1
|
Han C, Fu S, Chen M, Gou Y, Liu D, Zhang C, Huang X, Xiao L, Zhao M, Zhang J, Xiao Q, Peng D, Xue Y. GPSD: a hybrid learning framework for the prediction of phosphatase-specific dephosphorylation sites. Brief Bioinform 2024; 26:bbae694. [PMID: 39749667 PMCID: PMC11695897 DOI: 10.1093/bib/bbae694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/30/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Protein phosphorylation is dynamically and reversibly regulated by protein kinases and protein phosphatases, and plays an essential role in orchestrating a wide range of biological processes. Although a number of tools have been developed for predicting kinase-specific phosphorylation sites (p-sites), computational prediction of phosphatase-specific dephosphorylation sites remains to be a great challenge. In this study, we manually curated 4393 experimentally identified site-specific phosphatase-substrate relationships for 3463 dephosphorylation sites occurring on phosphoserine, phosphothreonine, and/or phosphotyrosine residues, from the literature and public databases. Then, we developed a hybrid learning framework, the group-based prediction system for the prediction of phosphatase-specific dephosphorylation sites (GPSD). For model training, we integrated 10 types of sequence features and utilized three types of machine learning methods, including penalized logistic regression, deep neural networks, and transformer neural networks. First, a pretrained model was constructed using 561 416 nonredundant p-sites and then fine-tuned to generate computational models for predicting general dephosphorylation sites. In addition, 103 individual phosphatase-specific predictors were constructed via transfer learning and meta-learning. For site prediction, one or multiple protein sequences in FASTA format could be inputted, and the prediction results will be shown together with additional annotations, such as protein-protein interactions, structural information, and disorder propensity. The online service of GPSD is freely available at https://gpsd.biocuckoo.cn/. We believe that GPSD can serve as a valuable tool for further analysis of dephosphorylation.
Collapse
Affiliation(s)
- Cheng Han
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Shanshan Fu
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Miaomiao Chen
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Yujie Gou
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Dan Liu
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Chi Zhang
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Xinhe Huang
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Leming Xiao
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Miaoying Zhao
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Jiayi Zhang
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Qiang Xiao
- School of Artificial Intelligence and Automation, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Di Peng
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Yu Xue
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| |
Collapse
|
2
|
Seyrek K, Ivanisenko NV, König C, Lavrik IN. Modulation of extrinsic apoptotic pathway by intracellular glycosylation. Trends Cell Biol 2024; 34:728-741. [PMID: 38336591 DOI: 10.1016/j.tcb.2024.01.003] [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: 09/20/2023] [Revised: 12/20/2023] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
The importance of post-translational modifications (PTMs), particularly O-GlcNAcylation, of cytoplasmic proteins in apoptosis has been neglected for quite a while. Modification of cytoplasmic proteins by a single N-acetylglucosamine sugar is a dynamic and reversible PTM exhibiting properties more like phosphorylation than classical O- and N-linked glycosylation. Due to the sparse information existing, we have only limited understanding of how GlcNAcylation affects cell death. Deciphering the role of GlcNAcylation in cell fate may provide further understanding of cell fate decisions. This review focus on the modulation of extrinsic apoptotic pathway via GlcNAcylation carried out by O-GlcNAc transferase (OGT) or by other bacterial effector proteins.
Collapse
Affiliation(s)
- Kamil Seyrek
- Translational Inflammation Research, Medical Faculty, Center of Dynamic Systems (CDS), Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Nikita V Ivanisenko
- Translational Inflammation Research, Medical Faculty, Center of Dynamic Systems (CDS), Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Corinna König
- Translational Inflammation Research, Medical Faculty, Center of Dynamic Systems (CDS), Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Inna N Lavrik
- Translational Inflammation Research, Medical Faculty, Center of Dynamic Systems (CDS), Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany.
| |
Collapse
|
3
|
Kang YL, Kim J, Kwak SB, Kim YS, Huh J, Park JW. The polyol pathway and nuclear ketohexokinase A signaling drive hyperglycemia-induced metastasis of gastric cancer. Exp Mol Med 2024; 56:220-234. [PMID: 38200154 PMCID: PMC10834943 DOI: 10.1038/s12276-023-01153-3] [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: 01/10/2023] [Revised: 10/20/2023] [Accepted: 11/07/2023] [Indexed: 01/12/2024] Open
Abstract
Diabetes might be associated with increased cancer risk, with several studies reporting hyperglycemia as a primary oncogenic stimulant. Since glucose metabolism is linked to numerous metabolic pathways, it is difficult to specify the mechanisms underlying hyperglycemia-induced cancer progression. Here, we focused on the polyol pathway, which is dramatically activated under hyperglycemia and causes diabetic complications. We investigated whether polyol pathway-derived fructose facilitates hyperglycemia-induced gastric cancer metastasis. We performed bioinformatics analysis of gastric cancer datasets and immunohistochemical analyses of gastric cancer specimens, followed by transcriptomic and proteomic analyses to evaluate phenotypic changes in gastric cancer cells. Consequently, we found a clinical association between the polyol pathway and gastric cancer progression. In gastric cancer cell lines, hyperglycemia enhanced cell migration and invasion, cytoskeletal rearrangement, and epithelial-mesenchymal transition (EMT). The hyperglycemia-induced acquisition of metastatic potential was mediated by increased fructose derived from the polyol pathway, which stimulated the nuclear ketohexokinase-A (KHK-A) signaling pathway, thereby inducing EMT by repressing the CDH1 gene. In two different xenograft models of cancer metastasis, gastric cancers overexpressing AKR1B1 were found to be highly metastatic in diabetic mice, but these effects of AKR1B1 were attenuated by KHK-A knockdown. In conclusion, hyperglycemia induces fructose formation through the polyol pathway, which in turn stimulates the KHK-A signaling pathway, driving gastric cancer metastasis by inducing EMT. Thus, the polyol and KHK-A signaling pathways could be potential therapeutic targets to decrease the metastatic risk in gastric cancer patients with diabetes.
Collapse
Affiliation(s)
- Ye-Lim Kang
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Jiyoung Kim
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Su-Bin Kwak
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Yi-Sook Kim
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - June Huh
- Department of Chemical and Biological Engineering, Korea University, Anam-ro, Seongbuk-gu, Seoul, 02841, Korea
| | - Jong-Wan Park
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| |
Collapse
|
4
|
Schofield JH, Schafer ZT. Regulators mount up: the metabolic roles of apoptotic proteins. FRONTIERS IN CELL DEATH 2023; 2:1223926. [PMID: 37521407 PMCID: PMC10373711 DOI: 10.3389/fceld.2023.1223926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
The induction of apoptosis, a programmed cell death pathway governed by activation of caspases, can result in fundamental changes in metabolism that either facilitate or restrict the execution of cell death. In addition, metabolic adaptations can significantly impact whether cells in fact initiate the apoptotic cascade. In this mini-review, we will highlight and discuss the interconnectedness of apoptotic regulation and metabolic alterations, two biological outcomes whose regulators are intertwined.
Collapse
Affiliation(s)
- James H. Schofield
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| | - Zachary T. Schafer
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| |
Collapse
|
5
|
Garufi A, D'Orazi V, Pistritto G, Cirone M, D'Orazi G. The Sweet Side of HIPK2. Cancers (Basel) 2023; 15:2678. [PMID: 37345014 DOI: 10.3390/cancers15102678] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 06/23/2023] Open
Abstract
HIPK2 is an evolutionary conserved protein kinase which modulates many molecular pathways involved in cellular functions such as apoptosis, DNA damage response, protein stability, and protein transcription. HIPK2 plays a key role in the cancer cell response to cytotoxic drugs as its deregulation impairs drug-induced cancer cell death. HIPK2 has also been involved in regulating fibrosis, angiogenesis, and neurological diseases. Recently, hyperglycemia was found to positively and/or negatively regulate HIPK2 activity, affecting not only cancer cell response to chemotherapy but also the progression of some diabetes complications. The present review will discuss how HIPK2 may be influenced by the high glucose (HG) metabolic condition and the consequences of such regulation in medical conditions.
