaudia Zielinska
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland
| | - Anna Lewinska
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland.
| | - Maciej Wnuk
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland.
| |
Collapse
|
12
|
Li J, Li X, Guo Q. Drug Resistance in Cancers: A Free Pass for Bullying. Cells 2022; 11:3383. [PMID: 36359776 PMCID: PMC9654341 DOI: 10.3390/cells11213383] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 08/13/2023] Open
Abstract
The cancer burden continues to grow globally, and drug resistance remains a substantial challenge in cancer therapy. It is well established that cancerous cells with clonal dysplasia generate the same carcinogenic lesions. Tumor cells pass on genetic templates to subsequent generations in evolutionary terms and exhibit drug resistance simply by accumulating genetic alterations. However, recent evidence has implied that tumor cells accumulate genetic alterations by progressively adapting. As a result, intratumor heterogeneity (ITH) is generated due to genetically distinct subclonal populations of cells coexisting. The genetic adaptive mechanisms of action of ITH include activating "cellular plasticity", through which tumor cells create a tumor-supportive microenvironment in which they can proliferate and cause increased damage. These highly plastic cells are located in the tumor microenvironment (TME) and undergo extreme changes to resist therapeutic drugs. Accordingly, the underlying mechanisms involved in drug resistance have been re-evaluated. Herein, we will reveal new themes emerging from initial studies of drug resistance and outline the findings regarding drug resistance from the perspective of the TME; the themes include exosomes, metabolic reprogramming, protein glycosylation and autophagy, and the relates studies aim to provide new targets and strategies for reversing drug resistance in cancers.
Collapse
Affiliation(s)
| | | | - Qie Guo
- The Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
13
|
Mitachi K, Mingle D, Effah W, Sánchez‐Ruiz A, Hevener KE, Narayanan R, Clemons WM, Sarabia F, Kurosu M. Concise Synthesis of Tunicamycin V and Discovery of a Cytostatic DPAGT1 Inhibitor. Angew Chem Int Ed Engl 2022; 61:e202203225. [PMID: 35594368 PMCID: PMC9329268 DOI: 10.1002/anie.202203225] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Indexed: 11/11/2022]
Abstract
A short total synthesis of tunicamycin V (1), a non-selective phosphotransferase inhibitor, is achieved via a Büchner-Curtius-Schlotterbeck type reaction. Tunicamycin V can be synthesized in 15 chemical steps from D-galactal with 21 % overall yield. The established synthetic scheme is operationally very simple and flexible to introduce building blocks of interest. The inhibitory activity of one of the designed analogues 28 against human dolichyl-phosphate N-acetylglucosaminephosphotransferase 1 (DPAGT1) is 12.5 times greater than 1. While tunicamycins are cytotoxic molecules with a low selectivity, the novel analogue 28 displays selective cytostatic activity against breast cancer cell lines including a triple-negative breast cancer.
Collapse
Affiliation(s)
- Katsuhiko Mitachi
- Department of Pharmaceutical Sciences College of Pharmacy University of Tennessee Health Science Center 881 Madison Avenue Memphis TN 38163 USA
| | - David Mingle
- Department of Pharmaceutical Sciences College of Pharmacy University of Tennessee Health Science Center 881 Madison Avenue Memphis TN 38163 USA
| | - Wendy Effah
- Department of Medicine University of Tennessee Health Science Center 19 S. Manassas, Room 120 Memphis TN 38103 USA
| | - Antonio Sánchez‐Ruiz
- Faculty of Pharmacy Campus de Albacete Universidad de Castilla-La Mancha Avda. Dr. José María Sánchez Ibáñez S/N 02008 Albacete Spain
| | - Kirk E. Hevener
- Department of Pharmaceutical Sciences College of Pharmacy University of Tennessee Health Science Center 881 Madison Avenue Memphis TN 38163 USA
| | - Ramesh Narayanan
- Department of Medicine University of Tennessee Health Science Center 19 S. Manassas, Room 120 Memphis TN 38103 USA
| | - William M. Clemons
- Division of Chemistry and Chemical Engineering California Institute of Technology 1200 E. California Blvd. Pasadena CA 91125 USA
| | - Francisco Sarabia
- Department of Organic Chemistry Faculty of Sciences Universidad de Málaga, Campus de Teatinos 29071 Málaga Spain
| | - Michio Kurosu
- Department of Pharmaceutical Sciences College of Pharmacy University of Tennessee Health Science Center 881 Madison Avenue Memphis TN 38163 USA
| |
Collapse
|
14
|
Mitachi K, Mingle D, Effah W, Sánchez-Ruiz A, Hevener KE, Narayanan R, Clemons WM, Sarabia F, Kurosu M. Concise Synthesis of Tunicamycin V and Discovery of a Cytostatic DPAGT1 Inhibitor. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202203225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Katsuhiko Mitachi
- The University of Tennessee Health Science Center College of Pharmacy Pharmacy 881 Madison AvenueROOM 557 38163 MEMPHS UNITED STATES
| | - David Mingle
- The University of Tennessee Health Science Center College of Pharmacy Pharmacy 881 MADISON AVE 38163 MEMPHS UNITED STATES
| | - Wendy Effah
- University of Tennessee College of Medicine: The University of Tennessee Health Science Center College of Medicine Medicine UNITED STATES
| | | | - Kirk E. Hevener
- UTHSC College of Pharmacy Memphis: The University of Tennessee Health Science Center College of Pharmacy Pharmacy UNITED STATES
| | - Ramesh Narayanan
- University of Tennessee College of Medicine: The University of Tennessee Health Science Center College of Medicine Medicine 19, S. Manassas 38013 Memphis UNITED STATES
| | - William M. Clemons
- Caltech: California Institute of Technology Chemistry and Chemical Engineering UNITED STATES
| | - Francisco Sarabia
- University of Malaga: Universidad de Malaga Organic Chemistry UNITED STATES
| | - Michio Kurosu
- UTHSC College of Pharmacy Memphis: The University of Tennessee Health Science Center College of Pharmacy Department of Pharmaceutical Sciences, College of Pharmacy 881 MADISON AVEROOM 557 38163 Memphis UNITED STATES
| |
Collapse
|
15
|
Ibezim A, Osigwe S, Uzor P, Engel N, Ramanathan K, Nwodo N. Computational studies reveal potential dolichyl-phosphate N-acetylglucosaminephosphotransferase inhibitors amidst existing drugs. J Biomol Struct Dyn 2022:1-8. [PMID: 35467485 DOI: 10.1080/07391102.2022.2064916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Dolichyl-phosphate N-acetylglucosaminephosphotransferase (dpagt1) inhibition is reported to kill tumor cells whose growth progression requires increased branching of N-linked glycans. Available dpagt1 inhibitors are grossly limited and are faced with problems of heamolytic effect and aqueous solubility thereby necessitating the search for new, safe and effective dpagt1 inhibitors. We employed computational methods to screen a dataset of ∼1300 FDA approved drugs in order to obtain theoretical dpagt1 inhibitors which could be repurposed as chemotherapeutic drugs. Top six better performing drugs, binding affinity for dpagt1 at the range of -17.63 to -20.40 kcal/mol, than the reference ligand (tunicamycin; -14.86 kcal/mol) were obtained at the end of structure-based-pharmacophore- and virtual-screening and 'induced fit' docking calculations. Analysis of their binding poses identified essential pharmacophores involved in target-ligand complexation that could be targeted in chemical modification to develop more effective and safe dpagt1 inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Akachukwu Ibezim
- Department of Pharmaceutical and Medicinal, University of Nigeria, Nsukka, Nigeria.,Department of Biotechnology, Vellore Institute of Technology, Vellore, Tamilnadu, India
| | - Sochi Osigwe
- Department of Pharmaceutical and Medicinal, University of Nigeria, Nsukka, Nigeria
| | - Philip Uzor
- Department of Pharmaceutical and Medicinal, University of Nigeria, Nsukka, Nigeria
| | - Nadja Engel
- Department of Oral, Maxillofacial and Plastic Surgery, Rostock University Medical Center, Rostock, Germany
| | | | - Ngozi Nwodo
- Department of Pharmaceutical and Medicinal, University of Nigeria, Nsukka, Nigeria
| |
Collapse
|
16
|
Chen S, Wang Y, Liu W, Liang Y, Wang Y, Wu Z, Xu L, Liang X, Ma C, Gao L. N-Glycosylation at Asn291 Stabilizes TIM-4 and Promotes the Metastasis of NSCLC. Front Oncol 2022; 12:730530. [PMID: 35433445 PMCID: PMC9008408 DOI: 10.3389/fonc.2022.730530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 03/07/2022] [Indexed: 01/30/2023] Open
Abstract
T-cell immunoglobulin domain and mucin domain 4 (TIM-4) is a transmembrane protein that promotes epithelial-mesenchymal transition (EMT), migration and invasion of non-small cell lung cancer (NSCLC) cells. Most transmembrane proteins are modified by N-glycosylation and the importance of protein N-glycosylation in cancer cell metastasis has been well appreciated. However, whether TIM-4 is modified by N-glycosylation and the role of TIM-4 N-glycosylation in NSCLC remains largely unknown. In the current study, we reported that TIM-4 was extensively N-glycosylated at Asn291. After the removal of N-glycosylation, the stability of TIM-4 protein was decreased and TIM-4 was more susceptible to degradation by ER-localized ubiquitin ligase-mediated ERAD. Thus, the expression of TIM-4 on the cell surface was decreased, which suppressed TIM-4-mediated metastasis in NSCLC. In summary, the present study identifies TIM-4 N-glycosylation and its role in NSCLS migration, which would provide a valuable biomarker for developing drugs targeting N-glycosylation at Asn291 on TIM-4.
Collapse
Affiliation(s)
- Siyuan Chen
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuzhen Wang
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wen Liu
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yan Liang
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yingchun Wang
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhuanchang Wu
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Liyun Xu
- Cell and Molecular Biology Laboratory, Zhoushan Hospital, Zhoushan, China
| | - Xiaohong Liang
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunhong Ma
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lifen Gao
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
17
|
Zhen Z, Shen Z, Sun P. Downregulation of Low-density lipoprotein receptor-related protein 1B (LRP1B) inhibits the progression of hepatocellular carcinoma cells by activating the endoplasmic reticulum stress signaling pathway. Bioengineered 2022; 13:9467-9481. [PMID: 35389768 PMCID: PMC9161869 DOI: 10.1080/21655979.2022.2060778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has a high recurrence rate and mortality rate even after surgery. Low-density lipoprotein receptor-related protein 1B (LRP1B) has proven to be involved in tumor development and progression of multiple malignancies. However, the function of LRP1B in HCC progression has not been fully elucidated. Thus, we conducted this study to explore the relationship between LRP1B and HCC. Bioinformatic analyses implied that LRP1B was highly expressed in HCC tissues. High LRP1B expression was shown to be related to poor outcomes and the determination of HCC patients’ tumor stage. LRP1B deletion impeded the proliferation, migration, and invasion of HCC cells. Further investigation demonstrated that silencing LRP1B expression enhanced the sensitivity of HCC cells to doxorubicin. LRP1B deletion inhibited HCC progression by regulating the PERK-ATF4-CHOP signaling pathway. Additionally, we probed the genomic alterations of LRP1B in HCC and the impact on the prognosis of patients. Collectively, our results suggest that LRP1B plays an essential role in the promotion of HCC progression by regulating the PERK-ATF4-CHOP signaling pathway, which is a potential prognostic biomarker and a promising therapeutic target of HCC.
