1
|
Ding LJ, Jiang X, Li T, Wang S. Role of UFMylation in tumorigenesis and cancer immunotherapy. Front Immunol 2024; 15:1454823. [PMID: 39247188 PMCID: PMC11377280 DOI: 10.3389/fimmu.2024.1454823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Protein post-translational modifications (PTMs) represent a crucial aspect of cellular regulation, occurring after protein synthesis from mRNA. These modifications, which include phosphorylation, ubiquitination, acetylation, methylation, glycosylation, Sumoylation, and palmitoylation, play pivotal roles in modulating protein function. PTMs influence protein localization, stability, and interactions, thereby orchestrating a variety of cellular processes in response to internal and external stimuli. Dysregulation of PTMs is linked to a spectrum of diseases, such as cancer, inflammatory diseases, and neurodegenerative disorders. UFMylation, a type of PTMs, has recently gained prominence for its regulatory role in numerous cellular processes, including protein stability, response to cellular stress, and key signaling pathways influencing cellular functions. This review highlights the crucial function of UFMylation in the development and progression of tumors, underscoring its potential as a therapeutic target. Moreover, we discuss the pivotal role of UFMylation in tumorigenesis and malignant progression, and explore its impact on cancer immunotherapy. The article aims to provide a comprehensive overview of biological functions of UFMylation and propose how targeting UFMylation could enhance the effectiveness of cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Li-Juan Ding
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Te Li
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Shudong Wang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
2
|
Boutaud M, Auger C, Verdier M, Christou N. Metformin Treatment Reduces CRC Aggressiveness in a Glucose-Independent Manner: An In Vitro and Ex Vivo Study. Cancers (Basel) 2023; 15:3724. [PMID: 37509386 PMCID: PMC10378121 DOI: 10.3390/cancers15143724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
(1) Background: Metformin, an anti-diabetic drug, seems to protect against aggressive acquisition in colorectal cancers (CRCs). However, its mechanisms are still really unknown, raising questions about the possibility of its positive impact on non-diabetic patients with CRC. (2) Methods: An in vitro study based on human colon cancer cell lines and an ex vivo study with different colon cancer stages with proteomic and transcriptomic analyses were initiated. (3) Results: Metformin seems to protect from colon cancer invasive acquisition, irrespective of glucose concentration. (4) Conclusions: Metformin could be used as an adjuvant treatment to surgery for both diabetic and non-diabetic patients in order to prevent the acquisition of aggressiveness and, ultimately, recurrences.
Collapse
Affiliation(s)
- Marie Boutaud
- UMR-INSERM 1308 CAPTuR, Faculté de Médecine, Institut OmegaHealth, Université de Limoges, 2 Rue du Dr Raymond Marcland, CEDEX, 87025 Limoges, France
| | - Clément Auger
- UMR-INSERM 1308 CAPTuR, Faculté de Médecine, Institut OmegaHealth, Université de Limoges, 2 Rue du Dr Raymond Marcland, CEDEX, 87025 Limoges, France
| | - Mireille Verdier
- UMR-INSERM 1308 CAPTuR, Faculté de Médecine, Institut OmegaHealth, Université de Limoges, 2 Rue du Dr Raymond Marcland, CEDEX, 87025 Limoges, France
| | - Niki Christou
- UMR-INSERM 1308 CAPTuR, Faculté de Médecine, Institut OmegaHealth, Université de Limoges, 2 Rue du Dr Raymond Marcland, CEDEX, 87025 Limoges, France
- Service de Chirurgie Digestive, Centre Hospitalier Universitaire de Limoges, 2 Av. Martin Luther King, CEDEX, 87000 Limoges, France
| |
Collapse
|
3
|
Cheng X, Zhang J, Xiao Y, Wang Z, He J, Ke M, Liu S, Wang Q, Zhang L. Mitochondrial Regulation of Ferroptosis in Cancer Therapy. Int J Mol Sci 2023; 24:10037. [PMID: 37373183 DOI: 10.3390/ijms241210037] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Ferroptosis, characterized by glutamate overload, glutathione depletion, and cysteine/cystine deprivation during iron- and oxidative-damage-dependent cell death, is a particular mode of regulated cell death. It is expected to effectively treat cancer through its tumor-suppressor function, as mitochondria are the intracellular energy factory and a binding site of reactive oxygen species production, closely related to ferroptosis. This review summarizes relevant research on the mechanisms of ferroptosis, highlights mitochondria's role in it, and collects and classifies the inducers of ferroptosis. A deeper understanding of the relationship between ferroptosis and mitochondrial function may provide new strategies for tumor treatment and drug development based on ferroptosis.