Collapse
Affiliation(s)
- Alessia Garufi
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Valerio D'Orazi
- Department of Surgery, Sapienza University, 00185 Rome, Italy
| | - Giuseppa Pistritto
- Centralized Procedures Office, Italian Medicines Agency (AIFA), 00187 Rome, Italy
| | - Mara Cirone
- Laboratory Affiliated to Pasteur Institute Italy Foundation Cenci Bolognetti, Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Gabriella D'Orazi
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Department of Neurosciences, Imaging and Clinical Sciences, University "G. D'Annunzio", 66013 Chieti, Italy
| |
Collapse
|
6
|
NRF2 in Cancer: Cross-Talk with Oncogenic Pathways and Involvement in Gammaherpesvirus-Driven Carcinogenesis. Int J Mol Sci 2022; 24:ijms24010595. [PMID: 36614036 PMCID: PMC9820659 DOI: 10.3390/ijms24010595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
Expanding knowledge of the molecular mechanisms at the basis of tumor development, especially the cross-talk between oncogenic pathways, will possibly lead to better tailoring of anticancer therapies. Nuclear factor erythroid 2-related factor 2 (NRF2) plays a central role in cancer progression, not only because of its antioxidant activity but also because it establishes cross-talk with several oncogenic pathways, including Heat Shock Factor1 (HSF1), mammalian target of rapamycin (mTOR), and mutant (mut) p53. Moreover, the involvement of NRF2 in gammaherpesvirus-driven carcinogenesis is particularly interesting. These viruses indeed hijack the NRF2 pathway to sustain the survival of tumor cells in which they establish a latent infection and to avoid a too-high increase of reactive oxygen species (ROS) when these cancer cells undergo treatments that induce viral replication. Interestingly, NRF2 activation may prevent gammaherpesvirus-driven oncogenic transformation, highlighting how manipulating the NRF2 pathway in the different phases of gammaherpesvirus-mediated carcinogenesis may lead to different outcomes. This review will highlight the mechanistic interplay between NRF2 and some oncogenic pathways and its involvement in gammaherpesviruses biology to recapitulate published evidence useful for potential application in cancer therapy.
Collapse
|
7
|
Wang Y, Qu M, Qiu Z, Zhu S, Chen W, Guo K, Miao C, Zhang H. Surgical Stress and Cancer Progression: New Findings and Future Perspectives. Curr Oncol Rep 2022; 24:1501-1511. [PMID: 35763189 DOI: 10.1007/s11912-022-01298-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW The stress response to surgery is essential for maintaining homeostasis and exhibits anti-tumor effects; however, an ongoing and exaggerated stress response may have adverse clinical consequences and even promote cancer progression. This review will discuss the complex relationship between surgical stress and cancer progression. RECENT FINDINGS Surgical stress exhibits both anti-tumor and cancer-promoting effects by causing changes in the neuroendocrine, circulatory, and immune systems. Many studies have found that many mechanisms are involved in the process, and the corresponding targets could be applied for cancer therapy. Although surgical stress may have anti-tumor effects, it is necessary to inhibit an excessive stress response, mostly showing cancer-promoting effects.
Collapse
Affiliation(s)
- Yanghanzhao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Mengdi Qu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Zhiyun Qiu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Shuainan Zhu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Wankun Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Fudan University Jinshan Hospital, Shanghai, China.
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
8
|
Liu Y, Gu W. The complexity of p53-mediated metabolic regulation in tumor suppression. Semin Cancer Biol 2022; 85:4-32. [PMID: 33785447 PMCID: PMC8473587 DOI: 10.1016/j.semcancer.2021.03.010] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023]
Abstract
Although the classic activities of p53 including induction of cell-cycle arrest, senescence, and apoptosis are well accepted as critical barriers to cancer development, accumulating evidence suggests that loss of these classic activities is not sufficient to abrogate the tumor suppression activity of p53. Numerous studies suggest that metabolic regulation contributes to tumor suppression, but the mechanisms by which it does so are not completely understood. Cancer cells rewire cellular metabolism to meet the energetic and substrate demands of tumor development. It is well established that p53 suppresses glycolysis and promotes mitochondrial oxidative phosphorylation through a number of downstream targets against the Warburg effect. The role of p53-mediated metabolic regulation in tumor suppression is complexed by its function to promote both cell survival and cell death under different physiological settings. Indeed, p53 can regulate both pro-oxidant and antioxidant target genes for complete opposite effects. In this review, we will summarize the roles of p53 in the regulation of glucose, lipid, amino acid, nucleotide, iron metabolism, and ROS production. We will highlight the mechanisms underlying p53-mediated ferroptosis, AKT/mTOR signaling as well as autophagy and discuss the complexity of p53-metabolic regulation in tumor development.
Collapse
Affiliation(s)
- Yanqing Liu
- Institute for Cancer Genetics, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
| | - Wei Gu
- Institute for Cancer Genetics, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA.
| |
Collapse
|
9
|
The Impact of NRF2 Inhibition on Drug-Induced Colon Cancer Cell Death and p53 Activity: A Pilot Study. Biomolecules 2022; 12:biom12030461. [PMID: 35327653 PMCID: PMC8946796 DOI: 10.3390/biom12030461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/07/2022] [Accepted: 03/15/2022] [Indexed: 12/12/2022] Open
Abstract
Nuclear factor erythroid 2 (NF-E2) p45-related factor 2 (NRF2) protein is the master regulator of oxidative stress, which is at the basis of various chronic diseases including cancer. Hyperactivation of NRF2 in already established cancers can promote cell proliferation and resistance to therapies, such as in colorectal cancer (CRC), one of the most lethal and prevalent malignancies in industrialized countries with limited patient overall survival due to its escape mechanisms in both chemo- and targeted therapies. In this study, we generated stable NRF2 knockout colon cancer cells (NRF2-Cas9) to investigate the cell response to chemotherapeutic drugs with regard to p53 oncosuppressor, whose inhibition we previously showed to correlate with NRF2 pathway activation. Here, we found that NRF2 activation by sulforaphane (SFN) reduced cisplatin (CDDP)-induced cell death only in NRF2-proficient cells (NRF2-ctr) compared to NRF2-Cas9 cells. Mechanistically, we found that NRF2 activation protected NRF2-ctr cells from the drug-induced DNA damage and the apoptotic function of the unfolded protein response (UPR), in correlation with reduction of p53 activity, effects that were not observed in NRF2-Cas9 cells. Finally, we found that ZnCl2 supplementation rescued the cisplatin cytotoxic effects, as it impaired NRF2 activation, restoring p53 activity. These findings highlight NRF2′s key role in neutralizing the cytotoxic effects of chemotherapeutic drugs in correlation with reduced DNA damage and p53 activity. They also suggest that NRF2 inhibition could be a useful strategy for efficient anticancer chemotherapy and support the use of ZnCl2 to inhibit NRF2 pathway in combination therapies.
Collapse
|
10
|
Zhang Y, Cheng J, Li J, He J, Li X, Xu F. The GLP-1R Agonist Exendin-4 Attenuates Hyperglycemia-Induced Chemoresistance in Human Endometrial Cancer Cells Through ROS-Mediated Mitochondrial Pathway. Front Oncol 2022; 11:793530. [PMID: 34988025 PMCID: PMC8721044 DOI: 10.3389/fonc.2021.793530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/01/2021] [Indexed: 12/23/2022] Open
Abstract
This study aimed to assess the effects of the antidiabetic drug Exendin-4 (Exe-4), a GLP-1 receptor agonist, on the response of human endometrial cancer cells to chemotherapy under high glucose (HG) conditions. Cell viability was detected using a cell counting kit (CCK)-8. Cell apoptosis and reactive oxygen species (ROS) levels were measured by flow cytometry. Gene expression was evaluated by real-time PCR and immunoblotting. The chemotherapeutic drug cisplatin (DDP) dose-dependently inhibited both human endometrial adenocarcinoma Ishikawa and HEC1B cells, a response reversed by HG. Meanwhile, Exe-4 attenuated hyperglycemia’s effect by elevating intracellular lactate dehydrogenase (LDH) and ROS production. Similarly, DDP-induced elevation of intracellular rhodamine123 was attenuated by HG, and Exe-4 reversed HG’s impact. The chemoresistance genes multidrug resistance-associated protein 1 (MRP1) and P-glycoprotein (Pgp) were upregulated. At the same time, topoisomerase II (TOPO II) was downregulated under HG conditions, suggesting HG-induced chemoresistance. Exe-4 did not significantly influence the above genes. DDP downregulated Bcl-2 and Bcl-XL and upregulated Bax, cytosolic cytochrome c, and PARP under normal glucose (NG) versus HG conditions, and Exe-4 attenuated these effects. Upstream of Bax/Bcl, acetylated P53 was upregulated by DDP and downregulated by HG, whose effect was reversed by Exe-4. DPP treatment significantly induced apoptosis and cell cycle arrest in the S phase under NG, and HG reduced these effects. Prolonged exposure to HG induces DDP chemoresistance in human endometrial cancer cells but is alleviated by Exe-4.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Juan Cheng
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Jing Li
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Junxian He
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Xiaomao Li
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Fen Xu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| |
Collapse
|
11
|
Popescu-Vâlceanu HC, Stoicea MC, Enache V, Bratu RM, Mustăţea P, Drăguţ RM, Rusu E, Ionescu-Tîrgovişte C, Radulian G. Bcl-2 and p53 immunophenotypes in colorectal adenocarcinoma in type 2 diabetes mellitus versus non-diabetic patients. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2022; 63:521-528. [PMID: 36588490 PMCID: PMC9926153 DOI: 10.47162/rjme.63.3.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
We aimed to investigate immunohistochemical expression of the p53 tumor suppressor protein, and the B-cell lymphoma-2 (Bcl-2) apoptotic protein in colorectal adenocarcinoma patients with or without type 2 diabetes mellitus (T2DM). Tissue sections from 95 paraffin-embedded colorectal adenocarcinomas, originating from 52 T2DM and 43 non-diabetic patients, were immunostained for p53 [Ventana mouse monoclonal primary antibody (mAb) in vitro diagnostic (IVD) anti-p53, clone Bp53-11] and Bcl-2 (Ventana mAb IVD anti-Bcl-2, clone Bcl-2/124). Immunohistochemistry analysis did not find statistically significant differences between the two groups, but analysis on subgroups of patients in terms of presence or absence of obesity identified overexpression of p53 (>70% of cells) in the T2DM obese patients compared to non-diabetics. Overexpression of p53 was present in 80% of tumor cells coming from T2DM obese patients compared to 37.2% of tumor cells coming from non-diabetics obese and non-obese, and in 36.6% of tumor cells coming from non-diabetic non-obese patients (p=0.024). There was a single non-diabetic obese patient with p53 overexpression. Most cancer cells of T2DM obese patients presented more frequently p53 overexpression by comparison with cancer cells of the T2DM non-obese patients (80% vs 40.5%, p=0.028). Bcl-2/p53 co-expression was an infrequent event in T2DM patients' group. The results of this study suggest that patients with colorectal adenocarcinoma that associate T2DM and obesity exhibit higher p53 protein expression in malignant cells. In conclusion, our research highlights that obesity is a potential key factor in the relationship between T2DM and colorectal cancer.