Collapse
Affiliation(s)
- Zili Zhen
- Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai, P. R. China.,Department of Surgery, Shanghai Medical College, Fudan University, Shanghai, P. R. China.,Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, P. R. China
| | - Zhemin Shen
- Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai, P. R. China
| | - Peilong Sun
- Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai, P. R. China.,Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, P. R. China
| |
Collapse
|
18
|
Zeng W, Zheng S, Mao Y, Wang S, Zhong Y, Cao W, Su T, Gong M, Cheng J, Zhang Y, Yang H. Elevated N-Glycosylation Contributes to the Cisplatin Resistance of Non-Small Cell Lung Cancer Cells Revealed by Membrane Proteomic and Glycoproteomic Analysis. Front Pharmacol 2022; 12:805499. [PMID: 35002739 PMCID: PMC8728018 DOI: 10.3389/fphar.2021.805499] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/06/2021] [Indexed: 02/05/2023] Open
Abstract
Chemoresistance is the major restriction on the clinical use of cisplatin. Aberrant changes in protein glycosylation are closely associated with drug resistance. Comprehensive study on the role of protein glycosylation in the development of cisplatin resistance would contribute to precise elucidation of the complicated mechanism of resistance. However, comprehensive characterization of glycosylated proteins remains a big challenge. In this work, we integrated proteomic and N-glycoproteomic workflow to comprehensively characterize the cisplatin resistance-related membrane proteins. Using this method, we found that proteins implicated in cell adhesion, migration, response to drug, and signal transduction were significantly altered in both protein abundance and glycosylation level during the development of cisplatin resistance in the non-small cell lung cancer cell line. Accordingly, the ability of cell migration and invasion was markedly increased in cisplatin-resistant cells, hence intensifying their malignancy. In contrast, the intracellular cisplatin accumulation was significantly reduced in the resistant cells concomitant with the down-regulation of drug uptake channel protein, LRRC8A, and over-expression of drug efflux pump proteins, MRP1 and MRP4. Moreover, the global glycosylation was elevated in the cisplatin-resistant cells. Consequently, inhibition of N-glycosylation reduced cell resistance to cisplatin, whereas promoting the high-mannose or sialylated type of glycosylation enhanced the resistance, suggesting that critical glycosylation type contributes to cisplatin resistance. These results demonstrate the high efficiency of the integrated proteomic and N-glycoproteomic workflow in discovering drug resistance-related targets, and provide new insights into the mechanism of cisplatin resistance.
Collapse
Affiliation(s)
- Wenjuan Zeng
- NHC Key Laboratory of Transplant Engineering and Immunology, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Shanshan Zheng
- NHC Key Laboratory of Transplant Engineering and Immunology, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yonghong Mao
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Shisheng Wang
- NHC Key Laboratory of Transplant Engineering and Immunology, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Zhong
- NHC Key Laboratory of Transplant Engineering and Immunology, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Cao
- NHC Key Laboratory of Transplant Engineering and Immunology, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Su
- NHC Key Laboratory of Transplant Engineering and Immunology, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Meng Gong
- NHC Key Laboratory of Transplant Engineering and Immunology, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jingqiu Cheng
- NHC Key Laboratory of Transplant Engineering and Immunology, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Zhang
- NHC Key Laboratory of Transplant Engineering and Immunology, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Yang
- NHC Key Laboratory of Transplant Engineering and Immunology, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Kukal S, Guin D, Rawat C, Bora S, Mishra MK, Sharma P, Paul PR, Kanojia N, Grewal GK, Kukreti S, Saso L, Kukreti R. Multidrug efflux transporter ABCG2: expression and regulation. Cell Mol Life Sci 2021; 78:6887-6939. [PMID: 34586444 PMCID: PMC11072723 DOI: 10.1007/s00018-021-03901-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/24/2021] [Accepted: 07/15/2021] [Indexed: 12/15/2022]
Abstract
The adenosine triphosphate (ATP)-binding cassette efflux transporter G2 (ABCG2) was originally discovered in a multidrug-resistant breast cancer cell line. Studies in the past have expanded the understanding of its role in physiology, disease pathology and drug resistance. With a widely distributed expression across different cell types, ABCG2 plays a central role in ATP-dependent efflux of a vast range of endogenous and exogenous molecules, thereby maintaining cellular homeostasis and providing tissue protection against xenobiotic insults. However, ABCG2 expression is subjected to alterations under various pathophysiological conditions such as inflammation, infection, tissue injury, disease pathology and in response to xenobiotics and endobiotics. These changes may interfere with the bioavailability of therapeutic substrate drugs conferring drug resistance and in certain cases worsen the pathophysiological state aggravating its severity. Considering the crucial role of ABCG2 in normal physiology, therapeutic interventions directly targeting the transporter function may produce serious side effects. Therefore, modulation of transporter regulation instead of inhibiting the transporter itself will allow subtle changes in ABCG2 activity. This requires a thorough comprehension of diverse factors and complex signaling pathways (Kinases, Wnt/β-catenin, Sonic hedgehog) operating at multiple regulatory levels dictating ABCG2 expression and activity. This review features a background on the physiological role of transporter, factors that modulate ABCG2 levels and highlights various signaling pathways, molecular mechanisms and genetic polymorphisms in ABCG2 regulation. This understanding will aid in identifying potential molecular targets for therapeutic interventions to overcome ABCG2-mediated multidrug resistance (MDR) and to manage ABCG2-related pathophysiology.
Collapse
Affiliation(s)
- Samiksha Kukal
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi, 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Debleena Guin
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi, 110007, India
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Main Bawana Road, Delhi, 110042, India
| | - Chitra Rawat
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi, 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shivangi Bora
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi, 110007, India
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Main Bawana Road, Delhi, 110042, India
| | - Manish Kumar Mishra
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi, 110007, India
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Main Bawana Road, Delhi, 110042, India
| | - Priya Sharma
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi, 110007, India
| | - Priyanka Rani Paul
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi, 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Neha Kanojia
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi, 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Gurpreet Kaur Grewal
- Department of Biotechnology, Kanya Maha Vidyalaya, Jalandhar, Punjab, 144004, India
| | - Shrikant Kukreti
- Nucleic Acids Research Lab, Department of Chemistry, University of Delhi (North Campus), Delhi, 110007, India
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P. le Aldo Moro 5, 00185, Rome, Italy
| | - Ritushree Kukreti
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi, 110007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
20
|
Huang B, Yan X, Li Y. Cancer Stem Cell for Tumor Therapy. Cancers (Basel) 2021; 13:cancers13194814. [PMID: 34638298 PMCID: PMC8508418 DOI: 10.3390/cancers13194814] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/13/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Although many methods have been applied in clinical treatment for tumors, they still always show a poor prognosis. Molecule targeted therapy has revolutionized tumor therapy, and a proper target must be found urgently. With a crucial role in tumor development, metastasis and recurrence, cancer stem cells have been found to be a feasible and potential target for tumor therapy. We list the unique biological characteristics of cancer stem cells and summarize the recent strategies to target cancer stem cells for tumor therapy, through which we hope to provide a comprehensive understanding of cancer stem cells and find a better combinational strategy to target cancer stem cells for tumor therapy. Abstract Tumors pose a significant threat to human health. Although many methods, such as operations, chemotherapy and radiotherapy, have been proposed to eliminate tumor cells, the results are unsatisfactory. Targeting therapy has shown potential due to its specificity and efficiency. Meanwhile, it has been revealed that cancer stem cells (CSCs) play a crucial role in the genesis, development, metastasis and recurrence of tumors. Thus, it is feasible to inhibit tumors and improve prognosis via targeting CSCs. In this review, we provide a comprehensive understanding of the biological characteristics of CSCs, including mitotic pattern, metabolic phenotype, therapeutic resistance and related mechanisms. Finally, we summarize CSCs targeted strategies, including targeting CSCs surface markers, targeting CSCs related signal pathways, targeting CSC niches, targeting CSC metabolic pathways, inducing differentiation therapy and immunotherapy (tumor vaccine, CAR-T, oncolytic virus, targeting CSCs–immune cell crosstalk and immunity checkpoint inhibitor). We highlight the potential of immunity therapy and its combinational anti-CSC therapies, which are composed of different drugs working in different mechanisms.
Collapse
Affiliation(s)
- Binjie Huang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou 730030, China; (B.H.); (X.Y.)
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Xin Yan
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou 730030, China; (B.H.); (X.Y.)
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Yumin Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou 730030, China; (B.H.); (X.Y.)
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou 730030, China
- Correspondence: ; Tel.: +86-138-9361-5421
| |
Collapse
|
21
|
Liu Z, Yang S, Chen X, Dong S, Zhou S, Xu S. LncRNA LINC00467 acted as an oncogene in esophageal squamous cell carcinoma by accelerating cell proliferation and preventing cell apoptosis via the miR-485-5p/DPAGT1 axis. J Gastroenterol Hepatol 2021; 36:721-730. [PMID: 32720371 DOI: 10.1111/jgh.15201] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/15/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIM Esophageal carcinoma has been regarded as one of the top 10 common malignancies globally. Esophageal squamous cell carcinoma (ESCC) is an important subtype of esophageal carcinoma with approximately 20% survival rate. Long noncoding RNAs were documented to regulate the occurrence or progression of several tumors. However, neither the biological role nor the molecular mechanism of LINC00467 has been explored. This research is aimed to investigating the regulatory mechanism of LINC00467 in ESCC. METHODS In this study, a series of experiments including reverse transcription-quantitative polymerase chain reaction, Cell Counting Kit-8, luciferase reporter, western blot, and RNA immunoprecipitation were designed and conducted to explore the potential function and mechanism of LINC00467 in ESCC. RESULTS According to experimental results, we found out upregulated LINC00467 improved cell proliferation, but hindered cell apoptosis. In mechanism, miR-485-5p was predicted, screened out, and validated to combine with LINC00467, which displayed lower expression in ESCC. Additionally, miR-485-5p negatively regulated and directly targeted DPAGT1. Rescue assays suggested that DPAGT1 amplification was able to recover the influence of LINC00467 deficiency on cell proliferation and apoptosis. Furthermore, knockdown of LINC00467 suppressed tumor growth in vivo. CONCLUSION We proved that LINC00467 acted as an oncogene in ESCC by accelerating cell proliferation and preventing cell apoptosis via miR-485-5p/DPAGT1 axis. This may provide a potential diagnostic marker for ESCC treatment.
Collapse
Affiliation(s)
- Zhenghua Liu
- Department of Thoracic Surgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shize Yang
- Department of Thoracic Surgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xitao Chen
- Department of Thoracic Surgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Siyuan Dong
- Department of Thoracic Surgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Siyu Zhou
- Department of Thoracic Surgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shun Xu
- Department of Thoracic Surgery, First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
22
|
Bartos Z, Homolya L. Identification of Specific Trafficking Defects of Naturally Occurring Variants of the Human ABCG2 Transporter. Front Cell Dev Biol 2021; 9:615729. [PMID: 33634118 PMCID: PMC7900420 DOI: 10.3389/fcell.2021.615729] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/05/2021] [Indexed: 01/04/2023] Open
Abstract
Proper targeting of the urate and xenobiotic transporter ATP-binding transporter subfamily G member 2 (ABCG2) to the plasma membrane (PM) is essential for its normal function. The naturally occurring Q141K and M71V polymorphisms in ABCG2, associated with gout and hyperuricemia, affect the cellular routing of the transporter, rather than its transport function. The cellular localization of ABCG2 variants was formerly studied by immunolabeling, which provides information only on the steady-state distribution of the protein, leaving the dynamics of its cellular routing unexplored. In the present study, we assessed in detail the trafficking of the wild-type, M71V-, and Q141K-ABCG2 variants from the endoplasmic reticulum (ER) to the cell surface using a dynamic approach, the so-called Retention Using Selective Hooks (RUSH) system. This method also allowed us to study the kinetics of glycosylation of these variants. We found that the fraction of Q141K- and M71V-ABCG2 that passes the ER quality control system is only partially targeted to the PM; a subfraction is immobile and retained in the ER. Surprisingly, the transit of these variants through the Golgi apparatus (either the appearance or the exit) was unaffected; however, their PM delivery beyond the Golgi was delayed. In addition to identifying the specific defects in the trafficking of these ABCG2 variants, our study provides a novel experimental tool for studying the effect of drugs that potentially promote the cell surface delivery of mutant or polymorphic ABCG2 variants with impaired trafficking.