Collapse
Affiliation(s)
- Xiaoxia Cheng
- School of Basic Medical Science, Henan University, Kaifeng 475004, China
| | - Jiale Zhang
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Yichen Xiao
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Zhihang Wang
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Jin He
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Mengquan Ke
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Sijie Liu
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Qun Wang
- School of Basic Medical Science, Henan University, Kaifeng 475004, China
| | - Lei Zhang
- School of Basic Medical Science, Henan University, Kaifeng 475004, China
| |
Collapse
|
4
|
Zhong J, Shen X, Zhou J, Yu H, Wang B, Sun J, Wang J, Liu F. Development and validation of a combined hypoxia and ferroptosis prognostic signature for breast cancer. Front Oncol 2023; 13:1077342. [PMID: 36998462 PMCID: PMC10043308 DOI: 10.3389/fonc.2023.1077342] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundHypoxia is involved in tumor biological processes and disease progression. Ferroptosis, as a newly discovered programmed cell death process, is closely related to breast cancer (BC) occurrence and development. However, reliable prognostic signatures based on a combination of hypoxia and ferroptosis in BC have not been developed.MethodWe set The Cancer Genome Atlas (TCGA) breast cancer cohort as training set and the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) BC cohort as the validation set. Least Absolute Shrinkage and Selection Operator (LASSO) and COX regression approaches were used to construct ferroptosis-related genes (FRGs) and hypoxia-related genes (HRGs) prognostic signature (HFRS). The CIBERSORT algorithm and ESTIMATE score were used to explore the relationship between HFRS and tumor immune microenvironment. Immunohistochemical staining was used to detect protein expression in tissue samples. A nomogram was developed to advance the clinical application of HFRS signature.ResultsTen ferroptosis-related genes and hypoxia-related genes were screened to construct the HFRS prognostic signature in TCGA BC cohort, and the predictive capacity was verified in METABRIC BC cohort. BC patients with high-HFRS had shorter survival time, higher tumor stage, and a higher rate of positive lymph node. Moreover, high HFRS was associated with high hypoxia, ferroptosis, and immunosuppression status. A nomogram that was constructed with age, stage, and HFRS signature showed a strong prognostic capability to predict overall survival (OS) for BC patients.ConclusionWe developed a novel prognostic model with hypoxia and ferroptosis-related genes to predict OS, and characterize the immune microenvironment of BC patients, which might provide new cures for clinical decision-making and individual treatment of BC patients.
Collapse
Affiliation(s)
- Jianxin Zhong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xi Shen
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junjie Zhou
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heping Yu
- Department of Thyroid and Breast Surgery, Wuhan Fourth Hospital, Wuhan, China
| | - Birong Wang
- Department of Thyroid and Breast Surgery, Wuhan Fourth Hospital, Wuhan, China
| | - Jianbin Sun
- Department of Thyroid and Breast Surgery, Wuhan Fourth Hospital, Wuhan, China
| | - Jing Wang
- Department of Thoracic Surgery, Wuhan Fourth Hospital, Wuhan, China
- *Correspondence: Jing Wang, ; Feng Liu,
| | - Feng Liu
- Department of Thyroid and Breast Surgery, Wuhan Fourth Hospital, Wuhan, China
- *Correspondence: Jing Wang, ; Feng Liu,
| |
Collapse
|
5
|
Wu XY, Xu WW, Huan XK, Wu GN, Li G, Zhou YH, Najafi M. Mechanisms of cancer cell killing by metformin: a review on different cell death pathways. Mol Cell Biochem 2023; 478:197-214. [PMID: 35771397 DOI: 10.1007/s11010-022-04502-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 06/08/2022] [Indexed: 01/17/2023]
Abstract
Cancer resistance to anti-tumour agents has been one of the serious challenges in different types of cancer treatment. Usually, an increase in the cell death markers can predict a higher rate of survival among patients diagnosed with cancer. By increasing the regulation of survival genes, cancer cells can display a higher resistance to therapy through the suppression of anti-tumour immunity and inhibition of cell death signalling pathways. Administration of certain adjuvants may be useful in order to increase the therapeutic efficiency of anti-cancer therapy through the stimulation of different cell death pathways. Several studies have demonstrated that metformin, an antidiabetic drug with anti-cancer properties, amplifies cell death mechanisms, especially apoptosis in a broad-spectrum of cancer cells. Stimulation of the immune system by metformin has been shown to play a key role in the induction of cell death. It seems that the induction or suppression of different cell death mechanisms has a pivotal role in either sensitization or resistance of cancer cells to therapy. This review explains the cellular and molecular mechanisms of cell death following anticancer therapy. Then, we discuss the modulatory roles of metformin on different cancer cell death pathways including apoptosis, mitotic catastrophe, senescence, autophagy, ferroptosis and pyroptosis.
