1
|
Mashayekhi-Sardoo H, Rezaee R, Yarmohammadi F, Karimi G. Targeting Endoplasmic Reticulum Stress by Natural and Chemical Compounds Ameliorates Cisplatin-Induced Nephrotoxicity: A Review. Biol Trace Elem Res 2024:10.1007/s12011-024-04351-w. [PMID: 39212819 DOI: 10.1007/s12011-024-04351-w] [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] [Received: 05/17/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
Cisplatin is a chemotherapeutic that dose-dependently causes renal complications such as decreased kidney function and acute kidney injury. The endoplasmic reticulum (ER) is responsible for calcium homeostasis and protein folding and plays a major part in cisplatin's nephrotoxicity. The current article reviews how chemical and natural compounds modulate cisplatin-induced apoptosis, autophagy, and inflammation by inhibiting ER stress signaling pathways. The available evidence indicates that natural compounds (Achyranthes aspera water-soluble extract, morin hydrate, fucoidan, isoliquiritigenin, leonurine, epigallocatechin-3-gallate, grape seed proanthocyanidin, and ginseng polysaccharide) and chemicals (Sal003, NSC228155, TUG891, dorsomorphin (compound C), HC-030031, dexmedetomidine, and recombinant human erythropoietin (rHuEpo)) can alleviate cisplatin nephrotoxicity by suppression of ER stress signaling pathways including IRE1α/ASK1/JNK, PERK-eIF2α-ATF4, and ATF6, as well as PI3K/AKT signaling pathway. Since ER and related signaling pathways are important in cisplatin nephrotoxicity, agents that can inhibit the abovementioned signaling pathways may hold promise in alleviating this untoward adverse effect.
Collapse
Affiliation(s)
- Habibeh Mashayekhi-Sardoo
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Ramin Rezaee
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Yarmohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical, P. O. Box, Sciences, Mashhad, 1365-91775, Iran.
| |
Collapse
|
2
|
Penugurti V, Manne RK, Bai L, Kant R, Lin HK. AMPK: The energy sensor at the crossroads of aging and cancer. Semin Cancer Biol 2024; 106-107:15-27. [PMID: 39197808 DOI: 10.1016/j.semcancer.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024]
Abstract
AMP-activated protein kinase (AMPK) is a protein kinase that plays versatile roles in response to a variety of physiological stresses, including glucose deprivation, hypoxia, and ischemia. As a kinase with pleiotropic functions, it plays a complex role in tumor progression, exhibiting both tumor-promoting and tumor-suppressing activities. On one hand, AMPK enhances cancer cell proliferation and survival, promotes cancer metastasis, and impairs anti-tumor immunity. On the other hand, AMPK inhibits cancer cell growth and survival and stimulates immune responses in a context-dependent manner. Apart from these functions, AMPK plays a key role in orchestrating aging and aging-related disorders, including cardiovascular diseases (CVD), Osteoarthritis (OA), and Diabetes. In this review article, we summarized the functions of AMPK pathway based on its oncogenic and tumor-suppressive roles and highlighted the importance of AMPK pathway in regulating cellular aging. We also spotlighted the significant role of various signaling pathways, activators, and inhibitors of AMPK in serving as therapeutic strategies for anti-cancer and anti-aging therapy.
Collapse
Affiliation(s)
- Vasudevarao Penugurti
- Department of Pathology, School of Medicine, Duke University, Durham, NC 27710, United States
| | - Rajesh Kumar Manne
- Department of Pathology, School of Medicine, Duke University, Durham, NC 27710, United States
| | - Ling Bai
- Department of Pathology, School of Medicine, Duke University, Durham, NC 27710, United States
| | - Rajni Kant
- Department of Pathology, School of Medicine, Duke University, Durham, NC 27710, United States
| | - Hui-Kuan Lin
- Department of Pathology, School of Medicine, Duke University, Durham, NC 27710, United States.
| |
Collapse
|
3
|
Jang M, Park R, Yamamoto A, Park YI, Park Y, Lee S, Park J. AMPK inhibitor, compound C, inhibits coronavirus replication in vitro. PLoS One 2023; 18:e0292309. [PMID: 37788269 PMCID: PMC10547180 DOI: 10.1371/journal.pone.0292309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 09/18/2023] [Indexed: 10/05/2023] Open
Abstract
The coronavirus disease (COVID-19) pandemic has resulted in more than six million deaths by October 2022. Vaccines and antivirals for severe acute respiratory syndrome coronavirus 2 are now available; however, more effective antiviral drugs are required for effective treatment. Here, we report that a potent AMP-activated protein kinase (AMPK) inhibitor, compound C/dorsomorphin, inhibits the replication of the human coronavirus OC43 strain (HCoV-OC43). We examined HCoV-OC43 replication in control and AMPK-knockout (KO) cells and found that the virus replication decreased in AMPK-KO cells. Next, we examined the effect of the AMPK inhibitor, compound C on coronavirus replication. Compound C treatment efficiently inhibited the replication and decreased the coronavirus-induced cytotoxicity, further inhibiting autophagy. In addition, treatment with compound C in combination with chloroquine synergistically inhibited coronavirus replication. These results suggest that compound C can be considered as a potential drug candidate for COVID-19.