Collapse
Affiliation(s)
- Zsuzsa Bartos
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Centre of Excellence, Budapest, Hungary
| | - László Homolya
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Centre of Excellence, Budapest, Hungary
| |
Collapse
|
23
|
Choi YJ, Lee JE, Ji HD, Lee BR, Lee SB, Kim KS, Lee IK, Chin J, Cho SJ, Lee J, Lee SW, Ha JH, Jeon YH. Tunicamycin as a Novel Redifferentiation Agent in Radioiodine Therapy for Anaplastic Thyroid Cancer. Int J Mol Sci 2021; 22:ijms22031077. [PMID: 33499100 PMCID: PMC7865976 DOI: 10.3390/ijms22031077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 11/16/2022] Open
Abstract
The silencing of thyroid-related genes presents difficulties in radioiodine therapy for anaplastic thyroid cancers (ATCs). Tunicamycin (TM), an N-linked glycosylation inhibitor, is an anticancer drug. Herein, we investigated TM-induced restoration of responsiveness to radioiodine therapy in radioiodine refractory ATCs. 125I uptake increased in TM-treated ATC cell lines, including BHT101 and CAL62, which was inhibited by KClO4, a sodium-iodide symporter (NIS) inhibitor. TM upregulated the mRNA expression of iodide-handling genes and the protein expression of NIS. TM blocked pERK1/2 phosphorylation in both cell lines, but AKT (protein kinase B) phosphorylation was only observed in CAL62 cells. The downregulation of glucose transporter 1 protein was confirmed in TM-treated cells, with a significant reduction in 18F-fluorodeoxyglucose (FDG) uptake. A significant reduction in colony-forming ability and marked tumor growth inhibition were observed in the combination group. TM was revealed to possess a novel function as a redifferentiation inducer in ATC as it induces the restoration of iodide-handling gene expression and radioiodine avidity, thereby facilitating effective radioiodine therapy.
Collapse
Affiliation(s)
- Yoon Ju Choi
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41566, Korea; (Y.J.C.); (H.D.J.); (J.L.)
- Department of pharmacology, School of Medicine, Kyungpook National University, Daegu 41405, Korea
| | - Jae-Eon Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41404, Korea; (J.-E.L.); (B.-R.L.); (K.S.K.)
| | - Hyun Dong Ji
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41566, Korea; (Y.J.C.); (H.D.J.); (J.L.)
- Department of pharmacology, School of Medicine, Kyungpook National University, Daegu 41405, Korea
| | - Bo-Ra Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41404, Korea; (J.-E.L.); (B.-R.L.); (K.S.K.)
| | - Sang Bong Lee
- Vaccine Commerialization Center, Gyeongbuk Institute for Bioindustry, 88, Saneodanjigil, Pungsan-eup, Andong-si, Gyeongbuk 36618, Korea;
| | - Kil Soo Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41404, Korea; (J.-E.L.); (B.-R.L.); (K.S.K.)
- College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41404, Korea
| | - Jungwook Chin
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41404, Korea; (J.C.); (S.J.C.)
| | - Sung Jin Cho
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41404, Korea; (J.C.); (S.J.C.)
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41566, Korea; (Y.J.C.); (H.D.J.); (J.L.)
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41566, Korea; (Y.J.C.); (H.D.J.); (J.L.)
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea
- Correspondence: (S.-W.L.); (J.-H.H.); (Y.H.J.); Tel.: +82-53-200-2851 (S.-W.L.); +82-53-950-4232 (J.-H.H.); +82-10-2455-6046 or +82-53-200-3149 (Y.H.J.)
| | - Jeoung-Hee Ha
- Department of pharmacology, School of Medicine, Kyungpook National University, Daegu 41405, Korea
- Correspondence: (S.-W.L.); (J.-H.H.); (Y.H.J.); Tel.: +82-53-200-2851 (S.-W.L.); +82-53-950-4232 (J.-H.H.); +82-10-2455-6046 or +82-53-200-3149 (Y.H.J.)
| | - Yong Hyun Jeon
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41404, Korea; (J.-E.L.); (B.-R.L.); (K.S.K.)
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea
- Correspondence: (S.-W.L.); (J.-H.H.); (Y.H.J.); Tel.: +82-53-200-2851 (S.-W.L.); +82-53-950-4232 (J.-H.H.); +82-10-2455-6046 or +82-53-200-3149 (Y.H.J.)
| |
Collapse
|
24
|
Schmidtova S, Dorssers LCJ, Kalavska K, Gillis AJM, Oosterhuis JW, Stoop H, Miklikova S, Kozovska Z, Burikova M, Gercakova K, Durinikova E, Chovanec M, Mego M, Kucerova L, Looijenga LHJ. Napabucasin overcomes cisplatin resistance in ovarian germ cell tumor-derived cell line by inhibiting cancer stemness. Cancer Cell Int 2020; 20:364. [PMID: 32774158 PMCID: PMC7397611 DOI: 10.1186/s12935-020-01458-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/25/2020] [Indexed: 02/08/2023] Open
Abstract
Background Cisplatin resistance of ovarian yolk sac tumors (oYST) is a clinical challenge due to dismal patient prognosis, even though the disease is extremely rare. We investigated potential association between cisplatin resistance and cancer stem cell (CSC) markers in chemoresistant oYST cells and targeting strategies to overcome resistance in oYST. Methods Chemoresistant cells were derived from chemosensitive human oYST cells by cultivation in cisplatin in vitro. Derivative cells were characterized by chemoresistance, functional assays, flow cytometry, gene expression and protein arrays focused on CSC markers. RNAseq, methylation and microRNA profiling were performed. Quail chorioallantoic membranes (CAM) with implanted oYST cells were used to analyze the micro-tumor extent and interconnection with the CAM. Tumorigenicity in vivo was determined on immunodeficient mouse model. Chemoresistant cells were treated by inhibitors intefering with the CSC properties to examine the chemosensitization to cisplatin. Results Long-term cisplatin exposure resulted in seven-fold higher IC50 value in resistant cells, cross-resistance to oxaliplatin and carboplatin, and increased migratory capacity, invasiveness and tumorigenicity, associated with hypomethylation of differentially methylated genes/promotors. Resistant cells exhibited increased expression of prominin-1 (CD133), ATP binding cassette subfamily G member 2 (ABCG2), aldehyde dehydrogenase 3 isoform A1 (ALDH3A1), correlating with reduced gene and promoter methylation, as well as increased expression of ALDH1A3 and higher overall ALDH enzymatic activity, rendering them cross-resistant to DEAB, disulfiram and napabucasin. Salinomycin and tunicamycin were significantly more toxic to resistant cells. Pretreatment with napabucasin resensitized the cells to cisplatin and reduced their tumorigenicity in vivo. Conclusions The novel chemoresistant cells represent unique model of refractory oYST. CSC markers are associated with cisplatin resistance being possible targets in chemorefractory oYST.
Collapse
Affiliation(s)
- Silvia Schmidtova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia.,Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia
| | - Lambert C J Dorssers
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Katarina Kalavska
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia.,Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia.,2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia
| | - Ad J M Gillis
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.,Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - J Wolter Oosterhuis
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Hans Stoop
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Svetlana Miklikova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Zuzana Kozovska
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Monika Burikova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Katarina Gercakova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Erika Durinikova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Michal Chovanec
- Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia.,2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia
| | - Michal Mego
- Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia.,2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia
| | - Lucia Kucerova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Leendert H J Looijenga
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.,Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| |
Collapse
|
25
|
Liu H, Xie S, Fang F, Kalvakolanu DV, Xiao W. SHQ1 is an ER stress response gene that facilitates chemotherapeutics-induced apoptosis via sensitizing ER-stress response. Cell Death Dis 2020; 11:445. [PMID: 32522979 PMCID: PMC7286909 DOI: 10.1038/s41419-020-2656-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022]
Abstract
SHQ1 was reported to control the biogenesis and assembly of H/ACA ribonucleoprotein particles (RNPs). It was independently isolated as a growth suppressor, GRIM1, in a genetic screen. Recent studies have indicated that SHQ1 inhibits prostate cancer growth and metastasis. SHQ1 facilitates MYC RNA splicing to promote T-acute lymphoblastic leukemia (T-ALL) development. Thus, the mechanisms of SHQ1 in cancers remain largely unknown. We report here that SHQ1 promotes tumor apoptosis and chemo-sensitivity in hepatocellular carcinoma (HCC) cells. In HCC tissues from patients, expression of SHQ1 was significantly decreased in the tumor compared to adjacent tissues. Experiments with HCC xenograft models revealed that restoring SHQ1 levels enhanced the anti-tumor activity of the endoplasmic reticulum (ER) stress inducer tunicamycin (TM) and common chemotherapy drug paclitaxel (PTX). Mechanistically, SHQ1 is an ER-stress response gene which is regulated by p50ATF6 and XBP1s through an ER stress response like element located on the SHQ1 promoter. SHQ1 interacts with the ER chaperone GRP78 to release ER sensors PERK/IRE1α/ATF6 from GRP78/ER-sensor complexes, leading to hyper-activation of unfolded protein response (UPR). In the persistent ER stress conditions of a HepG2 xenograft tumor model, SHQ1-mediated hyper-activation of ER-sensor signaling induces apoptosis. Our study thus demonstrates a SHQ1-mediated ER-stress response feedback loop that promotes tumor sensitivity to chemotherapeutics.