Collapse
Affiliation(s)
- Xiao-Yu Wu
- Department of Surgical Oncology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Wen-Wen Xu
- Department of Gynaecology, The Affiliated Hospital of Nanjing University of Chinese Medi-Cine, Nanjing, 210029, Jiangsu, China
| | - Xiang-Kun Huan
- Department of Surgical Oncology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Guan-Nan Wu
- Department of Surgical Oncology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Gang Li
- Department of General Surgery, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Yu-Hong Zhou
- Digestive Endoscopy Center, The Affiliated Hospital of Nanjing University of Chinese Medi-Cine, Nanjing, 210029, Jiangsu, China.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
6
|
Gong C, Ji Q, Wu M, Tu Z, Lei K, Luo M, Liu J, Lin L, Li K, Li J, Huang K, Zhu X. Ferroptosis in tumor immunity and therapy. J Cell Mol Med 2022; 26:5565-5579. [DOI: 10.1111/jcmm.17529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Chuandong Gong
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Qiankun Ji
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Miaojing Wu
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Zewei Tu
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Kunjian Lei
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Min Luo
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Junzhe Liu
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Li Lin
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Kuangxun Li
- College of Queen Mary Nanchang University Nanchang China
| | - Jingying Li
- Department of Comprehensive Intensive Care Unit Second Affiliated Hospital of Nanchang University Nanchang China
| | - Kai Huang
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Xingen Zhu
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| |
Collapse
|
7
|
Metformin and Cancer, an Ambiguanidous Relationship. Pharmaceuticals (Basel) 2022; 15:ph15050626. [PMID: 35631452 PMCID: PMC9144507 DOI: 10.3390/ph15050626] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 01/27/2023] Open
Abstract
The deregulation of energetic and cellular metabolism is a signature of cancer cells. Thus, drugs targeting cancer cell metabolism may have promising therapeutic potential. Previous reports demonstrate that the widely used normoglycemic agent, metformin, can decrease the risk of cancer in type 2 diabetics and inhibit cell growth in various cancers, including pancreatic, colon, prostate, ovarian, and breast cancer. While metformin is a known adenosine monophosphate-activated protein kinase (AMPK) agonist and an inhibitor of the electron transport chain complex I, its mechanism of action in cancer cells as well as its effect on cancer metabolism is not clearly established. In this review, we will give an update on the role of metformin as an antitumoral agent and detail relevant evidence on the potential use and mechanisms of action of metformin in cancer. Analyzing antitumoral, signaling, and metabolic impacts of metformin on cancer cells may provide promising new therapeutic strategies in oncology.