Collapse
Affiliation(s)
- Minsu Jang
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Rackhyun Park
- Department of Life Science, Yong-In University, Yongin, Republic of Korea
| | - Ayane Yamamoto
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Yea-In Park
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Yeonjeong Park
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Siyun Lee
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Junsoo Park
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| |
Collapse
|
4
|
Lee YC, Chiou JT, Chang LS. AMPK inhibition induces MCL1 mRNA destabilization via the p38 MAPK/miR-22/HuR axis in chronic myeloid leukemia cells. Biochem Pharmacol 2023; 209:115442. [PMID: 36720359 DOI: 10.1016/j.bcp.2023.115442] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/15/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023]
Abstract
The oncogenic and tumor-suppressive roles of AMPK in chronic myeloid leukemia (CML) are controvertible. This study aimed to investigate the cytotoxic effects of the AMPK inhibitor Compound C in the CML cell lines K562, KU812, and MEG-01. Compared to K562 cells, KU812 and MEG-01 cells were more sensitive to Compound C-mediated cytotoxicity. Moreover, Compound C induced SIRT3 upregulation in K562 cells but not in KU812 or MEG-01 cells. SIRT3 silencing increased the sensitivity of K562 cells to Compound C. Additionally; Compound C-induced autophagy attenuated its induced apoptosis in KU812 and MEG-01 cells. Compound C-induced ROS-mediated AMPKα inactivation resulted in the downregulation of apoptotic regulator MCL1 in KU812 and MEG-01 cells. Mechanistically, AMPK inhibition activated p38 MAPK-mediated miR-22 expression, which in turn inhibited HuR expression, thereby reducing MCL1 mRNA stability. Overexpression of constitutively active AMPKα1 and abolishment of the activation of p38 MAPK inhibited Compound C-induced cell death and MCL1 downregulation. Furthermore, Compound C synergistically enhanced the cytotoxicity of BCR-ABL inhibitors and the BCL2 inhibitor ABT-199. Collectively, this study indicates that Compound C induces MCL1 downregulation through the AMPK/p38 MAPK/miR-22/HuR pathway, thereby inducing apoptosis of KU812 and MEG-01 cells. Furthermore, our findings suggest that AMPK inhibition is a promising strategy for improving CML therapy.
Collapse
Affiliation(s)
- Yuan-Chin Lee
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Jing-Ting Chiou
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
5
|
Liu Z, Shan S, Yuan Z, Wu F, Zheng M, Wang Y, Gui J, Xu W, Wang C, Ren T, Wen Z. Mitophagy bridges DNA sensing with metabolic adaption to expand lung cancer stem-like cells. EMBO Rep 2023; 24:e54006. [PMID: 36416244 PMCID: PMC9900345 DOI: 10.15252/embr.202154006] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/01/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
While previous studies have identified cancer stem-like cells (CSCs) as a crucial driver for chemoresistance and tumor recurrence, the underlying mechanisms for populating the CSC pool remain unclear. Here, we identify hypermitophagy as a feature of human lung CSCs, promoting metabolic adaption via the Notch1-AMPK axis to drive CSC expansion. Specifically, mitophagy is highly active in CSCs, resulting in increased mitochondrial DNA (mtDNA) content in the lysosome. Lysosomal mtDNA acts as an endogenous ligand for Toll-like receptor 9 (TLR9) that promotes Notch1 activity. Notch1 interacts with AMPK to drive lysosomal AMPK activation by inducing metabolic stress and LKB1 phosphorylation. This TLR9-Notch1-AMPK axis supports mitochondrial metabolism to fuel CSC expansion. In patient-derived xenograft chimeras, targeting mitophagy and TLR9-dependent Notch1-AMPK pathway restricts tumor growth and CSC expansion. Taken together, mitochondrial hemostasis is interlinked with innate immune sensing and Notch1-AMPK activity to increase the CSC pool of human lung cancer.
Collapse
Affiliation(s)
- Zhen Liu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| | - Shan Shan
- Department of Respiratory MedicineShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zixin Yuan
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| | - Fengying Wu
- Department of Medical Oncology, Shanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Ming Zheng
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| | - Ying Wang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| | - Jun Gui
- State Key Laboratory of Oncogenes and Related Genes; Renji‐Med X Clinical Stem Cell Research Center, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wei Xu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| | - Chunhong Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology of Jiangsu Province, State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouChina
| | - Tao Ren
- Department of Respiratory MedicineShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Kay Laboratory of Sleep Disordered BreathingShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhenke Wen
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| |
Collapse
|
6
|
Hasei S, Yamamotoya T, Nakatsu Y, Ohata Y, Itoga S, Nonaka Y, Matsunaga Y, Sakoda H, Fujishiro M, Kushiyama A, Asano T. Carnosic Acid and Carnosol Activate AMPK, Suppress Expressions of Gluconeogenic and Lipogenic Genes, and Inhibit Proliferation of HepG2 Cells. Int J Mol Sci 2021; 22:ijms22084040. [PMID: 33919842 PMCID: PMC8070802 DOI: 10.3390/ijms22084040] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/25/2022] Open
Abstract
Carnosic acid (CA), carnosol (CL) and rosmarinic acid (RA), components of the herb rosemary, reportedly exert favorable metabolic actions. This study showed that both CA and CL, but not RA, induce significant phosphorylation of AMP-dependent kinase (AMPK) and its downstream acetyl-CoA carboxylase 1 (ACC1) in HepG2 hepatoma cells. Glucose-6-phosphatase (G6PC) and phosphoenolpyruvate carboxykinase 1 (PCK1), rate-limiting enzymes of hepatic gluconeogenesis, are upregulated by forskolin stimulation, and this upregulation was suppressed when incubated with CA or CL. Similarly, a forskolin-induced increase in CRE transcriptional activity involved in G6PC and PCK1 regulations was also stymied when incubated with CA or CL. In addition, mRNA levels of ACC1, fatty acid synthase (FAS) and sterol regulatory element-binding protein 1c (SREBP-1c) were significantly reduced when incubated with CA or CL. Finally, it was shown that CA and CL suppressed cell proliferation and reduced cell viability, possibly as a result of AMPK activation. These findings raise the possibility that CA and CL exert a protective effect against diabetes and fatty liver disease, as well as subsequent cases of hepatoma.