Collapse
Affiliation(s)
- Huimin Liu
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027, Anhui, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China.,Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, 230027, Anhui, Hefei, China
| | - Siqi Xie
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027, Anhui, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China.,Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, 230027, Anhui, Hefei, China
| | - Fang Fang
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027, Anhui, Hefei, China. .,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China. .,Institute of Immunology, University of Science and Technology of China, Hefei, China. .,Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, 230027, Anhui, Hefei, China.
| | - Dhananjaya V Kalvakolanu
- Department of Microbiology and Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Weihua Xiao
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027, Anhui, Hefei, China. .,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China. .,Institute of Immunology, University of Science and Technology of China, Hefei, China. .,Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, 230027, Anhui, Hefei, China.
| |
Collapse
|
26
|
Ju M, Qi A, Bi J, Zhao L, Jiang L, Zhang Q, Wei Q, Guan Q, Li X, Wang L, Wei M, Zhao L. A five-mRNA signature associated with post-translational modifications can better predict recurrence and survival in cervical cancer. J Cell Mol Med 2020; 24:6283-6297. [PMID: 32306508 PMCID: PMC7294153 DOI: 10.1111/jcmm.15270] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/04/2020] [Accepted: 03/27/2020] [Indexed: 12/24/2022] Open
Abstract
High mortality of patients with cervical cancer (CC) stresses the imperative of prognostic biomarkers for CC patients. Additionally, the vital status of post‐translational modifications (PTMs) in the progression of cancers has been reported by numerous researches. Therefore, the purpose of this research was to dig a prognostic signature correlated with PTMs for CC. We built a five‐mRNA (GALNTL6, ARSE, DPAGT1, GANAB and FURIN) prognostic signature associated with PTMs to predict both disease‐free survival (DFS) (hazard ratio [HR] = 3.967, 95% CI = 1.985‐7.927; P < .001) and overall survival (HR = 2.092, 95% CI = 1.138‐3.847; P = .018) for CC using data from The Cancer Genome Atlas database. Then, the robustness of the signature was validated using GSE44001 and the Human Protein Atlas (HPA) database. CIBERSORT algorithm analysis displayed that activated CD4 memory T cell was also an independent indicator for DFS (HR = 0.426, 95% CI = 0.186‐0.978; P = .044) which could add additional prognostic value to the signature. Collectively, the PTM‐related signature and activated CD4 memory T cell can provide new avenues for the prognostic predication of CC. These findings give further insights into effective treatment strategies for CC, providing opportunities for further experimental and clinical validations.
Collapse
Affiliation(s)
- Mingyi Ju
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang City, Liaoning, China
| | - Aoshuang Qi
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang City, Liaoning, China
| | - Jia Bi
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang City, Liaoning, China
| | - Lan Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang City, Liaoning, China
| | - Longyang Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang City, Liaoning, China
| | - Qiang Zhang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang City, Liaoning, China
| | - Qian Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang City, Liaoning, China
| | - Qiutong Guan
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang City, Liaoning, China
| | - Xueping Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang City, Liaoning, China
| | - Lin Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang City, Liaoning, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang City, Liaoning, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang City, Liaoning, China
| |
Collapse
|
27
|
Takahashi S. Mutations of FLT3 receptor affect its surface glycosylation, intracellular localization, and downstream signaling. Leuk Res Rep 2019; 13:100187. [PMID: 31853441 PMCID: PMC6911968 DOI: 10.1016/j.lrr.2019.100187] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/29/2019] [Accepted: 11/23/2019] [Indexed: 11/29/2022] Open
Abstract
This review describes the effects of FLT3 mutations that alter its intracellular localization and modify its glycosylation, leading to differences in downstream signaling pathways. The most common type of FLT3 mutation, internal tandem duplication (FLT3-ITD), leads to localization in the endoplasmic reticulum and constitutive strong activation of STAT5. In contrast, the ligand-activated FLT3-wild type is mainly expressed on the cell surface and activates MAP kinases. Based on these backgrounds, several reports have demonstrated that glycosylation inhibitors are effective for inhibition of FLT3-ITD expression and intracellular localization. The general subcellular localization regulatory mechanisms for receptor tyrosine kinases are also discussed.
Collapse
Affiliation(s)
- Shinichiro Takahashi
- Division of Laboratory Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Japan
| |
Collapse
|
28
|
Li R, Xu T, Wang H, Wu N, Liu F, Jia X, Mi J, Lv J, Gao H. Dysregulation of the miR-325-3p/DPAGT1 axis supports HBV-positive HCC chemoresistance. Biochem Biophys Res Commun 2019; 519:358-365. [PMID: 31519321 DOI: 10.1016/j.bbrc.2019.08.116] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 08/22/2019] [Indexed: 01/30/2023]
Abstract
BACKGROUND Chemotherapeutic resistance in hepatitis B virus (HBV)-positive hepatocellular carcinoma (HCC) patients is an unfortunate side effect of standard chemotherapy. This situation necessitates a better understanding of the molecular pathways underlying HBV + HCC chemoresistance in order to aid the development of novel chemotherapeutic targets. METHODS We generated two doxorubicin (DOX)-resistant HBV + HCC sublines HepG2.2.15 and Huh7-1.3. qRT-PCR was used to evaluate dysregulation in hexosamine pathway genes in chemosensitive and chemoresistant HBV + HCC cell lines in vitro. Western blots, luciferase reporter assays, and in vivo xenograft tumor studies were conducted to reveal the role of the miRNA-325-3p/DPAGT1 axis in HBV + HCC chemoresistance. RESULTS The hexosamine pathway gene dolichyl-phosphate N-acetylglucosamine phosphotransferase 1 (DPAGT1) was found to be upregulated in both DOX-resistant cell lines. Enhancing DPAGT1 activity significantly improved the survival of DOX-resistant cells. Silencing or pharmacological inhibition of DPAGT1 inhibited xenograft tumor growth under DOX-treated conditions. DPAGT1 upregulation was associated with higher levels of stemness-related markers and ATP-binding cassette (ABC) drug efflux transporters in DOX-resistant cell lines. miR-325-3p was found to negatively modulate DPAGT1 expression and phenocopied the effects of DPAGT1 silencing in vitro and in vivo. In HBV + HCC patients treated with transarterial chemoembolization (TACE), high and low levels of tumor DPAGT1 and miR-325-3p expression, respectively, were associated with a poor chemotherapeutic response. CONCLUSIONS Our findings provide novel insights into the role of miR-325-3p/DPAGT1 axis dysregulation in supporting HBV + HCC chemoresistance.
Collapse
Affiliation(s)
- Rui Li
- Department of Immunology, Anhui Key Laboratory of Infection and Immunity at Bengbu Medical College, Bengbu, Anhui, China
| | - Tao Xu
- Department of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, China
| | - Hongtao Wang
- Department of Immunology, Anhui Key Laboratory of Infection and Immunity at Bengbu Medical College, Bengbu, Anhui, China
| | - Nan Wu
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical College, China
| | - Fei Liu
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical College, China
| | - Xianjie Jia
- Department of Epidemiology, School of Public Health, Bengbu Medical College, Bengbu, Anhui, China
| | - Jing Mi
- Department of Epidemiology, School of Public Health, Bengbu Medical College, Bengbu, Anhui, China
| | - Jingzhu Lv
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu, Anhui, China.
| | - Huaiquan Gao
- Department of Epidemiology, School of Public Health, Bengbu Medical College, Bengbu, Anhui, China.
| |
Collapse
|
29
|
Gabani BB, Sulochana SP, Kiran V, Todmal U, Mullangi R. Validated LC–ESI–MS/MS method for the determination of tunicamycin in rat plasma: Application to a pharmacokinetic study. Biomed Chromatogr 2019; 33:e4661. [DOI: 10.1002/bmc.4661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/10/2019] [Accepted: 07/15/2019] [Indexed: 11/09/2022]
Affiliation(s)
| | | | - Vinay Kiran
- Drug Metabolism and PharmacokineticsJubilant Biosys Bangalore India
| | - Umesh Todmal
- Drug Metabolism and PharmacokineticsJubilant Biosys Bangalore India
| | - Ramesh Mullangi
- Drug Metabolism and PharmacokineticsJubilant Biosys Bangalore India
| |
Collapse
|
30
|
Shu J, Dang L, Zhang D, Shah P, Chen L, Zhang H, Sun S. Dynamic analysis of proteomic alterations in response to N-linked glycosylation inhibition in a drug-resistant ovarian carcinoma cell line. FEBS J 2019; 286:1594-1605. [PMID: 30884134 PMCID: PMC7360092 DOI: 10.1111/febs.14811] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 01/04/2019] [Accepted: 11/03/2019] [Indexed: 12/20/2022]
Abstract
Glycosylation inhibition can improve the efficacy of antitumor drugs and enhance the apoptosis of cancer cells, thus holding great potential for cancer treatment. Inhibition of N-glycosylation induces endoplasmic reticulum (ER) stress and the unfolded protein response (UPR), and eventually triggers ER stress-related apoptosis. Unfortunately, the detailed timeline of these cell responses and protein expression alterations related to N-glycosylation inhibition is not explicit yet, and the pathways involved in different stages of N-glycosylation inhibition still need to be characterized. In this study, the dynamic proteome alterations related to N-glycosylation inhibition were investigated by further analyzing our previously published quantitative proteomics data from tunicamycin (TM)-treated ovarian carcinoma (OVCAR-3) cells. The results revealed that N-glycosylation inhibition not only directly affects the expression of glycosylated proteins but also alters an extended scale of proteins. Functional annotation of these altered proteins demonstrated that proteins related to ER stress start changing within 6 h, followed by UPR within 24 h, and eventually ER stress-related apoptosis is triggered after 48 h, indicating the conversion of cellular response from positive to negative. The dynamic proteome data presented here provide important information for better understanding of the significance of N-glycosylation to cell survival and TM-related cancer treatment.
Collapse
Affiliation(s)
- Jian Shu
- College of Life Sciences, Northwest University, Xi'an, China
| | - Liuyi Dang
- College of Life Sciences, Northwest University, Xi'an, China
| | - Dandan Zhang
- College of Life Sciences, Northwest University, Xi'an, China
| | - Punit Shah
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Lijun Chen
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Hui Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Shisheng Sun
- College of Life Sciences, Northwest University, Xi'an, China
| |
Collapse
|
31
|
Deng FL, Li L, Huang ZS. Role of breast cancer resistance protein in gastrointestinal tumors. Shijie Huaren Xiaohua Zazhi 2019; 27:395-401. [DOI: 10.11569/wcjd.v27.i6.395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The breast cancer resistance protein, also known as ABCG2, is a member of the drug efflux membrane transporters. As a drug discharge pump, ABCG2 can reduce the concentration of intracellular drugs and protect cells from toxic substances. ABCG2 is not only able to protect normal cells, but also to influence the chemotherapy effect by making tumor cells resistant to various anti-cancer drugs. In order to provide a reference for the basic and clinical research of gastrointestinal tumors, we review the physiological function of ABCG2, factors affecting ABCG2 expression, and its relationship with gastrointestinal tumors.
Collapse
Affiliation(s)
- Feng-Lian Deng
- Guangxi Clinical Medical Research Center of Hepatobiliary Diseases, the Affiliated Hospital of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Li Li
- Guangxi Clinical Medical Research Center of Hepatobiliary Diseases, the Affiliated Hospital of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Zan-Song Huang
- Guangxi Clinical Medical Research Center of Hepatobiliary Diseases, the Affiliated Hospital of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China,Graduate School of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
32
|
Structure-based drug discovery by targeting N-glycan biosynthesis, dolichyl-phosphate N-acetylglucosaminephosphotransferase. Future Med Chem 2019; 11:927-933. [PMID: 30907628 DOI: 10.4155/fmc-2018-0405] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
33
|
Mitachi K, Kurosu SM, Eslamimehr S, Lemieux MR, Ishizaki Y, Clemons WM, Kurosu M. Semisynthesis of an Anticancer DPAGT1 Inhibitor from a Muraymycin Biosynthetic Intermediate. Org Lett 2019; 21:876-879. [PMID: 30698984 PMCID: PMC6447083 DOI: 10.1021/acs.orglett.8b03716] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We have explored a method to convert a muraymycin biosynthetic intermediate 3 to an anticancer drug lead 2 for in vivo and thorough preclinical studies. Cu(OAc)2 forms a stable complex with the amide 4 and prevents electrophilic reactions at the 2-((3-aminopropyl)amino)acetamide moiety. Under the present conditions, the desired 5″-primary amine was selectively protected with (Boc)2O to yield 6. The intermediate 6 was converted to 2 in two steps with 90% yield.