Collapse
|
8
|
Aoun R, El Hadi C, Tahtouh R, El Habre R, Hilal G. Microarray analysis of breast cancer gene expression profiling in response to 2-deoxyglucose, metformin, and glucose starvation. Cancer Cell Int 2022; 22:123. [PMID: 35305635 PMCID: PMC8933915 DOI: 10.1186/s12935-022-02542-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is the most frequently diagnosed cancer in women. Altering glucose metabolism and its effects on cancer progression and treatment resistance is an emerging interest in BC research. For instance, combining chemotherapy with glucose-lowering drugs (2-deoxyglucose (2-DG), metformin (MET)) or glucose starvation (GS) has shown better outcomes than with chemotherapy alone. However, the genes and molecular mechanisms that govern the action of these glucose deprivation conditions have not been fully elucidated. Here, we investigated the differentially expressed genes in MCF-7 and MDA-MB-231 BC cell lines upon treatment with glucose-lowering drugs (2-DG, MET) and GS using microarray analysis to study the difference in biological functions between the glucose challenges and their effect on the vulnerability of BC cells. METHODS MDA-MB-231 and MCF-7 cells were treated with 20 mM MET or 4 mM 2-DG for 48 h. GS was performed by gradually decreasing the glucose concentration in the culture medium to 0 g/L, in which the cells remained with fetal bovine serum for one week. Expression profiling was carried out using Affymetrix Human Clariom S microarrays. Differentially expressed genes were obtained from the Transcriptome Analysis Console and enriched using DAVID and R packages. RESULTS Our results showed that MDA-MB-231 cells were more responsive to glucose deprivation than MCF-7 cells. Endoplasmic reticulum stress response and cell cycle inhibition were detected after all three glucose deprivations in MDA-MB-231 cells and only under the metformin and GS conditions in MCF-7 cells. Induction of apoptosis and inhibition of DNA replication were observed with all three treatments in MDA-MB-231 cells and metformin-treated MCF-7 cells. Upregulation of cellular response to reactive oxygen species and inhibition of DNA repair mechanisms resulted after metformin and GS administration in MDA-MB-231 cell lines and metformin-treated MCF-7 cells. Autophagy was induced after 2-DG treatment in MDA-MB-231 cells and after metformin in MCF-7 cells. Finally, inhibition of DNA methylation were observed only with GS in MDA-MB-231 cells. CONCLUSION The procedure used to process cancer cells and analyze their expression data distinguishes our study from others. GS had the greatest effect on breast cancer cells compared to 2-DG and MET. Combining MET and GS could restrain both cell lines, making them more vulnerable to conventional chemotherapy.
Collapse
Affiliation(s)
- Rita Aoun
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | | | - Roula Tahtouh
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Rita El Habre
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - George Hilal
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon.
| |
Collapse
|
9
|
Zeng J, Yi J, Tan S, Zeng Y, Zou L, Zhang C, Liu L, Yi P, Fan P, Yu J. GPI: An indicator for immune infiltrates and prognosis of human breast cancer from a comprehensive analysis. Front Endocrinol (Lausanne) 2022; 13:995972. [PMID: 36246907 PMCID: PMC9554491 DOI: 10.3389/fendo.2022.995972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/01/2022] [Indexed: 11/15/2022] Open
Abstract
Glucose-6-phosphate isomerase (GPI) plays an important part in gluconeogenesis and glycolysis through the interconversion of d-glucose-6-phosphate and d-fructose-6-phosphate, and its clinical significance still remains unclear in breast cancer (BRCA). We analyzed the expressions of GPI in BRCA patients to determine prognostic values. Our results showed that the expression levels of GPI were upregulated in BRCA patients, and a high GPI expression is correlated with poor overall survival (OS) in BRCA. At the same time, a high GPI expression is correlated with poor clinicopathological characteristics, such as stage III, over 60 years old, N3, HER2 negative, and estrogen receptor (ER) positive. Further analysis of the influence of GPI on the prognosis of BRCA suggested that 50 genes and 10 proteins were positively correlated with GPI, and these genes and proteins were mainly involved in cell cycle signaling pathways. In addition, in this study, we observed that GPI was closely related to N 6-methyladenosine (m6A) RNA methylation modification and immune cell infiltration and ferroptosis-related gene expression in BRCA, and there was a difference in m6A RNA methylation alterations, immune cell infiltration, and ferroptosis-related gene expression between the high GPI expression group and the low GPI expression group. Finally, we found that GPI in BRCA had 2.6% gene alterations, and BRCA patients with gene alteration of GPI had a poor prognosis in disease-free survival (DFS). Altogether, our work strongly suggested that GPI may serve as a new prognostic biomarker for BRCA patients.