Collapse
Affiliation(s)
- Shun Hasei
- Department of Medical Chemistry, Division of Molecular Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan; (S.H.); (T.Y.); (Y.N.); (Y.O.)
| | - Takeshi Yamamotoya
- Department of Medical Chemistry, Division of Molecular Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan; (S.H.); (T.Y.); (Y.N.); (Y.O.)
| | - Yusuke Nakatsu
- Department of Medical Chemistry, Division of Molecular Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan; (S.H.); (T.Y.); (Y.N.); (Y.O.)
| | - Yukino Ohata
- Department of Medical Chemistry, Division of Molecular Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan; (S.H.); (T.Y.); (Y.N.); (Y.O.)
| | - Shota Itoga
- Research Institute, Suntory Global Innovation Center Limited, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan; (S.I.); (Y.N.)
| | - Yuji Nonaka
- Research Institute, Suntory Global Innovation Center Limited, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan; (S.I.); (Y.N.)
| | - Yasuka Matsunaga
- Center for Translational Research in Infection & Inflammation, School of Medicine, Tulane University, 6823 St. Charles Avenue, New Orleans, LA 70118, USA;
| | - Hideyuki Sakoda
- Department of Internal Medicine, Division of Neurology, Respirology, Endocrinology and Metabolism, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan;
| | - Midori Fujishiro
- Department of Internal Medicine, Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, Tokyo 173-8610, Japan;
| | - Akifumi Kushiyama
- Department of Pharmacotherapy, Meiji Pharmaceutical University, Kiyose City, Tokyo 204-8588, Japan;
| | - Tomoichiro Asano
- Department of Medical Chemistry, Division of Molecular Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan; (S.H.); (T.Y.); (Y.N.); (Y.O.)
- Correspondence:
| |
Collapse
|
7
|
Li F, Sun A, Cheng G, Liu D, Xiao J, Zhao Z, Dong Z. Compound C Protects Against Cisplatin-Induced Nephrotoxicity Through Pleiotropic Effects. Front Physiol 2021; 11:614244. [PMID: 33424637 PMCID: PMC7785967 DOI: 10.3389/fphys.2020.614244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/03/2020] [Indexed: 12/17/2022] Open
Abstract
AICAR (Acadesine/AICA riboside) as an activator of AMPK, can protect renal tubular cells from cisplatin induced apoptosis. But in our experiment, the dorsomorphin (compound C, an inhibitor of AMPK) also significantly reduced cisplatin induced renal tubular cells apoptosis. Accordingly, we tested whether compound C can protect cisplatin-induced nephrotoxicity and the specific mechanism. Here, we treated Boston University mouse proximal tubular cells (BUMPT-306) with cisplatin and/or different dosages of AICAR (Acadesine/AICA riboside) or compound C to confirm the effect of AICAR and compound C in vitro. The AMPK-siRNA treated cells to evaluate whether the protective effect of compound C was through inhibiting AMPK. Male C57BL/6 mice were used to verify the effect of compound C in vivo. Both compound C and AICAR can reduce renal tubular cells apoptosis in dose-dependent manners, and compound C decreased serum creatinine and renal tubular injury induced by cisplatin. Mechanistically, compound C inhibited P53, CHOP and p-IREα during cisplatin treatment. Our results demonstrated that compound C inhibited AMPK, but the renal protective effects of compound C were not through AMPK. Instead, compound C protected cisplatin nephrotoxicity by inhibiting P53 and endoplasmic reticulum (ER) stress. Therefore, compound C may protect against cisplatin-induced nephrotoxicity through pleiotropic effects.