Collapse
Affiliation(s)
- Katsuhiko Mitachi
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Tennessee Health Science Center , 881 Madison Avenue , Memphis , Tennessee 38163 , United States
| | - Shou M Kurosu
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Tennessee Health Science Center , 881 Madison Avenue , Memphis , Tennessee 38163 , United States
| | - Shakiba Eslamimehr
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Tennessee Health Science Center , 881 Madison Avenue , Memphis , Tennessee 38163 , United States
| | - Maddie R Lemieux
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Tennessee Health Science Center , 881 Madison Avenue , Memphis , Tennessee 38163 , United States
| | - Yoshimasa Ishizaki
- Laboratory of Microbiology , Institute of Microbial Chemistry (BIKAKEN) , 3-14-23, Kamiosaki , Shinagawa-ku, Tokyo 141-0021 , Japan
| | - William M Clemons
- Division of Chemistry and Chemical Engineering , California Institute of Technology , 1200 E. California Boulevard , Pasadena , California 91125 , United States
| | - Michio Kurosu
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Tennessee Health Science Center , 881 Madison Avenue , Memphis , Tennessee 38163 , United States
| |
Collapse
|
34
|
Seo J, Lee SH, Park SY, Jeong MH, Lee SY, Kim MJ, Yoo JY, Jang S, Choi KC, Yoon HG. GPR177 promotes gastric cancer proliferation by suppressing endoplasmic reticulum stress-induced cell death. J Cell Biochem 2019; 120:2532-2539. [PMID: 30206979 DOI: 10.1002/jcb.27545] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/06/2018] [Indexed: 01/24/2023]
Abstract
Gastric cancer is the fourth most common cancer worldwide. Despite the high incidence of gastric cancer, efficient chemotherapy treatments still need to be developed. In this study, we examined the anticancer effects of endoplasmic reticulum (ER) stress inducer tunicamycin in gastric cancer. Previously, we found that overexpression of WLS1/GPR177 correlated with poor prognosis in patients with gastric cancer. Furthermore, tunicamycin treatment downregulated GPR177 expression in a dose-dependent manner. GPR177 transports WNT ligand from ER to the plasma membrane, mediating its secretion to the extracellular matrix. In gastric cancer cells, GPR177 preferentially localizes to the ER. Small interfering RNA-mediated knockdown of GPR177 leads to sensitization to ER stress and induces apoptosis of cancer cells along with tunicamycin treatment. GPR177 suppression promoted the ER stress-mediated proapoptotic pathway, such as PERK-CHOP cascade. Furthermore, fluorouracil treatment combined with tunicamycin dramatically reduced cancer cell proliferation. Efficacy of tunicamycin chemotherapy treatments depended on GPR177 expression in gastric cancer cell lines. Together, our results indicate that ER stress can potentiate anticancer effects and suggest GPR177 as a potential gastric cancer therapeutic target.
Collapse
Affiliation(s)
- Jaesung Seo
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-H Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Soo-Y Park
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Mi-H Jeong
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Y Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Mi-J Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Jung-Y Yoo
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Subhin Jang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung-C Choi
- Department of Biomedical Sciences, Asan Medical Center, AMIST, University of Ulsan College of Medicine, Seoul, Korea.,Department of Pharmacology, University of Ulsan College of Medicine, Seoul, Korea
| | - Ho-G Yoon
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
35
|
Wu J, Chen S, Liu H, Zhang Z, Ni Z, Chen J, Yang Z, Nie Y, Fan D. Tunicamycin specifically aggravates ER stress and overcomes chemoresistance in multidrug-resistant gastric cancer cells by inhibiting N-glycosylation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:272. [PMID: 30413206 PMCID: PMC6230241 DOI: 10.1186/s13046-018-0935-8] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023]
Abstract
Background Multidrug resistance remains a major obstacle to successful treatment for patients with gastric cancer (GC). Recently, glycosylation has been demonstrated to play a vital role in the acquisition of multidrug resistance. As a potent inhibitor of glycosylation, tunicamycin (Tu) has shown marked antitumor activities in various cancers. In the present study, we attempted to determine the exact effect of Tu on the chemoresistance of GC. Methods The cytotoxic effects of drugs on GC cells were evaluated by cell viability assays, and apoptosis was detected by flow cytometry. PCR, western blot analysis, immunofluorescence staining and canonical inhibitors were employed to identify the underlying mechanisms of the specific effects of Tu on multidrug-resistant (MDR) GC cells. Results For the first time, we found that MDR GC cells were more sensitive to Tu-induced cell death than the parental cells and that the increased sensitivity might correlate with basal endoplasmic reticulum (ER) stress. In addition, Tu dramatically increased chemotherapy-induced apoptosis by evoking ER stress in GC cells, particularly MDR cells. Further study indicated that these effects were highly dependent on glycosylation inhibition by Tu, rather than its role as a canonical ER stress inducer. Besides, autophagy was markedly triggered by Tu, and blocking autophagy enhanced the combined effects of Tu and chemotherapy on MDR GC cells. Conclusions Our results suggest that tumor-targeted glycosylation inhibition may be a feasible strategy to reverse chemoresistance in GC patients. Electronic supplementary material The online version of this article (10.1186/s13046-018-0935-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jian Wu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Sheng Chen
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Hao Liu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Zhe Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Zhen Ni
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Jie Chen
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Zhiping Yang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China.
| | - Daiming Fan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
36
|
Tsitsipatis D, Jayavelu AK, Müller JP, Bauer R, Schmidt-Arras D, Mahboobi S, Schnöder TM, Heidel F, Böhmer FD. Synergistic killing of FLT3ITD-positive AML cells by combined inhibition of tyrosine-kinase activity and N-glycosylation. Oncotarget 2018; 8:26613-26624. [PMID: 28460451 PMCID: PMC5432283 DOI: 10.18632/oncotarget.15772] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 02/16/2017] [Indexed: 01/05/2023] Open
Abstract
Fms-like tyrosine kinase 3 (FLT3) with internal tandem duplications (ITD) is a major oncoprotein in acute myeloid leukemia (AML), and confers an unfavorable prognosis. Interference with FLT3ITD signaling is therefore pursued as a promising therapeutic strategy. In this study we show that abrogation of FLT3ITD glycoprotein maturation using low doses of the N-glycosylation inhibitor tunicamycin has anti-proliferative and pro-apoptotic effects on FLT3ITD-expressing human and murine cell lines. This effect is mediated in part by arresting FLT3ITD in an underglycosylated state and thereby attenuating FLT3ITD-driven AKT and ERK signaling. In addition, tunicamycin caused pronounced endoplasmatic reticulum stress and apoptosis through activation of protein kinase RNA-like endoplasmic reticulum kinase (PERK) and activation of the gene encoding CCAAT-enhancer-binding protein homologous protein (CHOP). PERK inhibition with a small molecule attenuated CHOP induction and partially rescued cells from apoptosis. Combination of tunicamycin with potent FLT3ITD kinase inhibitors caused synergistic cell killing, which was highly selective for cell lines and primary AML cells expressing FLT3ITD. Although tunicamycin is currently not a clinically applicable drug, we propose that mild inhibition of N-glycosylation may have therapeutic potential in combination with FLT3 kinase inhibitors for FLT3ITD-positive AML.
Collapse
Affiliation(s)
- Dimitrios Tsitsipatis
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany.,Current address: Institute of Nutrition, Department of Nutrigenomics, Friedrich-Schiller-University, Jena, Germany
| | - Ashok Kumar Jayavelu
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany.,Current address: Max-Planck Institute of Biochemistry, Department of Proteomics and Signal Transduction, Martinsried, Germany
| | - Jörg P Müller
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| | - Reinhard Bauer
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| | - Dirk Schmidt-Arras
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Siavosh Mahboobi
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Regensburg, Germany
| | - Tina M Schnöder
- Innere Medizin II, Hämatologie und Onkologie, Universitätsklinikum Jena, Jena, Germany.,Leibniz Institute on Aging, Fritz-Lipmann-Institute, Jena, Germany
| | - Florian Heidel
- Innere Medizin II, Hämatologie und Onkologie, Universitätsklinikum Jena, Jena, Germany.,Leibniz Institute on Aging, Fritz-Lipmann-Institute, Jena, Germany
| | - Frank-D Böhmer
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| |
Collapse
|
37
|
Hou H, Ge C, Sun H, Li H, Li J, Tian H. Tunicamycin inhibits cell proliferation and migration in hepatocellular carcinoma through suppression of CD44s and the ERK1/2 pathway. Cancer Sci 2018; 109:1088-1100. [PMID: 29377347 PMCID: PMC5891198 DOI: 10.1111/cas.13518] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 01/02/2018] [Accepted: 01/22/2018] [Indexed: 12/17/2022] Open
Abstract
Tunicamycin (TM) is an N‐linked glycosylation (NLG) inhibitor with strong antitumor activity, the exact underlying molecular mechanism of which remains to be elucidated. In our previous studies, we found that TM reversed drug resistance and improved the efficacy of combination treatments for hepatocellular carcinomas (HCC). Here, we investigated the effects of TM on HCC cell proliferation and migration as well as the mechanism of those effects. Our results showed that TM inhibited cell proliferation and migration as well as induced apoptosis of hepatocellular carcinoma cells. TM inhibited proliferation of HCC cells by inducing cell apoptosis and cell cycle arrest at the G2/M phase. Meanwhile, TM inhibited migration of HCC cells by suppressing CD44s‐mediated epithelial‐mesenchymal transition (EMT). TM inhibited migration and invasion of HCC cells by decreasing CD44 expression and altering its glycosylation. In addition, CD44s is involved in promoting EMT and is associated with a poor prognosis in HCC patients. Overexpression of CD44s promoted tumor migration and activated phosphorylation of ERK1/2 in HCC cells, whereas TM inhibited CD44s overexpression‐associated cell migration. The ability of TM to inhibit cell migration and invasion was enhanced or reversed in CD44s knockdown cells and cells overexpressing CD44s, respectively. The MEK/ERK inhibitor U0126 and TM inhibited hyaluronic acid‐induced cell migration in HCC cells. Furthermore, TM inhibited exogenous transforming growth factor beta (TGF‐β)‐mediated EMT by an ERK1/2‐dependent mechanism and restored the TGF‐β‐mediated loss of E‐cadherin. In summary, our study provides evidence that TM inhibits proliferation and migration of HCC cells through inhibition of CD44s and the ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Helei Hou
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Chao Ge
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hefen Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hong Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jinjun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
38
|