Collapse
Affiliation(s)
| | - Mihai Ciprian Stoicea
- Department of Pathology, Regina Maria Central Reference Laboratory, Bucharest, Romania
| | - Valentin Enache
- Department of Pathology, Emergency Clinical Hospital, Bucharest, Romania
| | - Răzvan Matei Bratu
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Petronel Mustăţea
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Ramona Maria Drăguţ
- Department of Diabetes, Nutrition and Metabolic Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Emilia Rusu
- Department of Diabetes, Nutrition and Metabolic Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Constantin Ionescu-Tîrgovişte
- Department of Diabetes, Nutrition and Metabolic Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania , Department of Diabetes, Nutrition and Metabolic Diseases, Prof. Dr. N. C. Paulescu National Institute of Diabetes, Nutrition and Metabolic Diseases, Bucharest, Romania
| | - Gabriela Radulian
- Department of Diabetes, Nutrition and Metabolic Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania , Department of Diabetes, Nutrition and Metabolic Diseases, Prof. Dr. N. C. Paulescu National Institute of Diabetes, Nutrition and Metabolic Diseases, Bucharest, Romania
| |
Collapse
|
12
|
Lau MF, Chua KH, Sabaratnam V, Kuppusamy UR. In vitro Anti-colorectal Cancer Potential of the Medicinal Mushroom Ganoderma neo-japonicum Imazeki in Hyperglycemic Condition: Impact on Oxidative Stress, Cell Cycle and Apoptosis. Nutr Cancer 2021; 74:978-995. [PMID: 34085886 DOI: 10.1080/01635581.2021.1931701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Clinical efficacy of chemotherapy is often compromised by diabetogenic glucose on colorectal cancer (CRC). High glucose has been shown to diminish the cytotoxicity of anticancer drugs. The issue can potentially be addressed with natural products. Recently, we revealed that Ganoderma neo-japonicum exhibits inhibitory activities against human colonic carcinoma cells. In this study, the impacts of hexane fraction (Hex, sterol-enriched) and chloroform fraction (Chl, terpenoid-enriched) were further elucidated. The cellular responses, including oxidative stress, cell cycle, and apoptosis were compared between the presence of normal glucose (NG, 5.5 mM) and high glucose (HG, 25 mM). HG promoted cell viability with concomitant elevation of GSH level. Both Hex and Chl fractions stimulated NO production, in addition, induced cell cycle arrest. The apoptotic effect of Hex fraction was glucose-dependent, but Chl fraction triggered apoptosis with an equivalent extent in NG and HG conditions. Overall, the active fractions from G. neo-japonicum show therapeutic potential in managing hyperglycemia-associated CRC.
Collapse
Affiliation(s)
- Meng-Fei Lau
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Mushroom Research Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Kek-Heng Chua
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Mushroom Research Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Vikineswary Sabaratnam
- Mushroom Research Centre, University of Malaya, Kuala Lumpur, Malaysia.,Institute of Biological Science, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Umah Rani Kuppusamy
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Mushroom Research Centre, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
13
|
Bao Y, Suvesh M, Li X, Bai X, Li H, Li X, Xu D, Liu L. Ebp1 p48 promotes oncogenic properties in hepatocellular carcinoma through p38 MAPK/HIF1α activation and p53 downregulation. Mol Carcinog 2021; 60:252-264. [PMID: 33634940 DOI: 10.1002/mc.23288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/21/2022]
Abstract
The ErbB3 binding protein 1 (Ebp1) has been reported in several cancers, in which it can act as either a pro-oncogenic regulator or a tumor suppressor. However, the biological function and molecular mechanism of Ebp1 p48 in hepatocellular carcinoma (HCC) remain unclear. Here, we report that the long isoform of Ebp1, p48, is highly expressed in HCC tissues compared with normal tissues. Ebp1 p48 expression was correlated with the tumor size in HCC patients. Silencing Ebp1 p48 by transduction with lentiviral shEbp1 dramatically reduced the proliferation rate, soft agar colony generation, and tumor formation in vivo. We further demonstrated that Ebp1 p48 knockdown resulted in decreased p38 phosphorylation, which subsequently reduced hypoxia-inducible factor 1α (HIF1α) expression. Moreover, Ebp1 p48 knockdown led to an upregulation of p53 expression through MDM2 downregulation. Taken together, these results suggest that the Ebp1/p38/HIF1α signaling pathway and the Ebp1-mediated downregulation of p53 are involved in hepatocarcinogenesis. Therefore, Ebp1 and its downstream signaling pathways may be promising therapeutic targets of HCC.
Collapse
Affiliation(s)
- Yanqiu Bao
- Department of Pathology, Affiliated Hospital of Yanbian University, Jilin, China
| | - Munakarmi Suvesh
- Division of GI and Hepatology, Departments of Internal Medicine, The Research Institute of Clinical Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Jeonbuk, Republic of Korea
| | - Xiaobo Li
- Department of Pathology, Affiliated Hospital of Yanbian University, Jilin, China
| | - Xin Bai
- Department of Pathology, Affiliated Hospital of Yanbian University, Jilin, China
| | - Hua Li
- Department of Pathology, Affiliated Hospital of Yanbian University, Jilin, China.,Division of GI and Hepatology, Departments of Internal Medicine, The Research Institute of Clinical Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Jeonbuk, Republic of Korea
| | - Xiangdan Li
- Center of Morphological Experiment, Medical College of Yanbian University, Jilin, China
| | - Dongyuan Xu
- Center of Morphological Experiment, Medical College of Yanbian University, Jilin, China
| | - Lan Liu
- Department of Pathology, Affiliated Hospital of Yanbian University, Jilin, China
| |
Collapse
|
14
|
Ahn HR, Kang SY, Youn HJ, Jung SH. Hyperglycemia during Adjuvant Chemotherapy as a Prognostic Factor in Breast Cancer Patients without Diabetes. J Breast Cancer 2020; 23:398-409. [PMID: 32908790 PMCID: PMC7462816 DOI: 10.4048/jbc.2020.23.e44] [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: 04/24/2020] [Accepted: 06/26/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose Breast cancer treatments, including chemotherapy, administered in combination with glucocorticoids can induce hyperglycemia. This study aimed to investigate the effect of hyperglycemia during adjuvant chemotherapy on the prognosis of breast cancer patients without a known history of diabetes. Methods In this study, 936 patients who underwent breast cancer surgery from 2010 to 2015 were initially selected as participants. Chemotherapy-related hyperglycemia was defined as fasting plasma glucose levels ≥ 100 mg/dL or random blood glucose levels ≥ 140 mg/dL during 2 or more cycles of adjuvant chemotherapy. After dividing the patients into the euglycemia and hyperglycemia groups, univariate and multivariate analyses were performed, and survival outcomes were analyzed by propensity score matching. Results The mean age of the patients was 47.4 ± 7.7 years, and the median follow-up period was 70.1 months. Eighty-two patients (19.4%) were diagnosed as having hyperglycemia. There were significant differences between the euglycemia and hyperglycemia groups with respect to age, hypertension, body mass index, axillary surgery extents, nodal stage, and total steroid dosage. T stage, vascular invasion, and hyperglycemia were identified as prognostic factors of relapse-free survival (RFS). The 5-year RFS rates were 92.0% and 82.3% in the euglycemia and hyperglycemia groups, respectively, and there was a statistically significant difference between the 2 groups (p = 0.011). The 5-year overall survival rates were 94.6% and 92.0% in the euglycemia and hyperglycemia groups, respectively, showing no statistically significant difference between the 2 groups (p = 0.113). Conclusion These data suggest that hyperglycemia during adjuvant chemotherapy is a prognostic factor for RFS in breast cancer patients without diabetes.