Collapse
Affiliation(s)
- Fanghua Li
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Anbang Sun
- Department of Anatomy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Genyang Cheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dong Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Xiao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhanzheng Zhao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
8
|
Synthesis and biological evaluation of novel pyrazolo[1,5-a]pyrimidines: Discovery of a selective inhibitor of JAK1 JH2 pseudokinase and VPS34. Bioorg Med Chem Lett 2020; 30:126813. [DOI: 10.1016/j.bmcl.2019.126813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/01/2019] [Accepted: 11/06/2019] [Indexed: 12/15/2022]
|
9
|
Chakraborty S, Datta S, Ghosh S. Induction of autophagy under nitrosative stress: A complex regulatory interplay between SIRT1 and AMPK in MCF7 cells. Cell Signal 2019; 64:109411. [PMID: 31491460 DOI: 10.1016/j.cellsig.2019.109411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 08/30/2019] [Accepted: 08/31/2019] [Indexed: 12/18/2022]
Abstract
Induction of nitrosative stress has been observed in various cancer types and in tumor environment. However, it is still unclear how cancer cells combat the effect of nitrosative stress. The main targets of nitrosative stress in cells are cellular lipids, proteins and DNA. Autophagy or self-cleaning generates energy for cell survival under stress conditions. In the present study we investigated the role of autophagy under nitrosative stress in MCF7, a breast cancer cell line. Interestingly, we observed induction of autophagy associated with cell death when MCF7 cells were treated with NO donor compound DETA-NONOate for eight hours. While investigating the mode of cell death under nitrosative stress in MCF7 cells, it was found that it was neither apoptotic nor necrotic. Moreover, nitrosative stress did not alter mitochondrial membrane potential and cellular redox status in MCF7 cells. But we observed an increase in NAD+/NADH and a drop in NADH level in MCF7 cells following NO donor treatment. Sirtuins having NAD+ dependent deacetylase activity, play an important role in cell survival mechanisms. So we further checked the status of SIRT1 under nitrosative stress in MCF7 cells. Surprisingly, we observed an induction of SIRT1, phospho-AMPK and p53 in MCF7 cells under nitrosative stress. Interestingly, autophagy markers were down regulated in MCF7 cells upon treatment with nicotinamide, an inhibitor of SIRT1 activity and dorsomorphin, a phospho-AMPK inhibitor when treated separately under nitrosative stress. To further confirm the role of SIRT1 in the induction of autophagy associated cell death, it was knocked down using si-RNA and nitrosative stress was applied. SIRT1 knock down led to increase in MCF7 cell viability along with down regulation of autophagic markers and phospho-AMPK as well as accumulation of acetylated p53. The increase in p53 controlled DRAM1 mRNA expression in MCF7 cells under nitrosative stress further confirmed a complex interplay between p53, SIRT1 and AMPK under nitrosative stress in MCF7 cells. Altogether our work for the first time suggests a complex inter-twined partnership between AMPK, SIRT1 and p53 in regulating autophagy in response to nitrosative stress in MCF7 cells.
Collapse
Affiliation(s)
- Subhamoy Chakraborty
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Sampurna Datta
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Sanjay Ghosh
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| |
Collapse
|
10
|
Li N, Wang T, Li Z, Ye X, Deng B, Zhuo S, Yao P, Yang M, Mei H, Chen X, Zhu T, Chen S, Wang H, Wang J, Le Y. Dorsomorphin induces cancer cell apoptosis and sensitizes cancer cells to HSP90 and proteasome inhibitors by reducing nuclear heat shock factor 1 levels. Cancer Biol Med 2019; 16:220-233. [PMID: 31516744 PMCID: PMC6713636 DOI: 10.20892/j.issn.2095-3941.2018.0235] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective Heat shock factor 1 (HSF1), a transcriptional regulator of heat shock proteins (HSPs), is an attractive therapeutic target for cancer. However, only a few HSF1 inhibitors have been identified so far. Methods The mRNA and protein levels of HSF1, HSPs, cleaved PARP, and phosphorylated HSF1 were examined by real-time PCR and Western blot. Forced expression, RNA interference, and immunofluorescence assay were used for mechanistic studies. Cell viability and apoptosis were measured by WST-8 assay and flow cytometry, respectively. Xenograft studies were performed in nude mice to evaluate the effect of dorsomorphin and an HSP90 inhibitor on tumor growth. Results Dorsomorphin suppressed multiple stimuli-induced and constitutive HSPs expression in cancer cells. Mechanistic studies revealed that dorsomorphin reduced heat-induced HSP expression independent of adenosine monophosphate activated protein kinase. Dorsomorphin reduced heat-stimulated HSF1 Ser320 phosphorylation and nuclear translocation, as well as resting nuclear HSF1 levels in cancer cells. Dorsomorphin induced cancer cell apoptosis by inhibiting HSF1 expression. A structure-activity study revealed that the 4-pyridyl at the 3-site of the pyrazolo [1, 5-a]pyrimidine ring is critical for the anti-HSF1 activities of dorsomorphin. Dorsomorphin sensitized cancer cells to HSP90 and proteasome inhibitors and inhibited HSP70 expression induced by these inhibitors in vitro. In tumor-bearing nude mice, dorsomorphin enhanced HSP90 inhibitor-induced cancer cell apoptosis, tumor growth inhibition, and HSP70 expression.
Conclusions Dorsomorphin is an HSF1 inhibitor. It induces cancer cell apoptosis, sensitizes cancer cells to both HSP90 and proteasome inhibitors, and suppresses HSP upregulation by these drugs, which may prevent the development of drug resistance. Hence, dorsomorphin and its derivates may serve as potential precursors for developing drugs against cancer.