Yu XS, Du J, Fan YJ, Liu FJ, Cao LL, Liang N, Xu DG, Zhang JD. Activation of endoplasmic reticulum stress promotes autophagy and apoptosis and reverses chemoresistance of human small cell lung cancer cells by inhibiting the PI3K/AKT/mTOR signaling pathway. Oncotarget 2018; 7:76827-76839. [PMID: 27765907 PMCID: PMC5363552 DOI: 10.18632/oncotarget.12718] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/28/2016] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE This study aims to investigate the effects of endoplasmic reticulum stress (ERS) on autophagy, apoptosis and chemoresistance of human small cell lung cancer (SCLC) cells via the PI3K/AKT/mTOR signaling pathway. RESULTS The expressions of ERS-related proteins (PEAK, eIF2α and CHOP) up-regulated, autophagy-related proteins (LC3, LC3-II and Beclin1) and apoptosis-related proteins (Bax and procaspase-3) down-regulated in NCI-H446 and H69 cells after tunicamycin treatment for 24 h. Compared with the blank group, the tunicamycin, BEZ235 and tunicamycin + BEZ235 groups exhibited decreased expressions of p-PI3K, p-AKT and p-mTOR, and increased expressions of autophagy-related proteins (LC3, LC3-II and Beclin1) and apoptosis proteins (Bax and procaspase-3), and the most obvious changes were observed in the tunicamycin + BEZ235 group. MATERIALS AND METHODS CCK-8 assay was applied to select the best cell line from five SCLC cell lines (NCI-H446, H69, H526, H146 and H209). Finally, NCI-H446 and H69 cells were selected for further experiments. NCI-H446/CDDP and H69/CDDP were selected and divided into the blank group, tunicamycin (an ESR inducer) group, BEZ235 (inhibitors of PI3K/AKT/mTOR pathway) group and tunicamycin + BEZ235 group. Cell apoptosis was detected by flow cytometry. Autophagy was observed by fluorescence microscopy and flow cytometry. Western blotting was used to detect the expressions of ERS-related proteins, autophagy-related proteins, apoptosis-related proteins and PI3K/AKT/mTOR pathway-related proteins. CONCLUSIONS Our findings provide evidence that the activation of ERS could promote autophagy and apoptosis and reverse chemoresistance of human SCLC cells by inhibiting the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Xin-Shuang Yu
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P.R. China
| | - Juan Du
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P.R. China.,Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P.R. China
| | - Yu-Jun Fan
- Medical Management Service Center of Shandong Provincial Health and Family Planning Commission, Jinan 250014, P.R. China
| | - Feng-Jun Liu
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P.R. China
| | - Li-Li Cao
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P.R. China
| | - Ning Liang
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P.R. China
| | - De-Guo Xu
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P.R. China
| | - Jian-Dong Zhang
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, P.R. China
| |
Collapse
|
39
|
Yotsumoto K, Akiyoshi T, Wada N, Imaoka A, Ohtani H. 5-Fluorouracil treatment alters the expression of intestinal transporters in rats. Biopharm Drug Dispos 2017; 38:509-516. [PMID: 28849584 DOI: 10.1002/bdd.2102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/05/2017] [Accepted: 08/14/2017] [Indexed: 01/26/2023]
Abstract
5-Fluorouracil (5-FU), an anticancer drug, causes severe gastrointestinal damage, which may affect the absorption of orally administered drugs including the substrates of intestinal uptake and efflux transporters. This study aimed to investigate quantitatively the effect of 5-FU-induced intestinal damage on the expression of intestinal transporters: P-glycoprotein (P-gp), breast cancer resistance protein (BCRP) and peptide transporter 1 (PEPT1) in rats. The rats were treated with 5-FU (30 mg/kg/day, p.o.) for 5 days to induce intestinal damage, and then the upper, middle and lower intestinal segments were removed. The mRNA and protein expression levels of these transporters in each segment were determined using quantitative real-time PCR and Western blotting, respectively. In the 5-FU-treated rats, the protein levels of P-gp and Bcrp in the upper segment were significantly increased to 15- and 2.6-fold of the control, respectively, while those in other segments were unaffected. Pept1 expression was increased by 5-FU in almost all segments. A remarkable increase in P-gp expression was shown, the uptake of digoxin, a P-gp substrate, in each intestinal segment was measured using a rat everted sac. As a result, the uptake of digoxin in the upper segments of 5-FU-treated rats was decreased compared with that of the control. In conclusion, 5-FU-induced intestinal damage was shown to alter the expression of these transporters, especially in the upper intestinal segment, while the characteristics of the influence varied among the transporters. The 5-FU-induced intestinal damage may affect transporter-mediated drug absorption of orally administered drugs in the clinical setting.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- Administration, Oral
- Animals
- Antimetabolites, Antineoplastic/toxicity
- Blotting, Western
- Digoxin/pharmacokinetics
- Drug Interactions
- Fluorouracil/toxicity
- Gene Expression Regulation/drug effects
- Intestines/drug effects
- Intestines/pathology
- Male
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Peptide Transporter 1/genetics
- Peptide Transporter 1/metabolism
- Rats
- Rats, Sprague-Dawley
- Real-Time Polymerase Chain Reaction
Collapse
Affiliation(s)
- Keiichi Yotsumoto
- Division of Clinical Pharmacy, Keio Graduate School of Pharmaceutical Sciences, Tokyo, Japan
| | - Takeshi Akiyoshi
- Division of Clinical Pharmacy, Keio Graduate School of Pharmaceutical Sciences, Tokyo, Japan
| | - Naoki Wada
- Division of Clinical Pharmacy, Keio Graduate School of Pharmaceutical Sciences, Tokyo, Japan
| | - Ayuko Imaoka
- Division of Clinical Pharmacy, Keio Graduate School of Pharmaceutical Sciences, Tokyo, Japan
| | - Hisakazu Ohtani
- Division of Clinical Pharmacy, Keio Graduate School of Pharmaceutical Sciences, Tokyo, Japan
| |
Collapse
|
40
|
Pedriali G, Rimessi A, Sbano L, Giorgi C, Wieckowski MR, Previati M, Pinton P. Regulation of Endoplasmic Reticulum-Mitochondria Ca 2+ Transfer and Its Importance for Anti-Cancer Therapies. Front Oncol 2017; 7:180. [PMID: 28913175 PMCID: PMC5583168 DOI: 10.3389/fonc.2017.00180] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/07/2017] [Indexed: 12/18/2022] Open
Abstract
Inter-organelle membrane contact sites are emerging as major sites for the regulation of intracellular Ca2+ concentration and distribution. Here, extracellular stimuli operate on a wide array of channels, pumps, and ion exchangers to redistribute intracellular Ca2+ among several compartments. The resulting highly defined spatial and temporal patterns of Ca2+ movement can be used to elicit specific cellular responses, including cell proliferation, migration, or death. Plasma membrane (PM) also can directly contact mitochondria and endoplasmic reticulum (ER) through caveolae, small invaginations of the PM that ensure inter-organelle contacts, and can contribute to the regulation of numerous cellular functions through scaffolding proteins such as caveolins. PM and ER organize specialized junctions. Here, many components of the receptor-dependent Ca2+ signals are clustered, including the ORAI1-stromal interaction molecule 1 complex. This complex constitutes a primary mechanism for Ca2+ entry into non-excitable cells, modulated by intracellular Ca2+. Several contact sites between the ER and mitochondria, termed mitochondria-associated membranes, show a very complex and specialized structure and host a wide number of proteins that regulate Ca2+ transfer. In this review, we summarize current knowledge of the particular action of several oncogenes and tumor suppressors at these specialized check points and analyze anti-cancer therapies that specifically target Ca2+ flow at the inter-organelle contacts to alter the metabolism and fate of the cancer cell.
Collapse
Affiliation(s)
- Gaia Pedriali
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Alessandro Rimessi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Luigi Sbano
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Carlotta Giorgi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Mariusz R Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Maurizio Previati
- Department of Morphology, Surgery and Experimental Medicine, Section of Human Anatomy and Histology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| |
Collapse
|
41
|
Endoplasmic Reticulum Stress Inducer Tunicamycin Alters Hepatic Energy Homeostasis in Mice. Int J Mol Sci 2017; 18:ijms18081710. [PMID: 28777337 PMCID: PMC5578100 DOI: 10.3390/ijms18081710] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 08/03/2017] [Accepted: 08/03/2017] [Indexed: 12/19/2022] Open
Abstract
Disorders of hepatic energy metabolism, which can be regulated by endoplasmic reticulum (ER) stress, lead to metabolic diseases such as hepatic steatosis and hypoglycemia. Tunicamycin, a pharmacological ER stress inducer, is used to develop an anti-cancer drug. However, the effects of tunicamycin on hepatic energy metabolism have not been well elucidated. Mice were intraperitoneally injected with tunicamycin or vehicle. Twenty-four hours later, hepatic triglyceride and glycogen content and serum lipids profiles were analyzed, as well as the expression of lipogenic and gluconeogenic genes. Tunicamycin significantly induced hepatic a yellowish color and ER stress, as well as increasing serum levels of aspartate transaminase and alanine transaminase. Besides, tunicamycin remarkably increased hepatic triglyceride content and suppressed the expression of apolipoprotein B100. In addition, tunicamycin-treated mice had lower serum levels of triglyceride, apolipoprotein B, low-density lipoprotein cholesterol and high-density lipoprotein cholesterol. Gene expression of peroxisome proliferator-activated receptor α was decreased by tunicamycin, but the protein level was increased. Furthermore, blood glucose level and hepatic glycogen content were decreased in tunicamycin-treated mice. Protein kinase B signaling was attenuated in the tunicamycin-treated liver, but the expression and activities of phosphoenolpyruvate carboxykinase and glucose-6-phosphatase were unchanged. Tunicamycin alters hepatic energy homeostasis by increasing triglyceride accumulation and decreasing glycogen content.
Collapse
|
42
|
Li XF, Huang QY, Yang WZ, Wang HJ, Li CW. Alterations in ACE and ABCG2 expression levels in the testes of rats subjected to atropine-induced toxicity. Mol Med Rep 2016; 14:5211-5216. [PMID: 27779686 DOI: 10.3892/mmr.2016.5857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 07/20/2016] [Indexed: 11/06/2022] Open
Abstract
Atropine-induced damage is associated with enzyme and protein alterations. The aim of the present study was to investigate atropine‑induced alterations in testicular expression levels of angiotensin‑converting enzyme (ACE) and adenosine 5'-triphosphate binding cassette sub‑family G member 2 (ABCG2) following atropine treatment. Male Wistar rats received 15 mg/kg/day atropine for 7 days; control rats received an identical volume of saline, Following treatment, the testes were harvested for immunohistochemistry and in situ hybridization to examine the protein and gene expression levels of ACE and ABCG2 by digital image analysis. ACE gene and protein expression levels were significantly reduced in the testes of atropine‑treated rats, compared with control rats (P=0.0001 and P<0.001, respectively). In addition, ABCG2 gene and protein expression levels were significantly increased in the testes of atropine‑treated rats, compared with control rats (P=0.0017 and P<0.001, respectively). Thus, the results of the present study demonstrate that testicular protein and gene expression levels of ACE and ABCG2 were altered as a result of atropine‑induced toxicity in the rats. These alterations may result in abnormal testicular function, and the proteins and genes identified in the present study may be useful to elucidate the mechanisms underlying atropine‑induced toxicity and provide a direction for further studies.