Collapse
Affiliation(s)
- Jie Zeng
- Department of Breast and Thyroid Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Jianing Yi
- Department of Breast and Thyroid Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Siyi Tan
- Department of Medical Laboratory, Huazhi Medical Laboratory Co., Ltd, Changsha, China
| | - Yuanjun Zeng
- Department of Pathology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Lianhong Zou
- Institute of Translational Medicine, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Chaojie Zhang
- Department of Breast and Thyroid Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Luyao Liu
- Department of Breast and Thyroid Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Pingyong Yi
- Department of Oncology, Changsha Jing Kai Hospital, Changsha, China
| | - Peizhi Fan
- Department of Breast and Thyroid Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
- *Correspondence: Jie Yu, ; Peizhi Fan,
| | - Jie Yu
- Department of Breast and Thyroid Surgery, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
- *Correspondence: Jie Yu, ; Peizhi Fan,
| |
Collapse
|
10
|
Noser AA, Abdelmonsef AH, El-Naggar M, Salem MM. New Amino Acid Schiff Bases as Anticancer Agents via Potential Mitochondrial Complex I-Associated Hexokinase Inhibition and Targeting AMP-Protein Kinases/mTOR Signaling Pathway. Molecules 2021; 26:molecules26175332. [PMID: 34500765 PMCID: PMC8434356 DOI: 10.3390/molecules26175332] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022] Open
Abstract
Two series of novel amino acid Schiff base ligands containing heterocyclic moieties, such as quinazolinone 3–11 and indole 12–20 were successfully synthesized and confirmed by spectroscopic techniques and elemental analysis. Furthermore, all compounds were investigated in silico for their ability to inhibit mitochondrial NADH: ubiquinone oxidoreductase (complex I) by targeting the AMPK/mTOR signaling pathway and inhibiting hexokinase, a key glycolytic enzyme to prevent the Warburg effect in cancer cells. This inhibitory pathway may be an effective strategy to cause cancer cell death due to an insufficient amount of ATP. Our results revealed that, out of 18 compounds, two (11 and 20) were top-ranked as they exhibited the highest binding energies of −8.8, −13.0, −7.9, and −10.0 kcal/mol in the docking analysis, so they were then selected for in vitro assessment. Compound 11 promoted the best cytotoxic effect on MCF-7 with IC50 = 64.05 ± 0.14 μg/mL (0.135 mM) while compound 20 exhibited the best cytotoxic effect on MDA-231 with IC50 = 46.29 ± 0.09 μg/mL (0.166 mM) Compounds 11 and 20 showed significant activation of AMPK protein and oxidative stress, which led to elevated expression of p53 and Bax, reduced Bcl-2 expression, and caused cell cycle arrest at the sub-G0/G1 phase. Moreover, compounds 11 and 20 showed significant inhibition of the mTOR protein, which led to the activation of aerobic glycolysis for survival. This alternative pathway was also blocked as compounds 11 and 20 showed significant inhibitory effects on the hexokinase enzyme. These findings demonstrate that compounds 11 and 20 obeyed Lipinski’s rule of five and could be used as privileged scaffolds for cancer therapy via their potential inhibition of mitochondrial complex I-associated hexokinase.
Collapse
Affiliation(s)
- Ahmed A. Noser
- Organic Chemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt;
| | - Aboubakr H. Abdelmonsef
- Chemistry Department, Faculty of Science, South Valley University, Qena 83523, Egypt
- Correspondence: ; Tel.: +20-10-989-65494
| | - Mohamed El-Naggar
- Chemistry Department, Faculty of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates;
| | - Maha M. Salem
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt;
| |
Collapse
|
11
|
New Insight into the Effects of Metformin on Diabetic Retinopathy, Aging and Cancer: Nonapoptotic Cell Death, Immunosuppression, and Effects beyond the AMPK Pathway. Int J Mol Sci 2021; 22:ijms22179453. [PMID: 34502359 PMCID: PMC8430477 DOI: 10.3390/ijms22179453] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
Under metabolic stress conditions such as hypoxia and glucose deprivation, an increase in the AMP:ATP ratio activates the AMP-activated protein kinase (AMPK) pathway, resulting in the modulation of cellular metabolism. Metformin, which is widely prescribed for type 2 diabetes mellitus (T2DM) patients, regulates blood sugar by inhibiting hepatic gluconeogenesis and promoting insulin sensitivity to facilitate glucose uptake by cells. At the molecular level, the most well-known mechanism of metformin-mediated cytoprotection is AMPK pathway activation, which modulates metabolism and protects cells from degradation or pathogenic changes, such as those related to aging and diabetic retinopathy (DR). Recently, it has been revealed that metformin acts via AMPK- and non-AMPK-mediated pathways to exert effects beyond those related to diabetes treatment that might prevent aging and ameliorate DR. This review focuses on new insights into the anticancer effects of metformin and its potential modulation of several novel types of nonapoptotic cell death, including ferroptosis, pyroptosis, and necroptosis. In addition, the antimetastatic and immunosuppressive effects of metformin and its hypothesized mechanism are also discussed, highlighting promising cancer prevention strategies for the future.