Collapse
Affiliation(s)
- Na Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ting Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zongmeng Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoli Ye
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bo Deng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shu Zhuo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Pengle Yao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mengmei Yang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hong Mei
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaofang Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tengfei Zhu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shiting Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing 100022, China
| | - Jiming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick 21702, MD, USA
| | - Yingying Le
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing 100022, China
| |
Collapse
|
11
|
Cheruiyot A, Li S, Nickless A, Roth R, Fitzpatrick JAJ, You Z. Compound C inhibits nonsense-mediated RNA decay independently of AMPK. PLoS One 2018; 13:e0204978. [PMID: 30289931 PMCID: PMC6173407 DOI: 10.1371/journal.pone.0204978] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 09/18/2018] [Indexed: 12/26/2022] Open
Abstract
The nonsense mediated RNA decay (NMD) pathway safeguards the integrity of the transcriptome by targeting mRNAs with premature translation termination codons (PTCs) for degradation. It also regulates gene expression by degrading a large number of non-mutant RNAs (including mRNAs and noncoding RNAs) that bear NMD-inducing features. Consequently, NMD has been shown to influence development, cellular response to stress, and clinical outcome of many genetic diseases. Small molecules that can modulate NMD activity provide critical tools for understanding the mechanism and physiological functions of NMD, and they also offer potential means for treating certain genetic diseases and cancer. Therefore, there is an intense interest in identifying small-molecule NMD inhibitors or enhancers. It was previously reported that both inhibition of NMD and treatment with the AMPK-selective inhibitor Compound C (CC) induce autophagy in human cells, raising the possibility that CC may be capable of inhibiting NMD. Here we show that CC indeed has a NMD-inhibitory activity. Inhibition of NMD by CC is, however, independent of AMPK activity. As a competitive ATP analog, CC does not affect the kinase activity of SMG1, an essential NMD factor and the only known kinase in the NMD pathway. However, CC treatment down-regulates the protein levels of several NMD factors. The induction of autophagy by CC treatment is independent of ATF4, a NMD target that has been shown to promote autophagy in response to NMD inhibition. Our results reveal a new activity of CC as a NMD inhibitor, which has implications for its use in basic research and drug development.
Collapse
Affiliation(s)
- Abigael Cheruiyot
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Shan Li
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Andrew Nickless
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Robyn Roth
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - James A. J. Fitzpatrick
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Biomedical Engineering Washington University, St. Louis, Missouri, United States of America
- Center for Cellular Imaging, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Zhongsheng You
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
12
|
Abstract
While cancer cell proliferation depends on access to extracellular nutrients, inadequate tumour perfusion means that glucose, amino acids and lipids are often in short supply. To overcome this obstacle to growth, cancer cells utilize multiple scavenging strategies, obtaining macromolecules from the microenvironment and breaking them down in the lysosome to produce substrates for ATP generation and anabolism. Recent studies have revealed four scavenging pathways that support cancer cell proliferation in low-nutrient environments: scavenging of extracellular matrix proteins via integrins, receptor-mediated albumin uptake and catabolism, macropinocytic consumption of multiple components of the tumour microenvironment and the engulfment and degradation of entire live cells via entosis. New evidence suggests that blocking these pathways alone or in combination could provide substantial benefits to patients with incurable solid tumours. Both US Food and Drug Administration (FDA)-approved drugs and several agents in preclinical or clinical development shut down individual or multiple scavenging pathways. These therapies may increase the extent and durability of tumour growth inhibition and/or prevent the development of resistance when used in combination with existing treatments. This Review summarizes the evidence suggesting that scavenging pathways drive tumour growth, highlights recent advances that define the oncogenic signal transduction pathways that regulate scavenging and considers the benefits and detriments of therapeutic strategies targeting scavenging that are currently under development.
Collapse
Affiliation(s)
- Brendan T Finicle
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Vaishali Jayashankar
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Aimee L Edinger
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
13
|
Zhao X, Luo G, Cheng Y, Yu W, Chen R, Xiao B, Xiang Y, Feng C, Fu W, Duan C, Yao F, Xia X, Tao Q, Wei M, Dai R. Compound C induces protective autophagy in human cholangiocarcinoma cells via Akt/mTOR‐independent pathway. J Cell Biochem 2018; 119:5538-5550. [DOI: 10.1002/jcb.26723] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/24/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Xiaofang Zhao
- Liver Diseases LaboratorySouthwest Medical UniversityLuzhouSichuanChina
- Department of Biochemistry and Molecular BiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Guosong Luo
- Liver Diseases LaboratorySouthwest Medical UniversityLuzhouSichuanChina
- Department of Hepatobiliary Surgery of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuanChina
| | - Ying Cheng
- Liver Diseases LaboratorySouthwest Medical UniversityLuzhouSichuanChina
- Department of Biochemistry and Molecular BiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Wenjing Yu
- Department of Biochemistry and Molecular BiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Run Chen
- Department of Public HealthSouthwest Medical UniversityLuzhouSichuanChina
| | - Bin Xiao
- Department of Biochemistry and Molecular BiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Yuancai Xiang
- Liver Diseases LaboratorySouthwest Medical UniversityLuzhouSichuanChina
| | - Chunhong Feng
- Department of Hepatobiliary Surgery of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuanChina
| | - Wenguang Fu
- Department of Hepatobiliary Surgery of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuanChina
| | - Chunyan Duan
- Department of Biochemistry and Molecular BiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Fuli Yao
- Department of Biochemistry and Molecular BiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Xianming Xia
- Department of Hepatobiliary Surgery of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuanChina
| | - Qinghua Tao
- MOE Key Laboratory of Protein SciencesTsinghua University School of Life SciencesBeijingChina
| | - Mei Wei
- Department of Liver Diseases of the Affiliated Hospital of Chinese Traditional MedicineSouthwest Medical UniversityLuzhouSichuanChina
| | - Rongyang Dai
- Liver Diseases LaboratorySouthwest Medical UniversityLuzhouSichuanChina
- Department of Liver Diseases of the Affiliated Hospital of Chinese Traditional MedicineSouthwest Medical UniversityLuzhouSichuanChina
| |
Collapse
|
14
|
Abstract
The evolutionary conserved energy sensor AMPK plays crucial roles in many biological processes-both during normal development and pathology. Loss-of-function genetic studies in mice as well as in lower organisms underscore its importance in embryonic development, stress physiology in the adult, and in key metabolic disorders including cardiovascular disease, diabetes, cancer, and metabolic syndrome. In contrast to several other kinases important in human health and medicine where specific/selective inhibitors are available, no AMPK-specific inhibitors are available. The only reagent called dorsomorphin or compound C that is occasionally used as an AMPK inhibitor unfortunately inhibits several other kinases much more potently than AMPK and is therefore highly non-specific. In this chapter, we discuss the pros and cons of using this reagent to study AMPK functions.