Collapse
Affiliation(s)
- Ha Rim Ahn
- Department of Surgery, Research Institute of Clinical Medicine, Jeonbuk National University Hospital, Jeonbuk National University and Biomedical Research Institute, Jeonju, Korea
| | - Sang Yull Kang
- Department of Surgery, Research Institute of Clinical Medicine, Jeonbuk National University Hospital, Jeonbuk National University and Biomedical Research Institute, Jeonju, Korea
| | - Hyun Jo Youn
- Department of Surgery, Research Institute of Clinical Medicine, Jeonbuk National University Hospital, Jeonbuk National University and Biomedical Research Institute, Jeonju, Korea
| | - Sung Hoo Jung
- Department of Surgery, Research Institute of Clinical Medicine, Jeonbuk National University Hospital, Jeonbuk National University and Biomedical Research Institute, Jeonju, Korea
| |
Collapse
|
15
|
D'Orazi G, Garufi A, Cirone M. Nuclear factor erythroid 2 (NF-E2) p45-related factor 2 interferes with homeodomain-interacting protein kinase 2/p53 activity to impair solid tumors chemosensitivity. IUBMB Life 2020; 72:1634-1639. [PMID: 32593231 DOI: 10.1002/iub.2334] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022]
Abstract
Resistance to chemotherapy represents a major hurdle to successful cancer treatment. A key role for efficient response to anticancer therapies is played by TP53 oncosuppressor gene that indeed is mutated in 50% of human cancers or inactivated at protein level in the remaining 50%. Homeodomain-interacting protein kinase 2 (HIPK2) is the wild-type p53 (wtp53) apoptotic activator, and its inhibition by hypoxia or hyperglycemia may contribute to tumor chemoresistance mainly by impairing p53 apoptotic activity. Another important molecule able to induce chemoresistance is nuclear factor erythroid 2 (NF-E2) p45-related factor 2 (NRF2) transcription factor, whose activation by oxidative and/or electrophilic stress regulates a transcriptional antioxidant program allowing cancer cells to adapt and survive to stresses. NRF2 may shift from cytoprotective to tumor-promoting function, according to tumor phases. NRF2 may crosstalk with both wtp53 and mutant p53 (mutp53), inhibiting the wtp53 apoptotic function and strengthening the mutp53 oncogenic function. NRF2 has also been shown to induce HIPK2 mRNA expression cooperating in inducing cytoprotection. Although HIPK2, p53, and NRF2 have been individually extensively studied, their interplay has not been clearly addressed yet. On the basis of the background and our results, we aim at hypothesizing the unexpected pro-survival activity played by the NRF2/HIPK2/p53 interplay that can be hijacked by cancer cells to bypass drugs cytotoxicity.
Collapse
Affiliation(s)
- Gabriella D'Orazi
- Department of Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alessia Garufi
- Department of Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy.,Department of Medical Sciences, University 'G. d'Annunzio', Chieti, Italy
| | - Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.,Laboratory affiliated to Pasteur Institute, Italy-Foundation Cenci Bolognetti, Rome, Italy
| |
Collapse
|
16
|
Garufi A, Traversi G, Cirone M, D'Orazi G. HIPK2 role in the tumor-host interaction: Impact on fibroblasts transdifferentiation CAF-like. IUBMB Life 2019; 71:2055-2061. [PMID: 31414572 PMCID: PMC6899452 DOI: 10.1002/iub.2144] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/26/2019] [Indexed: 12/16/2022]
Abstract
The dialogue between cancer cells and the surrounding fibroblasts, tumor-associated macrophages (TAM), and immune cells can create a tumor microenvironment (TME) able to promote tumor progression and metastasis and induce resistance to anticancer therapies. Cancer cells, by producing growth factors and cytokines, can recruit and activate fibroblasts in the TME inducing their transdifferention in cancer-associated fibroblasts (CAFs). Then, CAFs, in a reciprocal cross-talk with cancer cells, sustain cancer growth and survival and support malignancy and tumor resistance to therapies. Therefore, the identification of the molecular mechanisms regulating the interplay between cancer cells and fibroblasts can offer an intriguing opportunity for novel diagnostic and therapeutic anticancer purpose. HIPK2 is a multifunctional tumor suppressor protein that modulates cancer cell growth and apoptosis in response to anticancer drugs and negatively regulates pathways involved in tumor progression and chemoresistance. HIPK2 protein downregulation is induced by hypoxia and hyperglycemia and HIPK2 knockdown favors tumor progression and resistance to therapy other than a pseudohypoxic, inflammatory, and angiogenic cancer phenotype. Therefore, we hypothesized that HIPK2 modulation in cancer cells could contribute to modify the tumor-host interaction. In support of our hypothesis, here we provide evidence that culturing human fibroblasts (hFB) with conditioned media derived from cancer cells undergoing HIPK2 knockdown (CMsiHIPK2 ) triggered their transdifferentiation CAF-like, compared to hFB cultured with CM-derived from HIPK2-carrying control cancer cells. CAF transdifferentiation was identified by expression of several markers including α-smooth muscle actin (α-SMA) and collagen I and correlated with autophagy-mediated caveolin-1 degradation. Although the molecular mechanisms dictating CAF-transdifferentiation need to be elucidated, these results open the way to further study the role of HIPK2 in TME remodeling for prognostic and therapeutic purpose.
Collapse
Affiliation(s)
- Alessia Garufi
- Department of Medical SciencesUniversity “G. d'Annunzio”ChietiItaly
- Department of ResearchIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Gianandrea Traversi
- Department of Medical SciencesUniversity “G. d'Annunzio”ChietiItaly
- Department of ResearchIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Mara Cirone
- Department of Experimental Medicine“Sapienza” University of Rome, Italy, Laboratory affiliated to Pasteur InstituteRomeItaly
| | - Gabriella D'Orazi
- Department of Medical SciencesUniversity “G. d'Annunzio”ChietiItaly
- Department of ResearchIRCCS Regina Elena National Cancer InstituteRomeItaly
| |
Collapse
|
17
|
Li W, Zhang X, Sang H, Zhou Y, Shang C, Wang Y, Zhu H. Effects of hyperglycemia on the progression of tumor diseases. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:327. [PMID: 31337431 PMCID: PMC6651927 DOI: 10.1186/s13046-019-1309-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/02/2019] [Indexed: 12/13/2022]
Abstract
Malignant tumors are often multifactorial. Epidemiological studies have shown that hyperglycemia raises the prevalence and mortality of certain malignancies, like breast, liver, bladder, pancreatic, colorectal, endometrial cancers. Hyperglycemia can promote the proliferation, invasion and migration, induce the apoptotic resistance and enhance the chemoresistance of tumor cells. This review focuses on the new findings in the relationship between hyperglycemia and tumor development.
Collapse
Affiliation(s)
- Wenjie Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Xuehui Zhang
- Department of Pharmacy, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, 215228, China
| | - Hui Sang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Ying Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Chunyu Shang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Yongqing Wang
- Department of Pharmacy, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, 215228, China. .,Research Division of Clinical Pharmacology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China.
| | - Hong Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China.
| |
Collapse
|
18
|
Garufi A, Traversi G, Gilardini Montani MS, D'Orazi V, Pistritto G, Cirone M, D'Orazi G. Reduced chemotherapeutic sensitivity in high glucose condition: implication of antioxidant response. Oncotarget 2019; 10:4691-4702. [PMID: 31384396 PMCID: PMC6659798 DOI: 10.18632/oncotarget.27087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/05/2019] [Indexed: 12/19/2022] Open
Abstract
Resistance to chemotherapy represents a major obstacle to successful treatment. The generation of reactive oxygen species (ROS) has been directly linked to the cytotoxic effects of several antitumor agents, including Adriamycin (ADR), and modulation of the oxidative balance has been implicated in the development and/or regulation of resistance to chemotherapeutic drugs. We recently showed that high glucose (HG) markedly diminished the cancer cell death induced by anticancer agents such as ADR. In the present study we attempted to evaluate the mechanism that impaired the cytotoxic effect of ADR in HG. We found that, in colon cancer cells, HG attenuated ADR-induced ROS production that consequently diminished ADR-induced H2AX phosphorylation and micronuclei (MN) formation. Mechanistically, HG attenuation of ADR-induced ROS production correlated with increased antioxidant response promoted by NRF2 activity. Thus, pharmacologic inhibition of NRF2 pathway by brusatol re-established the ADR cytotoxic effect impaired by HG. Together, the data provide new insights into chemotherapeutic-resistance mechanisms in HG condition dictated by increased NRF2-induced antioxidant response and how they may be overcome in order to restore chemosensitivity and ADR-induced cell death.