Collapse
|
12
|
Incorporation of Sulfonamide Moiety into Biguanide Scaffold Results in Apoptosis Induction and Cell Cycle Arrest in MCF-7 Breast Cancer Cells. Int J Mol Sci 2021; 22:ijms22115642. [PMID: 34073245 PMCID: PMC8198066 DOI: 10.3390/ijms22115642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 12/18/2022] Open
Abstract
Metformin, apart from its glucose-lowering properties, has also been found to demonstrate anti-cancer properties. Anti-cancer efficacy of metformin depends on its uptake in cancer cells, which is mediated by plasma membrane monoamine transporters (PMAT) and organic cation transporters (OCTs). This study presents an analysis of transporter mediated cellular uptake of ten sulfonamide-based derivatives of metformin in two breast cancer cell lines (MCF-7 and MDA-MB-231). Effects of these compounds on cancer cell growth inhibition were also determined. All examined sulfonamide-based analogues of metformin were characterized by greater cellular uptake in both MCF-7 and MDA-MB-231 cells, and stronger cytotoxic properties than those of metformin. Effective intracellular transport of the examined compounds in MCF-7 cells was accompanied by high cytotoxic activity. For instance, compound 2 with meta-methyl group in the benzene ring inhibited MCF-7 growth at micromolar range (IC50 = 87.7 ± 1.18 µmol/L). Further studies showed that cytotoxicity of sulfonamide-based derivatives of metformin partially results from their ability to induce apoptosis in MCF-7 and MDA-MB-231 cells and arrest cell cycle in the G0/G1 phase. In addition, these compounds were found to inhibit cellular migration in wound healing assay. Importantly, the tested biguanides are more effective in MCF-7 cells at relatively lower concentrations than in MDA-MB-231 cells, which proves that the effectiveness of transporter-mediated accumulation in MCF-7 cells is related to biological effects, including MCF-7 cell growth inhibition, apoptosis induction and cell cycle arrest. In summary, this study supports the hypothesis that effective transporter-mediated cellular uptake of a chemical molecule determines its cytotoxic properties. These results warrant a further investigation of biguanides as putative anti-cancer agents.
Collapse
|
13
|
Zhang X, Su Q, Zhou J, Yang Z, Liu Z, Ji L, Gao H, Jiang G. To betray or to fight? The dual identity of the mitochondria in cancer. Future Oncol 2021; 17:723-743. [DOI: 10.2217/fon-2020-0362] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are highly dynamic organelles that provide energy for oxidative phosphorylation in cells. Equally, they are the major sites for the metabolism of amino acids, lipids and iron. When cells become cancerous, the morphology, cellular location and metabolic mode of the mitochondria change accordingly. These mitochondrial changes can have two opposing effects on cancer: procancer and anticancer effects. Specifically, mitochondria play roles in the fight against cancer by participating in processes such as ferroptosis, mitophagy and antitumor immunity. Contrastingly, cancer cells can also enslave mitochondria to give them the conditions necessary for growth and metastasis. Moreover, through mitochondria, cancer cells can escape from immune surveillance, resulting in their immune escape and enhanced malignant transformation ability. At present, cancer-related studies of mitochondria are one-sided; therefore, we aim to provide a comprehensive understanding by systematically reviewing the two-sided cancer-related properties of mitochondria. Mitochondrial-targeted drugs are gradually emerging and showing significant advantages in cancer treatment; thus, our in-depth exploration of mitochondria in cancer will help to provide theoretical support for the future provision of efficient and low-toxicity cancer treatments.
Collapse
Affiliation(s)
- Xiaoyi Zhang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266021, PR China
| | - Quanzhong Su
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266021, PR China
| | - Ji Zhou
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266021, PR China
| | - Zhihong Yang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266021, PR China
| | - Zhantao Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266021, PR China
| | - Lixia Ji
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266021, PR China
| | - Hui Gao
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266021, PR China
| | - Guohui Jiang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266021, PR China
| |
Collapse
|