Collapse
Affiliation(s)
- Biplab Dasgupta
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - William Seibel
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
15
|
Lu X, Guo S, Cheng Y, Kim JH, Feng Y, Feng Y. Stimulation of ovarian follicle growth after AMPK inhibition. Reproduction 2017; 153:683-694. [DOI: 10.1530/rep-16-0577] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 01/31/2017] [Accepted: 02/28/2017] [Indexed: 12/20/2022]
Abstract
Previous studies showed that the protein kinase B (Akt)–mammalian target of rapamycin (mTOR) and Hippo signaling Yes-associated protein (YAP) pathways play important roles in promoting follicle growth. Additionally, other studies demonstrated that 5′ adenosine monophosphate-activated protein kinase (AMPK) is an upstream regulatory element of mTOR and YAP. Here, we used AMPK inhibitor (Compound C) toin vitrocultured ovaries from 10-day-old mice followed byin vivografting into adult hosts or toin situtreated ovaries of 3-week-old mice by intrabursal injection followed by gonadotropin stimulation. We found that the phosphorylation of ovarian mTOR and downstream proteins (ribosomal protein S6 (S6) and eukaryotic translation initiation factor 4B (eIF4B)) was upregulated following Compound C administration, whereas tuberous sclerosis complex 2 (TSC2) phosphorylation was downregulated. Additionally, treatment with Compound C increased hypoxia-inducible factor 1-alpha (Hif1a), vascular endothelial growth factor A (Vegfa), VEGF receptor 2 (Vegfr2) and connective tissue growth factor (Ctgf) mRNA levels. Furthermore, treatment of 10-day-old mice with Compound C promoted the growth of preantral and antral follicles accompanied by enhanced angiogenesis.In situintrabursal injection with Compound C, followed by controlled ovarian hyperstimulation, increased the number of ovulated oocytes in 3-week-old mice, and these oocytes could be successfully fertilized, leading to the delivery of healthy pups. Our results demonstrated that treatment with AMPK inhibitor resulted in the activation of the mTOR signaling pathway, increases inCtgfexpression in mouse ovaries, stimulation of follicle development and promotion of ovarian angiogenesis for ovary growth.
Collapse
|
16
|
Zhao X, Yang Y, Yao F, Xiao B, Cheng Y, Feng C, Duan C, Zhang C, Liu Y, Li H, Xiao B, Dai R. Unfolded Protein Response Promotes Doxorubicin-Induced Nonsmall Cell Lung Cancer Cells Apoptosis via the mTOR Pathway Inhibition. Cancer Biother Radiopharm 2016; 31:347-351. [DOI: 10.1089/cbr.2016.2079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Xiaofang Zhao
- Department of Biochemistry and Molecular Biology, Southwest Medical University, Luzhou, China
| | - Yan Yang
- Department of Public Health, Southwest Medical University, Luzhou, China
| | - Fuli Yao
- Department of Biochemistry and Molecular Biology, Southwest Medical University, Luzhou, China
| | - Bin Xiao
- Department of Biochemistry and Molecular Biology, Southwest Medical University, Luzhou, China
| | - Ying Cheng
- Department of Biochemistry and Molecular Biology, Southwest Medical University, Luzhou, China
| | - Chunhong Feng
- Department of Hepatobiliary Surgery of the Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Chunyan Duan
- Department of Biochemistry and Molecular Biology, Southwest Medical University, Luzhou, China
| | - Chunyan Zhang
- Department of Biochemistry and Molecular Biology, Southwest Medical University, Luzhou, China
| | - Youping Liu
- Department of Biochemistry and Molecular Biology, Southwest Medical University, Luzhou, China
| | - Hong Li
- Department of Biochemistry and Molecular Biology, Southwest Medical University, Luzhou, China
| | - Bo Xiao
- Department of Thoracic Surgery, Sichuan Cancer Hospital, Chengdu, China
| | - Rongyang Dai
- Department of Biochemistry and Molecular Biology, Southwest Medical University, Luzhou, China
| |
Collapse
|
17
|
Metformin and AICAR regulate NANOG expression via the JNK pathway in HepG2 cells independently of AMPK. Tumour Biol 2016; 37:11199-208. [PMID: 26939902 DOI: 10.1007/s13277-016-5007-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 02/25/2016] [Indexed: 12/21/2022] Open
Abstract