Collapse
Affiliation(s)
- Alessia Garufi
- IRCCS Regina Elena National Cancer Institute, Department of Research, Rome 00144, Italy.,University 'G. d'Annunzio', Department of Medical and Biotechnological Sciences, Chieti 66013, Italy
| | - Gianandrea Traversi
- IRCCS Regina Elena National Cancer Institute, Department of Research, Rome 00144, Italy.,University 'G. d'Annunzio', Department of Medical and Biotechnological Sciences, Chieti 66013, Italy
| | | | | | - Giuseppa Pistritto
- University Tor Vergata, Department of Systems Medicine, Rome 00133, Italy
| | - Mara Cirone
- Sapienza University, Department of Experimental Medicine, Rome 00161, Italy
| | - Gabriella D'Orazi
- IRCCS Regina Elena National Cancer Institute, Department of Research, Rome 00144, Italy.,University 'G. d'Annunzio', Department of Medical and Biotechnological Sciences, Chieti 66013, Italy
| |
Collapse
|
19
|
Cirone M, Gilardini Montani MS, Granato M, Garufi A, Faggioni A, D'Orazi G. Autophagy manipulation as a strategy for efficient anticancer therapies: possible consequences. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:262. [PMID: 31200739 PMCID: PMC6570888 DOI: 10.1186/s13046-019-1275-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 06/10/2019] [Indexed: 12/14/2022]
Abstract
Autophagy is a catabolic process whose activation may help cancer cells to adapt to cellular stress although, in some instances, it can induce cell death. Autophagy stimulation or inhibition has been considered an opportunity to treat cancer, especially in combination with anticancer therapies, although autophagy manipulation may be viewed as controversial. Thus, whether to induce or to inhibit autophagy may be the best option in the different cancer patients is still matter of debate. Her we will recapitulate the possible advantages or disadvantages of manipulating autophagy in cancer, not only with the aim to obtain cancer cell death and disable oncogenes, but also to evaluate its interplay with the immune response which is fundamental for the success of anticancer therapies.
Collapse
Affiliation(s)
- Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy. .,Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy.
| | - Maria Saveria Gilardini Montani
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.,Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Marisa Granato
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.,Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Alessia Garufi
- Department of Medical Science, University 'G. D'Annunzio', 66013, Chieti, Italy.,Department of Research, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Alberto Faggioni
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.,Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Gabriella D'Orazi
- Department of Medical Science, University 'G. D'Annunzio', 66013, Chieti, Italy. .,Department of Research, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy.
| |
Collapse
|
20
|
Bergandi L, Mungo E, Morone R, Bosco O, Rolando B, Doublier S. Hyperglycemia Promotes Chemoresistance Through the Reduction of the Mitochondrial DNA Damage, the Bax/Bcl-2 and Bax/Bcl-XL Ratio, and the Cells in Sub-G1 Phase Due to Antitumoral Drugs Induced-Cytotoxicity in Human Colon Adenocarcinoma Cells. Front Pharmacol 2018; 9:866. [PMID: 30150934 PMCID: PMC6099160 DOI: 10.3389/fphar.2018.00866] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/17/2018] [Indexed: 01/08/2023] Open
Abstract
Diabetes and cancer are common, chronic, and potentially fatal diseases that frequently co-exist. Observational studies clearly indicate that the risk of several types of cancer is increased in diabetic patients and a number of cancer types have shown a higher mortality rate in patients with hyperglycemic associated pathologies. This scenario could be due, at least in part, to a lower efficacy of the cancer treatments which needs to be better investigated. Here, we evaluated the effects of a prolonged exposure to high glucose (HG) to the response to chemotherapy on human colon adenocarcinoma HT29 and LOVO cell lines. We observed that hyperglycemia protected against the decreased cell viability and cytotoxicity and preserved from the mitochondrial DNA lesions induced by doxorubicin (DOX) and 5-fluorouracil (5-FU) treatments by lowering ROS production. In HT29 cells the amount of intracellular DOX and its nuclear localization were not modified by HG incubation in terms of Pgp, BCRP, MRP1, 5 and 8 activity and gene expression. On the contrary, in LOVO cells, the amount of intracellular DOX was significantly decreased after a bolus of DOX in HG condition and the expression and activity of MPR1 was increased, suggesting that HG promotes drug chemoresistance in both HT29 and LOVO cells, but in a different way. In both cell types, HG condition prevented the susceptibility to apoptosis by decreasing the ratio Bax/Bcl-2 and Bax/Bcl-XL and diminished the level of cytosolic cytochrome c and the cleavage of full length of PARP induced by DOX and 5-FU. Finally, hyperglycemia reduced cell death by decreasing the cell percentage in sub-G1 peak induced by DOX (via a cell cycle arrest in the G2/M phase) and 5-FU (via a cell cycle arrest in the S phase) in HT29 and LOVO cells. Taken together, our data showed that a prolonged exposure to HG protects human colon adenocarcinoma cells from the cytotoxic effects of two widely used chemotherapeutic drugs, impairing the effectiveness of the chemotherapy itself.
Collapse
Affiliation(s)
| | - Eleonora Mungo
- Department of Oncology, University of Turin, Turin, Italy
| | - Rosa Morone
- Department of Oncology, University of Turin, Turin, Italy
| | - Ornella Bosco
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Barbara Rolando
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | | |
Collapse
|
21
|
Giri B, Dey S, Das T, Sarkar M, Banerjee J, Dash SK. Chronic hyperglycemia mediated physiological alteration and metabolic distortion leads to organ dysfunction, infection, cancer progression and other pathophysiological consequences: An update on glucose toxicity. Biomed Pharmacother 2018; 107:306-328. [PMID: 30098549 DOI: 10.1016/j.biopha.2018.07.157] [Citation(s) in RCA: 221] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/15/2018] [Accepted: 07/31/2018] [Indexed: 02/09/2023] Open
Abstract
Chronic exposure of glucose rich environment creates several physiological and pathophysiological changes. There are several pathways by which hyperglycemia exacerbate its toxic effect on cells, tissues and organ systems. Hyperglycemia can induce oxidative stress, upsurge polyol pathway, activate protein kinase C (PKC), enhance hexosamine biosynthetic pathway (HBP), promote the formation of advanced glycation end-products (AGEs) and finally alters gene expressions. Prolonged hyperglycemic condition leads to severe diabetic condition by damaging the pancreatic β-cell and inducing insulin resistance. Numerous complications have been associated with diabetes, thus it has become a major health issue in the 21st century and has received serious attention. Dysregulation in the cardiovascular and reproductive systems along with nephropathy, retinopathy, neuropathy, diabetic foot ulcer may arise in the advanced stages of diabetes. High glucose level also encourages proliferation of cancer cells, development of osteoarthritis and potentiates a suitable environment for infections. This review culminates how elevated glucose level carries out its toxicity in cells, metabolic distortion along with organ dysfunction and elucidates the complications associated with chronic hyperglycemia.
Collapse
Affiliation(s)
- Biplab Giri
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India; Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India.
| | - Sananda Dey
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India; Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Tanaya Das
- Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Mrinmoy Sarkar
- Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Jhimli Banerjee
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India
| | - Sandeep Kumar Dash
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India.
| |
Collapse
|
22
|
Baldari S, Garufi A, Granato M, Cuomo L, Pistritto G, Cirone M, D'Orazi G. Hyperglycemia triggers HIPK2 protein degradation. Oncotarget 2018; 8:1190-1203. [PMID: 27901482 PMCID: PMC5352047 DOI: 10.18632/oncotarget.13595] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/08/2016] [Indexed: 12/23/2022] Open
Abstract
Homeodomain interacting protein kinase-2 (HIPK2) is an evolutionary conserved kinase that modulates several key molecular pathways to restrain tumor growth and induce p53-depending apoptotic cell-death in response to anticancer therapies. HIPK2 silencing in cancer cells leads to chemoresistance and cancer progression, in part due to p53 inhibition. Recently, hyperglycemia has been shown to reduce p53 phosphorylation at serine 46 (Ser46), the target residue of HIPK2, thus impairing p53 apoptotic function. Here we asked whether hyperglycemia could, upstream of p53, target HIPK2. We focused on the effect of high glucose (HG) on HIPK2 protein stability and the underlying mechanisms. We found that HG reduced HIPK2 protein levels, therefore impairing HIPK2-induced p53 apoptotic activity. HG-triggered HIPK2 protein downregulation was rescued by both proteasome inhibitor MG132 and by protein phosphatase inhibitors Calyculin A (CL-A) and Okadaic Acid (OA). Looking for the phosphatase involved, we found that protein phosphatase 2A (PP2A) induced HIPK2 degradation, as evidenced by directly activating PP2A with FTY720 or by silencing PP2A with siRNA in HG condition. The effect of PP2A on HIPK2 protein degradation could be in part due to hypoxia-inducible factor-1 (HIF-1) activity which has been previously shown to induce HIPK2 proteasomal degradation through several ubiquitin ligases. Validation analysed performed with HIF-1α dominant negative or with silencing of Siah2 ubiquitin ligase clearly showed rescue of HG-induced HIPK2 degradation. These findings demonstrate how hyperglycemia, through a complex protein cascade, induced HIPK2 downregulation and consequently impaired p53 apoptotic activity, revealing a novel link between diabetes/obesity and tumor resistance to therapies.