Collapse
Affiliation(s)
- Xue-Fang Li
- Library of Dali University, Dali, Yunnan 671003, P.R. China
| | - Quan-Yong Huang
- Department of Pathology, School of Basic Medical Sciences, Dali University, Dali, Yunnan 671000, P.R. China
| | - Wen-Zhong Yang
- Department of Pathology, School of Basic Medical Sciences, Dali University, Dali, Yunnan 671000, P.R. China
| | - Hui-Jie Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Dali University, Dali, Yunnan 671000, P.R. China
| | - Can-Wei Li
- Department of Epidemiology and Health Statistics, School of Public Health, Dali University, Dali, Yunnan 671000, P.R. China
| |
Collapse
|
43
|
Zhang L, Ge C, Zhao F, Zhang Y, Wang X, Yao M, Li J. NRBP2 Overexpression Increases the Chemosensitivity of Hepatocellular Carcinoma Cells via Akt Signaling. Cancer Res 2016; 76:7059-7071. [PMID: 27634758 DOI: 10.1158/0008-5472.can-16-0937] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/24/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022]
Abstract
Hepatocellular carcinoma is highly resistant to chemotherapy. Research data supported that cancer stem cells (CSC) may be responsible for the chemoresistance and strategies that suppress CSCs stemness could also inhibit the drug resistance. In this study, we found that nuclear receptor binding protein 2 (NRBP2) expression was downregulated in the CD133+ hepatocellular carcinoma CSCs. Most adjacent noncancerous liver tissue analyzed expressed higher level of NRBP2 compared with cancerous tissue in hepatocellular carcinoma patients, and high NRBP2 expression indicated a better prognosis. Real-time PCR results showed that NRBP2 negatively correlated with stemness-related genes, including Oct3/4, Nanog, Notch1, Ep300, and CD133 mRNA expression. High NRBP2 expression in hepatocellular carcinoma cells downregulated CK19 protein expression, inhibited tumorsphere formation, and tumorigenesis ability, indicating that high NRBP2 expression restrains the hepatocellular carcinoma cell stemness. Overexpression of NRBP2 reduced the IC50 of sorafenib in hepatocellular carcinoma cells, and NRBP2 expression was negatively correlated with hepatocellular carcinoma cell resistance to the chemotherapy agents, including cisplatin and the Akt signaling inhibitor perifosine. Coimmunoprecipitation results showed that NRBP2 could bind with Annexin A2 (ANXA2) and inhibit ANXA2 expression. Coexpression of ANXA2 restored the chemoresistant ability in NRBP2-overexpressing hepatocellular carcinoma cells. Further analysis showed that NRBP2 downregulated Akt and its downstream signaling target Bad phosphorylation level. ANXA2 coexpression partially restored the Akt phosphorylation. Analysis of the expression of Bcl2 family proteins showed that NRBP2 may increase hepatocellular carcinoma cell chemosensitivity by regulating expression of survival proteins involved in the Akt and Bcl2 pathway. These results suggest that NRBP2 plays an important role in the tumor progression and chemotherapeutic resistance of hepatocellular carcinoma. Cancer Res; 76(23); 7059-71. ©2016 AACR.
Collapse
Affiliation(s)
- Lixing Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chao Ge
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fangyu Zhao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xin Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jinjun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
44
|
Ranji P, Salmani Kesejini T, Saeedikhoo S, Alizadeh AM. Targeting cancer stem cell-specific markers and/or associated signaling pathways for overcoming cancer drug resistance. Tumour Biol 2016; 37:13059-13075. [DOI: 10.1007/s13277-016-5294-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023] Open
|
45
|
Nami B, Donmez H, Kocak N. Tunicamycin-induced endoplasmic reticulum stress reduces in vitro subpopulation and invasion of CD44+/CD24- phenotype breast cancer stem cells. ACTA ACUST UNITED AC 2016; 68:419-26. [PMID: 27350212 DOI: 10.1016/j.etp.2016.06.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 06/13/2016] [Indexed: 02/07/2023]
Abstract
Tunicamycin is an inhibitor of glycosylation that disturbs protein folding machinery in eukaryotic cells. Tunicamycin causes accumulation of unfolded proteins in cell endoplasmic reticulum (ER) and induces ER stress. ER stress is an essential mechanism for cellular homeostasis has role in cell death via reprogramming of protein processing, regulation of autophagy and apoptosis. In this study we show effect of tunicamycin on subpopulation and invasion of CD44+/CD24- MCF7 breast cancer stem cells. CD44+/CD24- cells were isolated from MCF7 cell line by fluorescence activated cell sorting (FACS) and treated with tunicamycin. ER stress was monitored by evaluation of X-box binding protein 1(XBP-1) mRNA splicing, cleaved activating transcription factor 6 (ATF6) nuclear translocation and CCAAT/enhancer-binding protein homologous protein (CHOP) expression. CD44+/CD24- subpopulation was analyzed using flow cytometry. Invasion was investigated by scratch assay, trypan blue staining, 3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyltetrazolium bromide (MTT) proliferation and in vitro migration assays. Increased level of spliced XBP-1, ATF6 nuclear translocation and CHOP protein expression were detected in CD44+/CD24- and original MCF7 cells treated with tunicamycin. Also, a significant decline in CD44+/CD24- cell subpopulation was determined in the cells treated with tunicamycin. The results also showed inhibited invasion, increased cell death, suppressed proliferation and reduced migration in the CD44+/CD24- and CD44+/CD24- rich MCF7 cell culture, under effect of tunicamycin. Our results indicate that CD44+/CD24- phenotype MCF7 cells are susceptible to tunicamycin. The results showed that tunicamycin-induced ER stress suppresses CD44+/CD24- phenotype cell subpopulation and in vitro invasion and accelerates tumorosphore formation. These results suggest that tunicamycin-induced ER stress inhibits CD44+/CD24- phenotype MCF7 breast cancer stem cells. We conclude that using ER-targeting chemicals like tunicamycin is an interesting approach to target breast cancer stem cells inside tumor.
Collapse
Affiliation(s)
- Babak Nami
- Department of Medical Genetics and Women and Children's Health Research Institute (WCHRI), University of Alberta, Edmonton, Canada.
| | - Huseyin Donmez
- Department of Medical Genetics, Selcuk University Medical Faculty, Konya, Turkey.
| | - Nadir Kocak
- Department of Medical Genetics, Selcuk University Medical Faculty, Konya, Turkey.
| |
Collapse
|
46
|
Yang L, Wang J, Yang J, Schamber R, Hu N, Nair S, Xiong L, Ren J. Antioxidant metallothionein alleviates endoplasmic reticulum stress-induced myocardial apoptosis and contractile dysfunction. Free Radic Res 2016; 49:1187-98. [PMID: 25968954 DOI: 10.3109/10715762.2015.1013952] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
AIMS Endoplasmic reticulum (ER) stress exerts myocardial oxidative stress, apoptosis, and contractile anomalies, although the precise interplay between ER stress and apoptosis remains elusive. This study was designed to examine the impact of the cysteine-rich free radical scavenger metallothionein on ER stress-induced myocardial contractile defect and underlying mechanisms. METHODS AND RESULTS Wild-type friendly virus B and transgenic mice with cardiac-specific overexpression of metallothionein were challenged with the ER stress inducer tunicamycin (1 mg/kg, intraperitoneal, 48 h) prior to the assessment of myocardial function, oxidative stress, and apoptosis. Our results revealed that tunicamycin promoted cardiac remodeling (enlarged left ventricular end systolic/diastolic diameters with little changes in left ventricular wall thickness), suppressed fractional shortening and cardiomyocyte contractile function, elevated resting Ca(2+), decreased stimulated Ca(2+) release, prolonged intracellular Ca(2+) clearance, and downregulated sarco(endo)plasmic reticulum Ca(2+)-ATPase levels, the effects of which were negated by metallothionein. Treatment with tunicamycin caused cardiomyocyte mitochondrial injury, as evidenced by decreased mitochondrial membrane potential (∆Ѱm, assessed by JC-1 staining), the effect of which was negated by the antioxidant. Moreover, tunicamycin challenge dramatically facilitated myocardial apoptosis as manifested by increased Bax, caspase 9, and caspase 12 protein levels, as well as elevated caspase 3 activity. Interestingly, metallothionein transgene significantly alleviated tunicamycin-induced myocardial apoptosis. CONCLUSION Taken together, our data favor a beneficial effect of metallothionein against ER stress-induced cardiac dysfunction possibly associated with attenuation of myocardial apoptosis.
Collapse
Affiliation(s)
- L Yang
- a Department of Anesthesiology , Xijing Hospital, the Fourth Military Medical University , Xi'an , P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Podolski-Renić A, Milošević Z, Dinić J, Stanković T, Banković J, Pešić M. Mutual regulation and targeting of multidrug resistance and cancer stem phenotype. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00391e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Targeting stemness mechanisms leads to the suppression of ABC transporter activity and elimination of CSCs.
Collapse
Affiliation(s)
- Ana Podolski-Renić
- Institute for Biological Research “Siniša Stanković”
- University of Belgrade
- 11060 Belgrade
- Serbia
| | - Zorica Milošević
- Institute for Biological Research “Siniša Stanković”
- University of Belgrade
- 11060 Belgrade
- Serbia
| | - Jelena Dinić
- Institute for Biological Research “Siniša Stanković”
- University of Belgrade
- 11060 Belgrade
- Serbia
| | - Tijana Stanković
- Institute for Biological Research “Siniša Stanković”
- University of Belgrade
- 11060 Belgrade
- Serbia
| | - Jasna Banković
- Institute for Biological Research “Siniša Stanković”
- University of Belgrade
- 11060 Belgrade
- Serbia
| | - Milica Pešić
- Institute for Biological Research “Siniša Stanković”
- University of Belgrade
- 11060 Belgrade
- Serbia
| |
Collapse
|
48
|
Dong F, Eibach M, Bartsch JW, Dolga AM, Schlomann U, Conrad C, Schieber S, Schilling O, Biniossek ML, Culmsee C, Strik H, Koller G, Carl B, Nimsky C. The metalloprotease-disintegrin ADAM8 contributes to temozolomide chemoresistance and enhanced invasiveness of human glioblastoma cells. Neuro Oncol 2015; 17:1474-85. [PMID: 25825051 PMCID: PMC4648299 DOI: 10.1093/neuonc/nov042] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/22/2015] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Despite multimodal treatment, glioblastoma (GBM) therapy with temozolomide (TMZ) remains inefficient due to chemoresistance. Matrix metalloproteinase (MMP) and a disintegrin and metalloprotease (ADAM), increased in GBM, could contribute to chemoresistance and TMZ-induced recurrence of glioblastoma. METHODS TMZ inducibility of metalloproteases was determined in GBM cell lines, primary GBM cells, and tissues from GBM and recurrent GBM. TMZ sensitivity and invasiveness of GBM cells were assessed in the presence of the metalloprotease inhibitors batimastat (BB-94) and marimastat (BB-2516). Metalloprotease-dependent effects of TMZ on mitochondria and pAkt/phosphatidylinositol-3 kinase (PI3K) and phosphorylated extracellular signal-regulated kinase 1/2 (pERK1/2) pathways were analyzed by fluorescence activated cell sorting, morphometry, and immunoblotting. Invasiveness of GBM cells was determined by Matrigel invasion assays. Potential metalloprotease substrates were identified by proteomics and tested for invasion using blocking antibodies. RESULTS TMZ induces expression of MMP-1, -9, -14, and ADAM8 in GBM cells and in recurrent GBM tissues. BB-94, but not BB-2516 (ADAM8-sparing) increased TMZ sensitivity of TMZ-resistant and -nonresistant GBM cells with different O(6)-methylguanine-DNA methyltransferase states, suggesting that ADAM8 mediates chemoresistance, which was confirmed by ADAM8 knockdown, ADAM8 overexpression, or pharmacological inhibition of ADAM8. Levels of pAkt and pERK1/2 were increased in GBM cells and correlated with ADAM8 expression, cell survival, and invasiveness. Soluble hepatocyte growth factor (HGF) R/c-met and CD44 were identified as metalloprotease substrates in TMZ-treated GBM cells. Blocking of HGF R/c-met prevented TMZ-induced invasiveness. CONCLUSIONS ADAM8 causes TMZ resistance in GBM cells by enhancing pAkt/PI3K, pERK1/2, and cleavage of CD44 and HGF R/c-met. Specific ADAM8 inhibition can optimize TMZ chemotherapy of GBM in order to prevent formation of recurrent GBM in patients.