NANOG, a marker of stemness, impacts tumor progression and therapeutic resistance in cancer cells. In human hepatocellular carcinoma (HCC), upregulation of NANOG is associated with metastasis and a low survival rate, while its downregulation results in a lower colony formation rate and enhanced chemosensitivity. Metformin, an agent widely used for diabetes treatment, and AICAR, another AMP-activated protein kinase (AMPK) activator, have been reported to inhibit the growth of several types of cancer. Although inhibitory effects of metformin on NANOG in pancreatic cancer cells and of AICAR in mouse embryonic stem cells have been described, the underlying molecular mechanisms remain uncertain in HCC. In this study, we used the HepG2 cell line and found that metformin/AICAR downregulated NANOG expression with decreased cell viability and enhanced chemosensitivity to 5-fluorouracil (5-FU). Moreover, metformin/AICAR inhibited c-Jun N-terminal kinase (JNK) activity, and blockade of either the JNK MAPK pathway or knockdown of JNK1 gene expression reduced NANOG levels. The upregulation of NANOG and phospho-JNK by basic fibroblast growth factor (bFGF) was abrogated by metformin/AICAR. Additionally, although transient upregulation of NANOG within 2 h of treatment with metformin/AICAR was concordant with both JNK and AMPK activation, increased NANOG expression with activation of JNK was also observed following AMPK inhibition with compound C. Taken together, our data suggest that metformin/AICAR regulate NANOG expression via the JNK MAPK pathway in HepG2 cells independently of AMPK, and that this JNK/NANOG signaling pathway may offer new therapeutic strategies for the treatment of HCC.
Collapse
|
18
|
Oosterman JE, Belsham DD. Glucose Alters Per2 Rhythmicity Independent of AMPK, Whereas AMPK Inhibitor Compound C Causes Profound Repression of Clock Genes and AgRP in mHypoE-37 Hypothalamic Neurons. PLoS One 2016; 11:e0146969. [PMID: 26784927 PMCID: PMC4718556 DOI: 10.1371/journal.pone.0146969] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/23/2015] [Indexed: 01/11/2023] Open
Abstract
Specific neurons in the hypothalamus are regulated by peripheral hormones and nutrients to maintain proper metabolic control. It is unclear if nutrients can directly control clock gene expression. We have therefore utilized the immortalized, hypothalamic cell line mHypoE-37, which exhibits robust circadian rhythms of core clock genes. mHypoE-37 neurons were exposed to 0.5 or 5.5 mM glucose, comparable to physiological levels in the brain. Per2 and Bmal1 mRNAs were assessed every 3 hours over 36 hours. Incubation with 5.5 mM glucose significantly shortened the period and delayed the phase of Per2 mRNA levels, but had no effect on Bmal1. Glucose had no significant effect on phospho-GSK3β, whereas AMPK phosphorylation was altered. Thus, the AMPK inhibitor Compound C was utilized, and mRNA levels of Per2, Bmal1, Cryptochrome1 (Cry1), agouti-related peptide (AgRP), carnitine palmitoyltransferase 1C (Cpt1c), and O-linked N-acetylglucosamine transferase (Ogt) were measured. Remarkably, Compound C dramatically reduced transcript levels of Per2, Bmal1, Cry1, and AgRP, but not Cpt1c or Ogt. Because AMPK was not inhibited at the same time or concentrations as the clock genes, we suggest that the effect of Compound C on gene expression occurs through an AMPK-independent mechanism. The consequences of inhibition of the rhythmic expression of clock genes, and in turn downstream metabolic mediators, such as AgRP, could have detrimental effects on overall metabolic processes. Importantly, the effects of the most commonly used AMPK inhibitor Compound C should be interpreted with caution, considering its role in AMPK-independent repression of specific genes, and especially clock gene rhythm dysregulation.
Collapse
Affiliation(s)
- Johanneke E. Oosterman
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Denise D. Belsham
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Cellular and Molecular Biology, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
19
|
Pros and cons of pDNA and mRNA transfection to study mRNA translation in mammalian cells. Gene 2015; 578:1-6. [PMID: 26680098 DOI: 10.1016/j.gene.2015.12.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 12/07/2015] [Indexed: 02/02/2023]
Abstract
Protein synthesis in eukaryotes is subject to stringent control. The misregulation of translation of certain mRNAs is often a hallmark of many diseases, including malignancies and autoimmune disorders. To understand why and how it happens, it is important to investigate the translational control of specific mRNAs. In this case, one could use reporter mRNAs in order to identify cis-acting elements responsible for regulation. Here we overview plasmid DNA (pDNA) and mRNA transfections, their pitfalls and limitations, as well as some emerging applications for mRNA transfection.