Collapse
Affiliation(s)
- Silvia Baldari
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Alessia Garufi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144 Rome, Italy.,Department of Medical Sciences, Tumor Biology Unit, University 'G. d'Annunzio', 66013 Chieti, Italy
| | - Marisa Granato
- Department of Experimental Medicine, Pasteur-Fondazione Cenci Bolognetti Institute, Sapienza University, 00100 Rome, Italy
| | - Laura Cuomo
- U.O.C. Clinical Pathology, A.C.O., San Filippo Neri Hospital, 00100 Rome, Italy
| | - Giuseppa Pistritto
- Department of Systems Medicine, University Tor Vergata, 00133 Rome, Italy
| | - Mara Cirone
- Department of Experimental Medicine, Pasteur-Fondazione Cenci Bolognetti Institute, Sapienza University, 00100 Rome, Italy
| | - Gabriella D'Orazi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144 Rome, Italy.,Department of Medical Sciences, Tumor Biology Unit, University 'G. d'Annunzio', 66013 Chieti, Italy
| |
Collapse
|
23
|
Hyperglycemia and aberrant O-GlcNAcylation: contributions to tumor progression. J Bioenerg Biomembr 2018; 50:175-187. [DOI: 10.1007/s10863-017-9740-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 12/26/2017] [Indexed: 12/17/2022]
|
24
|
Pistritto G, Trisciuoglio D, Ceci C, Garufi A, D'Orazi G. Apoptosis as anticancer mechanism: function and dysfunction of its modulators and targeted therapeutic strategies. Aging (Albany NY) 2017; 8:603-19. [PMID: 27019364 PMCID: PMC4925817 DOI: 10.18632/aging.100934] [Citation(s) in RCA: 1025] [Impact Index Per Article: 128.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/08/2016] [Indexed: 02/07/2023]
Abstract
Apoptosis is a form of programmed cell death that results in the orderly and efficient removal of damaged cells, such as those resulting from DNA damage or during development. Apoptosis can be triggered by signals from within the cell, such as genotoxic stress, or by extrinsic signals, such as the binding of ligands to cell surface death receptors. Deregulation in apoptotic cell death machinery is an hallmark of cancer. Apoptosis alteration is responsible not only for tumor development and progression but also for tumor resistance to therapies. Most anticancer drugs currently used in clinical oncology exploit the intact apoptotic signaling pathways to trigger cancer cell death. Thus, defects in the death pathways may result in drug resistance so limiting the efficacy of therapies. Therefore, a better understanding of the apoptotic cell death signaling pathways may improve the efficacy of cancer therapy and bypass resistance. This review will highlight the role of the fundamental regulators of apoptosis and how their deregulation, including activation of anti-apoptotic factors (i.e., Bcl-2, Bcl-xL, etc) or inactivation of pro-apoptotic factors (i.e., p53 pathway) ends up in cancer cell resistance to therapies. In addition, therapeutic strategies aimed at modulating apoptotic activity are briefly discussed.
Collapse
Affiliation(s)
- Giuseppa Pistritto
- Department of Systems Medicine, University "Tor Vergata", 00133 Rome, Italy
| | - Daniela Trisciuoglio
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00158 Rome, Italy
| | - Claudia Ceci
- Department of Systems Medicine, University "Tor Vergata", 00133 Rome, Italy
| | - Alessia Garufi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00158 Rome, Italy.,Department of Medical Oral and Biotechnological Sciences, Tumor Biology Unit, University "G. d'Annunzio", 66013 Chieti, Italy
| | - Gabriella D'Orazi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00158 Rome, Italy.,Department of Medical Oral and Biotechnological Sciences, Tumor Biology Unit, University "G. d'Annunzio", 66013 Chieti, Italy
| |
Collapse
|
25
|
Granato M, Gilardini Montani MS, Romeo MA, Santarelli R, Gonnella R, D'Orazi G, Faggioni A, Cirone M. Metformin triggers apoptosis in PEL cells and alters bortezomib-induced Unfolded Protein Response increasing its cytotoxicity and inhibiting KSHV lytic cycle activation. Cell Signal 2017; 40:239-247. [PMID: 28964970 DOI: 10.1016/j.cellsig.2017.09.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/26/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022]
Abstract
Metformin, the most used drug for the treatment of diabetes type 2 patients, has been shown to have anti-cancer properties. In this study, we found that metformin induced apoptosis in Primary Effusion Lymphoma (PEL) cells, an aggressive B cell lymphoma associated with KSHV against which the conventional therapies usually fail. The cytotoxic effect of metformin correlated with intracellular reactive oxygen species reduction, activation of AMPK, the inhibition of pro-survival pathways such as mTOR and STAT3 and the down-regulation of v-FLIP, a latent viral antigen that also plays a pivotal role in PEL cell survival. Interestingly, we found that metformin could be used to potentiate the bortezomib-mediated cytotoxicity against PEL cells and to inhibit the activation of KSHV lytic cycle, a side effect of this treatment that resulted in a block of autophagy in these cells. Mechanistically, metformin altered UPR activated by bortezomib, leading to a reduced expression of BiP, up-regulation of CHOP and down-regulation of Bcl-2. In summary, this study suggests that metformin could represent a promising strategy for the treatment of PEL alone or in combination with bortezomib. In the latter case, besides exerting a stronger cytotoxic effect, it might be used to restrain bortezomib-induced viral replication that is involved in the maintenance and progression of KSHV-associated malignancies.
Collapse
Affiliation(s)
- Marisa Granato
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | | | - Maria Anele Romeo
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Roberta Santarelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Roberta Gonnella
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Gabriella D'Orazi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144 Rome, Italy; Department of Medical, Oral and Biotechnological Sciences, Tumor Biology Section, University 'G. d'Annunzio', Chieti, Italy
| | - Alberto Faggioni
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161 Rome, Italy.
| | - Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161 Rome, Italy.
| |
Collapse
|
26
|
Garufi A, Pistritto G, Baldari S, Toietta G, Cirone M, D'Orazi G. p53-Dependent PUMA to DRAM antagonistic interplay as a key molecular switch in cell-fate decision in normal/high glucose conditions. J Exp Clin Cancer Res 2017; 36:126. [PMID: 28893313 PMCID: PMC5594515 DOI: 10.1186/s13046-017-0596-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/05/2017] [Indexed: 12/21/2022] Open
Abstract
Background As an important cellular stress sensor phosphoprotein p53 can trigger cell cycle arrest and apoptosis and regulate autophagy. The p53 activity mainly depends on its transactivating function, however, how p53 can select one or another biological outcome is still a matter of profound studies. Our previous findings indicate that switching cancer cells in high glucose (HG) impairs p53 apoptotic function and the transcription of target gene PUMA. Methods and results Here we report that, in response to drug adriamycin (ADR) in HG, p53 efficiently induced the expression of DRAM (damage-regulated autophagy modulator), a p53 target gene and a stress-induced regulator of autophagy. We found that ADR treatment of cancer cells in HG increased autophagy, as displayed by greater LC3II accumulation and p62 degradation compared to ADR-treated cells in low glucose. The increased autophagy in HG was in part dependent on p53-induced DRAM; indeed DRAM knockdown with specific siRNA reversed the expression of the autophagic markers in HG. A similar outcome was achieved by inhibiting p53 transcriptional activity with pifithrin-α. DRAM knockdown restored the ADR-induced cell death in HG to the levels obtained in low glucose. A similar outcome was achieved by inhibition of autophagy with cloroquine (CQ) or with silencing of autophagy gene ATG5. DRAM knockdown or inhibition of autophagy were both able to re-induce PUMA transcription in response to ADR, underlining a reciprocal interplay between PUMA to DRAM to unbalance p53 apoptotic activity in HG. Xenograft tumors transplanted in normoglycemic mice displayed growth delay after ADR treatment compared to those transplanted in diabetics mice and such different in vivo response correlated with PUMA to DRAM gene expression. Conclusions Altogether, these findings suggest that in normal/high glucose condition a mutual unbalance between p53-dependent apoptosis (PUMA) and autophagy (DRAM) gene occurred, modifying the ADR-induced cancer cell death in HG both in vitro and in vivo.