Collapse
Affiliation(s)
| | | | | | - Amalia M. Dolga
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany (F.D., M.E., J.W.B., U.S., C.Co., S.S., B.C., C.N.); Department of Neurosurgery, Tongji Hospital, Wuhan, China (F.D.); Philipps-University Marburg, Institute for Pharmacology and Clinical Pharmacy, Marburg, Germany (A.M.D., C.Cu.); Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany (O.S., M.L.B.); BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany (O.S.); Department of Neurology, Philipps-University Marburg, Marburg, Germany (H.S.); Biomaterials, Biomimetics and Biophotonics Research Group, King's College London Dental Institute, London, United Kingdom (G.K.)
| | - Uwe Schlomann
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany (F.D., M.E., J.W.B., U.S., C.Co., S.S., B.C., C.N.); Department of Neurosurgery, Tongji Hospital, Wuhan, China (F.D.); Philipps-University Marburg, Institute for Pharmacology and Clinical Pharmacy, Marburg, Germany (A.M.D., C.Cu.); Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany (O.S., M.L.B.); BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany (O.S.); Department of Neurology, Philipps-University Marburg, Marburg, Germany (H.S.); Biomaterials, Biomimetics and Biophotonics Research Group, King's College London Dental Institute, London, United Kingdom (G.K.)
| | - Catharina Conrad
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany (F.D., M.E., J.W.B., U.S., C.Co., S.S., B.C., C.N.); Department of Neurosurgery, Tongji Hospital, Wuhan, China (F.D.); Philipps-University Marburg, Institute for Pharmacology and Clinical Pharmacy, Marburg, Germany (A.M.D., C.Cu.); Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany (O.S., M.L.B.); BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany (O.S.); Department of Neurology, Philipps-University Marburg, Marburg, Germany (H.S.); Biomaterials, Biomimetics and Biophotonics Research Group, King's College London Dental Institute, London, United Kingdom (G.K.)
| | - Susanne Schieber
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany (F.D., M.E., J.W.B., U.S., C.Co., S.S., B.C., C.N.); Department of Neurosurgery, Tongji Hospital, Wuhan, China (F.D.); Philipps-University Marburg, Institute for Pharmacology and Clinical Pharmacy, Marburg, Germany (A.M.D., C.Cu.); Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany (O.S., M.L.B.); BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany (O.S.); Department of Neurology, Philipps-University Marburg, Marburg, Germany (H.S.); Biomaterials, Biomimetics and Biophotonics Research Group, King's College London Dental Institute, London, United Kingdom (G.K.)
| | - Oliver Schilling
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany (F.D., M.E., J.W.B., U.S., C.Co., S.S., B.C., C.N.); Department of Neurosurgery, Tongji Hospital, Wuhan, China (F.D.); Philipps-University Marburg, Institute for Pharmacology and Clinical Pharmacy, Marburg, Germany (A.M.D., C.Cu.); Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany (O.S., M.L.B.); BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany (O.S.); Department of Neurology, Philipps-University Marburg, Marburg, Germany (H.S.); Biomaterials, Biomimetics and Biophotonics Research Group, King's College London Dental Institute, London, United Kingdom (G.K.)
| | - Martin L. Biniossek
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany (F.D., M.E., J.W.B., U.S., C.Co., S.S., B.C., C.N.); Department of Neurosurgery, Tongji Hospital, Wuhan, China (F.D.); Philipps-University Marburg, Institute for Pharmacology and Clinical Pharmacy, Marburg, Germany (A.M.D., C.Cu.); Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany (O.S., M.L.B.); BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany (O.S.); Department of Neurology, Philipps-University Marburg, Marburg, Germany (H.S.); Biomaterials, Biomimetics and Biophotonics Research Group, King's College London Dental Institute, London, United Kingdom (G.K.)
| | - Carsten Culmsee
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany (F.D., M.E., J.W.B., U.S., C.Co., S.S., B.C., C.N.); Department of Neurosurgery, Tongji Hospital, Wuhan, China (F.D.); Philipps-University Marburg, Institute for Pharmacology and Clinical Pharmacy, Marburg, Germany (A.M.D., C.Cu.); Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany (O.S., M.L.B.); BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany (O.S.); Department of Neurology, Philipps-University Marburg, Marburg, Germany (H.S.); Biomaterials, Biomimetics and Biophotonics Research Group, King's College London Dental Institute, London, United Kingdom (G.K.)
| | - Herwig Strik
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany (F.D., M.E., J.W.B., U.S., C.Co., S.S., B.C., C.N.); Department of Neurosurgery, Tongji Hospital, Wuhan, China (F.D.); Philipps-University Marburg, Institute for Pharmacology and Clinical Pharmacy, Marburg, Germany (A.M.D., C.Cu.); Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany (O.S., M.L.B.); BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany (O.S.); Department of Neurology, Philipps-University Marburg, Marburg, Germany (H.S.); Biomaterials, Biomimetics and Biophotonics Research Group, King's College London Dental Institute, London, United Kingdom (G.K.)
| | - Garrit Koller
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany (F.D., M.E., J.W.B., U.S., C.Co., S.S., B.C., C.N.); Department of Neurosurgery, Tongji Hospital, Wuhan, China (F.D.); Philipps-University Marburg, Institute for Pharmacology and Clinical Pharmacy, Marburg, Germany (A.M.D., C.Cu.); Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany (O.S., M.L.B.); BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany (O.S.); Department of Neurology, Philipps-University Marburg, Marburg, Germany (H.S.); Biomaterials, Biomimetics and Biophotonics Research Group, King's College London Dental Institute, London, United Kingdom (G.K.)
| | - Barbara Carl
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany (F.D., M.E., J.W.B., U.S., C.Co., S.S., B.C., C.N.); Department of Neurosurgery, Tongji Hospital, Wuhan, China (F.D.); Philipps-University Marburg, Institute for Pharmacology and Clinical Pharmacy, Marburg, Germany (A.M.D., C.Cu.); Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany (O.S., M.L.B.); BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany (O.S.); Department of Neurology, Philipps-University Marburg, Marburg, Germany (H.S.); Biomaterials, Biomimetics and Biophotonics Research Group, King's College London Dental Institute, London, United Kingdom (G.K.)
| | - Christopher Nimsky
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany (F.D., M.E., J.W.B., U.S., C.Co., S.S., B.C., C.N.); Department of Neurosurgery, Tongji Hospital, Wuhan, China (F.D.); Philipps-University Marburg, Institute for Pharmacology and Clinical Pharmacy, Marburg, Germany (A.M.D., C.Cu.); Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany (O.S., M.L.B.); BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany (O.S.); Department of Neurology, Philipps-University Marburg, Marburg, Germany (H.S.); Biomaterials, Biomimetics and Biophotonics Research Group, King's College London Dental Institute, London, United Kingdom (G.K.)
| |
Collapse
|
49
|
Zhang L, Su J, Xie Q, Zeng L, Wang Y, Yi D, Yu Y, Liu S, Li S, Xu Y. 2-Deoxy-d-Glucose Sensitizes Human Ovarian Cancer Cells to Cisplatin by Increasing ER Stress and Decreasing ATP Stores in Acidic Vesicles. J Biochem Mol Toxicol 2015; 29:572-8. [PMID: 26241884 DOI: 10.1002/jbt.21730] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 06/15/2015] [Accepted: 06/17/2015] [Indexed: 01/07/2023]
Abstract
Cisplatin is a commonly used chemotherapeutic agent; however, the development of acquired resistance limits its application. Here, we demonstrate that 2-deoxy-d-glucose (2-DG) enhanced the antitumor effects of cisplatin in SKOV3 cells, which include inhibition of proliferation and promotion of apoptosis. Additionally, either cisplatin or 2-DG alone could upregulate the endoplasmic reticulum (ER) stress-associated protein glucose-regulated protein-78 (GRP78). Moreover, exposure to 2-DG increased the expression of GRP78 induced by cisplatin. Cisplatin also upregulated ER stress-associated apoptotic protein 153/C/EBP homology protein (CHOP) in SKOV3 cells. While treatment with 2-DG alone could not upregulate the CHOP expression, a combination of both 2-DG and cisplatin increased the protein levels of CHOP above those induced by Cisplatin alone. Finally, cisplatin mediated an increase in ATP stores within acidic vesicles, whereas 2-DG decreased this effect. These data demonstrate that 2-DG sensitizes SKOV3 cells to cisplatin by increasing ER stress and decreasing ATP stores in acidic vesicles.
Collapse
Affiliation(s)
- Lili Zhang
- School of Public Health, Jilin Medical University, Jilin, Jilin, People's Republic of China
| | - Jing Su
- Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin, People's Republic of China
| | - Qi Xie
- Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin, People's Republic of China
| | - Linchuan Zeng
- School of Public Health, Jilin Medical University, Jilin, Jilin, People's Republic of China
| | - Yan Wang
- School of Public Health, Jilin Medical University, Jilin, Jilin, People's Republic of China
| | - Dan Yi
- Medical Examination Center, Jilin Traditional Chinese and Western Medicine Hospital, Jilin, Jilin, People's Republic of China
| | - Yang Yu
- Medical Research Laboratory, Jilin Medical University, Jilin, Jilin, People's Republic of China
| | - Shibing Liu
- Medical Research Laboratory, Jilin Medical University, Jilin, Jilin, People's Republic of China
| | - Songyan Li
- Medical Research Laboratory, Jilin Medical University, Jilin, Jilin, People's Republic of China
| | - Ye Xu
- Medical Research Laboratory, Jilin Medical University, Jilin, Jilin, People's Republic of China. .,Department of Histology and Embryology, Jilin Medical University, Jilin, Jilin, People's Republic of China.
| |
Collapse
|
50
|
Rimessi A, Patergnani S, Bonora M, Wieckowski MR, Pinton P. Mitochondrial Ca(2+) Remodeling is a Prime Factor in Oncogenic Behavior. Front Oncol 2015; 5:143. [PMID: 26161362 PMCID: PMC4479728 DOI: 10.3389/fonc.2015.00143] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 06/11/2015] [Indexed: 12/30/2022] Open
Abstract
Cancer is sustained by defects in the mechanisms underlying cell proliferation, mitochondrial metabolism, and cell death. Mitochondrial Ca2+ ions are central to all these processes, serving as signaling molecules with specific spatial localization, magnitude, and temporal characteristics. Mutations in mtDNA, aberrant expression and/or regulation of Ca2+-handling/transport proteins and abnormal Ca2+-dependent relationships among the cytosol, endoplasmic reticulum, and mitochondria can cause the deregulation of mitochondrial Ca2+-dependent pathways that are related to these processes, thus determining oncogenic behavior. In this review, we propose that mitochondrial Ca2+ remodeling plays a pivotal role in shaping the oncogenic signaling cascade, which is a required step for cancer formation and maintenance. We will describe recent studies that highlight the importance of mitochondria in inducing pivotal “cancer hallmarks” and discuss possible tools to manipulate mitochondrial Ca2+ to modulate cancer survival.
Collapse
Affiliation(s)
- Alessandro Rimessi
- Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, University of Ferrara , Ferrara , Italy
| | - Simone Patergnani
- Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, University of Ferrara , Ferrara , Italy
| | - Massimo Bonora
- Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, University of Ferrara , Ferrara , Italy
| | - Mariusz R Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology , Warsaw , Poland
| | - Paolo Pinton
- Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, University of Ferrara , Ferrara , Italy
| |
Collapse
|