Collapse
|
20
|
EIF2A-dependent translational arrest protects leukemia cells from the energetic stress induced by NAMPT inhibition. BMC Cancer 2015; 15:855. [PMID: 26542945 PMCID: PMC4636066 DOI: 10.1186/s12885-015-1845-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 10/23/2015] [Indexed: 01/04/2023] Open
Abstract
Background Nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in NAD+ biosynthesis from nicotinamide, is one of the major factors regulating cancer cells metabolism and is considered a promising target for treating cancer. The prototypical NAMPT inhibitor FK866 effectively lowers NAD+ levels in cancer cells, reducing the activity of NAD+-dependent enzymes, lowering intracellular ATP, and promoting cell death. Results We show that FK866 induces a translational arrest in leukemia cells through inhibition of MTOR/4EBP1 signaling and of the initiation factors EIF4E and EIF2A. Specifically, treatment with FK866 is shown to induce 5′AMP-activated protein kinase (AMPK) activation, which, together with EIF2A phosphorylation, is responsible for the inhibition of protein synthesis. Notably, such an effect was also observed in patients’ derived primary leukemia cells including T-cell Acute Lymphoblastic Leukemia. Jurkat cells in which AMPK or LKB1 expression was silenced or in which a non-phosphorylatable EIF2A mutant was ectopically expressed showed enhanced sensitivity to the NAMPT inhibitor, confirming a key role for the LKB1-AMPK-EIF2A axis in cell fate determination in response to energetic stress via NAD+ depletion. Conclusions We identified EIF2A phosphorylation as a novel early molecular event occurring in response to NAMPT inhibition and mediating protein synthesis arrest. In addition, our data suggest that tumors exhibiting an impaired LBK1- AMPK- EIF2A response may be especially susceptible to NAMPT inhibitors and thus become an elective indication for this type of agents. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1845-1) contains supplementary material, which is available to authorized users.
Collapse
|
21
|
Berberine Regulated Lipid Metabolism in the Presence of C75, Compound C, and TOFA in Breast Cancer Cell Line MCF-7. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:396035. [PMID: 26351511 PMCID: PMC4550799 DOI: 10.1155/2015/396035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 05/04/2015] [Accepted: 05/07/2015] [Indexed: 12/21/2022]
Abstract
Berberine interfering with cancer reprogramming metabolism was confirmed in our previous study. Lipid metabolism and mitochondrial function were also the core parts in reprogramming metabolism. In the presence of some energy-related inhibitors, including C75, compound C, and TOFA, the discrete roles of berberine in lipid metabolism and mitochondrial function were elucidated. An altered lipid metabolism induced by berberine was observed under the inhibition of FASN, AMPK, and ACC in breast cancer cell MCF-7. And the reversion of berberine-induced lipid suppression indicated that ACC inhibition might be involved in that process instead of FASN inhibition. A robust apoptosis induced by berberine even under the inhibition of AMPK and lipid synthesis was also indicated. Finally, mitochondrial function regulation under the inhibition of AMPK and ACC might be in an ACL-independent manner. Undoubtedly, the detailed mechanisms of berberine interfering with lipid metabolism and mitochondrial function combined with energy-related inhibitors need further investigation, including the potential compensatory mechanisms for ATP production and the upregulation of ACL.
Collapse
|
22
|
Ali JL, Lagasse BJ, Minuk AJ, Love AJ, Moraya AI, Lam L, Arthur G, Gibson SB, Morrison LC, Werbowetski-Ogilvie TE, Fu Y, Nachtigal MW. Differential cellular responses induced by dorsomorphin and LDN-193189 in chemotherapy-sensitive and chemotherapy-resistant human epithelial ovarian cancer cells. Int J Cancer 2014; 136:E455-69. [PMID: 25227893 DOI: 10.1002/ijc.29220] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 08/21/2014] [Accepted: 09/10/2014] [Indexed: 12/11/2022]
Abstract
Inherent or acquired drug resistance is a major contributor to epithelial ovarian cancer (EOC) mortality. Novel drugs or drug combinations that produce EOC cell death or resensitize drug resistant cells to standard chemotherapy may improve patient treatment. After conducting drug tolerability studies for the multikinase inhibitors dorsomorphin (DM) and it is structural analogue LDN-193189 (LDN), these drugs were tested in a mouse intraperitoneal xenograft model of EOC. DM significantly increased survival, whereas LDN showed a trend toward increased survival. In vitro experiments using cisplatin (CP)-resistant EOC cell lines, A2780-cp or SKOV3, we determined that pretreatment or cotreatment with DM or LDN resensitized cells to the killing effect of CP or carboplatin (CB). DM was capable of blocking EOC cell cycle and migration, whereas LDN produced a less pronounced effect on cell cycle and no effect on migration. Subsequent analyses using primary human EOC cell samples or additional established EOC cells lines showed that DM or LDN induced a dose-dependent autophagic or cell death response, respectively. DM induced a characteristic morphological change with the appearance of numerous LC3B-containing acidic vacuoles and an increase in LC3BII levels. This was coincident with a decrease in cell growth and the altered cell cycle consistent with DM-induced cytostasis. By contrast, LDN produced a caspase 3-independent, reactive oxygen species-dependent cell death. Overall, DM and LDN possess drug characteristics suitable for adjuvant agents used to treat chemotherapy-sensitive and -resistant EOC.
Collapse
Affiliation(s)
- Jennifer L Ali
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|