Collapse
Affiliation(s)
- Alessia Garufi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy.,Department of Medical, Oral and Biotechnological Sciences, Tumor Biology Section, University "G. d'Annunzio", Via de Vestini, 31, 66013, Chieti, Italy
| | | | - Silvia Baldari
- Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy
| | - Mara Cirone
- Department of Experimental Medicine, Institute Pasteur Cenci Bolognetti Foundation, Sapienza University, Rome, Italy
| | - Gabriella D'Orazi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy. .,Department of Medical, Oral and Biotechnological Sciences, Tumor Biology Section, University "G. d'Annunzio", Via de Vestini, 31, 66013, Chieti, Italy.
| |
Collapse
|
27
|
Impact of hyperglycemia on the efficacy of chemotherapy-A systematic review of preclinical studies. Crit Rev Oncol Hematol 2017; 113:235-241. [PMID: 28427512 DOI: 10.1016/j.critrevonc.2017.03.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 01/05/2017] [Accepted: 03/08/2017] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Antineoplastic agents can provoke hyperglycemia in cancer patients with and without diabetes mellitus. We systematically reviewed the impact of hyperglycemia on the efficacy of chemotherapy. METHODS MEDLINE was searched for preclinical intervention studies which compared chemotherapy response in hyperglycemic and euglycemic conditions. RESULTS Thirteen preclinical studies, including 23 cell lines and 2 animal experiments were identified. In 14 cell lines and 2 animal studies, chemotherapy response was lower in a hyperglycemic (>15mmol/L) compared to a euglycemic environment (5mmol/L). The response was similar in 4 cell lines. In the remaining 5 cell lines, the hyperglycemic environment potentiated chemotherapy efficacy. CONCLUSION Hyperglycemia attenuated the antiproliferative effect of chemotherapy in preclinical experiments, but the results are inconsistent. Whether hyperglycemia influences efficacy of chemotherapy in patients needs to be explored.
Collapse
|
28
|
Wang L, Zhong N, Liu S, Zhu X, Liu Y. High glucose stimulates proliferation and inhibits apoptosis of non-small-cell lung cancer cells by JNK-mediated downregulation of p53 pathway. Acta Biochim Biophys Sin (Shanghai) 2017; 49:286-288. [PMID: 28104581 DOI: 10.1093/abbs/gmw137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Indexed: 11/14/2022] Open
Affiliation(s)
- Lumei Wang
- The Key Laboratory of Exercise and Health Sciences of Ministry of Education, School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
- Department of Pathology, Dongfang Hospital, Tongji University, Shanghai 200120, China
| | - Nanzhe Zhong
- Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Shujuan Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, China
| | - Xiaoyan Zhu
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Yujian Liu
- The Key Laboratory of Exercise and Health Sciences of Ministry of Education, School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|
29
|
Zhao M, Yu S, Zhang M. Differential expression of multidrug resistance‑related proteins in adriamycin‑resistant (pumc‑91/ADM) and parental (pumc‑91) human bladder cancer cell lines. Mol Med Rep 2016; 14:4741-4746. [PMID: 27748815 DOI: 10.3892/mmr.2016.5806] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 08/16/2016] [Indexed: 11/05/2022] Open
Abstract
Multidrug resistance (MDR) is the major obstacle to bladder cancer chemotherapy. Several mechanisms have been implicated in the development of MDR, including extrusion of the drug by cell membrane pumps, associated with P‑glycoprotein (P‑gp) and multidrug resistance‑associated protein (MRP); increased DNA damage repair, associated with topoisomerase II (Topo II); suppression of drug‑induced apoptosis, associated with p53; and regulation of cancer cell growth, associated with vascular endothelial growth factor (VEGF). In the present study, the expression levels of these five markers were detected in an adriamycin (ADM)‑resistant human bladder cancer cell line (pumc‑91/ADM) and its parental cell line (pumc‑91), in order to determine which marker is more important, or whether all of them participate in drug resistance. The expression levels of P‑gp, MRP, Topo II, VEGF and p53 were measured in the two cell lines by reverse transcription‑quantitative polymerase chain reaction, western blotting and immunohistochemistry. A significant increase in P‑gp, MRP and VEGF, and a decrease in Topo II mRNA expression were detected in the pumc‑91/ADM drug‑resistant cell line compared with the pumc‑91 cell line; however, no difference in p53 mRNA expression was detected between the cells. In pumc‑91/ADM cells, the protein expression levels of P‑gp and MRP were upregulated, whereas Topo II was significantly decreased. However, no marked differences in p53 or VEGF expression were detected between the two cell lines at the protein level. The cytoplasmic and cell membrane localization of P‑gp and MRP, the cytoplasmic localization of VEGF, and the nuclear localization of p53 and Topo II were confirmed in the two cell lines. The present study detected increased P‑gp and MRP, and reduced Topo II expression in pumc‑91/ADM cells compared with pumc‑91 cells; however, no difference was detected in p53 and VEGF expression between the cell lines. In conclusion, a significant upregulation of MRP and downregulation of Topo II were detected in the ADM‑resistant human bladder cancer cell line (pumc‑91/ADM) compared with in the parental cell line (pumc‑91).
Collapse
Affiliation(s)
- Man Zhao
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| | - Shuliang Yu
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| | - Man Zhang
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| |
Collapse
|
30
|
Thakur B, Ray P. p53 Loses grip on PIK3CA expression leading to enhanced cell survival during platinum resistance. Mol Oncol 2016; 10:1283-95. [PMID: 27401370 PMCID: PMC5423208 DOI: 10.1016/j.molonc.2016.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/25/2016] [Accepted: 06/19/2016] [Indexed: 12/28/2022] Open
Abstract
Tumour suppressor p53, a master transcriptional regulator determines cell fate through preferential activation/repression of a myriad of genes during stress. Till date, activation and preferential binding of p53 on different promoters was reported to be influenced by the nature, strength and duration of stress which mediates its post translational modifications. Cisplatin, a widely used cytotoxic drug represses PIK3CA promoter activity and attenuates PI3K/AKT cell survival pathway through p53 activation in sensitive cells. However, very little is understood about the overall mechanism of p53-PIK3CA interaction and influence of p53 on the transcriptional status of PIK3CA during cisplatin resistance. Here we showed that cisplatin could dynamically alter p53 occupancy between the p53 binding sequences present in PIK3CA promoter in ovarian and breast cancer cells. This altered occupancy is dictated by higher acetylation and hyper-phosphorylation at serine 15, serine 20 and serine 46 residues. Interestingly, cisplatin resistant cells when challenged with cisplatin demonstrated abolished PIK3CA promoter attenuation, low level of p53 binding, and loss of p53 serine 46 phosphorylation. A phosphorylation deficient S46A mutant failed to repress PIK3CA in p53 deficient cells. Elevated expression of Bcl2, P27 and cFLIP indicated a pro-survival state in these resistant cells. Non-invasive real time imaging using two different luciferase reporters showed that cisplatin could simultaneously induce PIK3CA attenuation and p53 activation with growth regression in sensitive tumours but not in the resistant tumours where only low level of p53 activation and sustained growth was observed. This is the first report on phosphorylation of p53 serine 46 as a modulator of p53-PIK3CA promoter interaction which influences altered binding of p53 at different consensus sequences in the same promoter in response to chemotherapeutic stress. Absence of such modulation in resistant cellular milieu influences cellular homoeostasis in platinum-resistant cells probably due to altered post translational modification of p53.
Collapse
Affiliation(s)
- Bhushan Thakur
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Pritha Ray
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, Maharashtra, India.
| |
Collapse
|
31
|
Granato M, Rizzello C, Romeo MA, Yadav S, Santarelli R, D'Orazi G, Faggioni A, Cirone M. Concomitant reduction of c-Myc expression and PI3K/AKT/mTOR signaling by quercetin induces a strong cytotoxic effect against Burkitt's lymphoma. Int J Biochem Cell Biol 2016; 79:393-400. [PMID: 27620077 DOI: 10.1016/j.biocel.2016.09.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/01/2016] [Accepted: 09/09/2016] [Indexed: 11/28/2022]
Abstract
Burkitt's lymphoma is an aggressive B cell lymphoma whose pathogenesis involves mainly c-Myc translocation and hyperexpression, in addition to antigen-independent BCR signaling and, in some cases, EBV infection. As result of BCR signaling activation, the PI3K/AKT/mTOR pathway results constitutively activated also in the absence of EBV, promoting cell survival and counterbalancing the pro-apoptotic function that c-Myc may also exert. In this study we found that quercetin, a bioflavonoid widely distributed in plant kingdom, reduced c-Myc expression and inhibited the PI3K/AKT/mTOR activity in BL, leading to an apoptotic cell death. We observed a higher cytotoxic effect against the EBV-negative BL cells in comparison with the positive ones, suggesting that this oncogenic gammaherpesvirus confers an additional resistance to the quercetin treatment. Besides cell survival, PI3K/AKT/mTOR pathway also regulates autophagy: we found that quercetin induced a complete autophagic flux in BL cells, that contributes to c-Myc reduction in some of these cells. Indeed, autophagy inhibition by chloroquine partially restored c-Myc expression in EBV-positive (Akata) and EBV-negative (2A8) cells that harbor c-Myc mutation. Interestingly, chloroquine did not affect the quercetin-mediated reduction of c-Myc expression in Ramos cells, that have no c-Myc mutation in the coding region, although autophagy was induced. These results suggest that mutant c-Myc could be partially degraded through autophagy in BL cells, as previously reported for other mutant oncogenic proteins.
Collapse
Affiliation(s)
- Marisa Granato
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Celeste Rizzello
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Maria Anele Romeo
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Shivangi Yadav
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Roberta Santarelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Gabriella D'Orazi
- Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy; Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Alberto Faggioni
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy.
| | - Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy.
| |
Collapse
|
32
|
Abstract
The p53 protein is essential for the implementation of the cellular response to challenging environmental conditions. Reacting to stochastic nutrient stress, p53 integrates the activity of key metabolite-sensing pathways to coordinate an appropriate cell response. During starvation, p53 activity augments cell survival pathways, inhibits unnecessary growth, and promotes efficient nutrient generation, utilization, and conservation. Similarly, during oxygen stress, p53 facilitates redirection of cellular metabolism toward energy generation through nonoxidative means, the suppression of reactive oxygen species (ROS) generation, and ROS detoxification-promoting cell survival. However, if adverse conditions are too acute or persistent, p53 can switch roles to implement canonical cell killing. The ability of p53 to regulate metabolism is a powerful feature of p53 biology that can both promote cell survival and act as a check on the inappropriate proliferation of cancer cells.
Collapse
|
33
|
Garufi A, Trisciuoglio D, Cirone M, D'Orazi G. ZnCl2 sustains the adriamycin-induced cell death inhibited by high glucose. Cell Death Dis 2016; 7:e2280. [PMID: 27362798 PMCID: PMC5108333 DOI: 10.1038/cddis.2016.178] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/14/2016] [Accepted: 05/30/2016] [Indexed: 12/15/2022]
Abstract
Hyperglycemia, the condition of high blood glucose, is typical of diabetes and obesity and represents a significant clinical problem. The relationship between hyperglycemia and cancer risk has been established by several studies. Moreover, hyperglycemia has been shown to reduce cancer cell response to therapies, conferring resistance to drug-induced cell death. Therefore, counteracting the negative effects of hyperglycemia may positively improve the cancer cell death induced by chemotherapies. Recent studies showed that zinc supplementation may have beneficial effects on glycemic control. Here we aimed at evaluating whether ZnCl2 could counteract the high-glucose (HG) effects and consequently restore the drug-induced cancer cell death. At the molecular level we found that the HG-induced expression of genes known to be involved in chemoresistance (such as HIF-1α, GLUT1, and HK2 glycolytic genes, as well as NF-κB activity) was reduced by ZnCl2 treatment. In agreement, the adryamicin (ADR)-induced apoptotic cancer cell death was significantly impaired by HG and efficiently re-established by ZnCl2 cotreatment. Mechanistically, the ADR-induced c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) phosphorylation, inhibited by HG, was efficiently restored by ZnCl2. The JNK involvement in apoptotic cell death was assessed by the use of JNK dominant-negative expression vector that indeed impaired the ZnCl2 ability to restore drug-induced cell death in HG condition. Altogether, these findings indicate that ZnCl2 supplementation efficiently restored the drug-induced cancer cell death, inhibited by HG, by both sustaining JNK activation and counteracting the glycolytic pathway.
Collapse
Affiliation(s)
- A Garufi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy.,Department of Medical, Oral and Biotechnological Sciences, Tumor Biology Section, University 'G. d'Annunzio', Chieti, Italy
| | - D Trisciuoglio
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - M Cirone
- Department of Experimental Medicine, Pasteur-Fondazione Cenci Bolognetti Institute, Sapienza University, Rome, Italy
| | - G D'Orazi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy.,Department of Medical, Oral and Biotechnological Sciences, Tumor Biology Section, University 'G. d'Annunzio', Chieti, Italy
| |
Collapse
|
34
|
Gilardini Montani MS, Granato M, Cuomo L, Valia S, Di Renzo L, D'Orazi G, Faggioni A, Cirone M. High glucose and hyperglycemic sera from type 2 diabetic patients impair DC differentiation by inducing ROS and activating Wnt/β-catenin and p38 MAPK. Biochim Biophys Acta Mol Basis Dis 2016; 1862:805-813. [PMID: 26769359 DOI: 10.1016/j.bbadis.2016.01.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/14/2015] [Accepted: 01/04/2016] [Indexed: 01/08/2023]
Abstract
Type 2 is the type of diabetes with higher prevalence in contemporary time, representing about 90% of the global cases of diabetes. In the course of diabetes, several complications can occur, mostly due to hyperglycemia and increased reactive oxygen species (ROS) production. One of them is represented by an increased susceptibility to microbial infections and by a reduced capacity to clear them. Therefore, knowing the impact of hyperglycemia on immune system functionality is of utmost importance for the management of the disease. In this study, we show that medium containing high glucose reduced the in-vitro differentiation of monocytes into functional DCs and their activation mediated by PAMPs or DAMPs. Most importantly, the same effects were mediated by the hyperglycemic sera derived by type 2 diabetic patients, mimicking a more physiologic condition. DC dysfunction caused by hyperglycemia may be involved in the inefficient control of infections observed in diabetic patients, given the pivotal role of these cells in both the innate and adaptive immune response. Searching for the molecular mechanisms underlying DC dysfunction, we found that canonical Wnt/β-catenin and p38 MAPK pathways were activated in the DCs differentiated either in the presence of high glucose or of hyper-glycemic sera. Interestingly, the activation of these pathways and the DC immune dysfunction were partially counteracted by the anti-oxidant quercetin, a flavonoid already known to exert several beneficial effects in diabetes.
Collapse
Affiliation(s)
| | - Marisa Granato
- Department of Experimental Medicine, Sapienza University of Rome, viale Regina Elena 324, 00161, Rome, Italy
| | - Laura Cuomo
- U.O.C. Patologia Clinica, A.C.O. San Filippo Neri, Rome, Italy
| | - Sandro Valia
- Department of Experimental Medicine, Sapienza University of Rome, viale Regina Elena 324, 00161, Rome, Italy
| | - Livia Di Renzo
- Department of Experimental Medicine, Sapienza University of Rome, viale Regina Elena 324, 00161, Rome, Italy
| | - Gabriella D'Orazi
- Department of Translational Oncology, Regina Elena National Cancer Institute, Rome, Italy; Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", 66013, Chieti, Italy
| | - Alberto Faggioni
- Department of Experimental Medicine, Sapienza University of Rome, viale Regina Elena 324, 00161, Rome, Italy.
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
35
|
Chen X, Guan X, Zhang H, Xie X, Wang H, Long J, Cai T, Li S, Liu Z, Zhang Y. DAL-1 attenuates epithelial-to mesenchymal transition in lung cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:3. [PMID: 25609022 PMCID: PMC4307741 DOI: 10.1186/s13046-014-0117-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 12/22/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Epithelial-to mesenchymal transition (EMT) involves in metastasis, causing loss of epithelial polarity. Metastasis is the major cause of carcinoma-induced death, but mechanisms are poorly understood. Here we identify differentially expressed in adenocarcinoma of the lung-1 (DAL-1), a protein belongs to the membrane-associated cytoskeleton protein 4.1 family, as an efficient suppressor of EMT in lung cancer. METHODS The relationship between DAL-1 and EMT markers were analyzed by using immunohistochemistry in the clinical lung cancer tissues. Quantitative real-time PCR and western blot were used to characterize the expression of the EMT indicator mRNAs and proteins in DAL-1 overexpressed or knockdown cells. DAL-1 combined proteins were assessed by co-immunoprecipitation. RESULTS DAL-1 levels were strongly reduced even lost in lymph node metastasis and advanced pathological stage of human lung carcinomas. Overexpression of DAL-1 altered the expression of numerous EMT markers, such as E-cadherin, β-catenin Vimentin and N-cadherin expression, meanwhile changed the morphological shape of lung cancer cells, and whereas silencing DAL-1 had an opposite effect. DAL-1 directly combined with E-cadherin promoter and regulated its expression that could be the reason for impairing EMT and decreasing cell migration and invasion. Strikingly, HSPA5 was found as DAL-1 direct binding protein. CONCLUSIONS These results suggest that tumor suppressor DAL-1 could also attenuate EMT and be important for tumor metastasis in the early transformation process in lung cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yajie Zhang
- Department of Pathology, School of Basic Medical Science, Guangzhou Medical University, 195# Dongfeng West Road, Guangzhou 510180, Guangdong, People's Republic of China.
| |
Collapse
|