1
|
Pandey A, Goswami A, Jithin B, Shukla S. Autophagy: The convergence point of aging and cancer. Biochem Biophys Rep 2025; 42:101986. [PMID: 40224538 PMCID: PMC11986642 DOI: 10.1016/j.bbrep.2025.101986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Autophagy, a dynamic intracellular degradation system, is critical for cellular renovation and maintaining equilibrium. By eliminating damaged components and recycling essential molecules, autophagy safeguards cellular integrity and function. The versatility of the autophagy process across various biological functions enable cells to adapt and maintain homeostasis under unfavourable conditions. Disruptions in autophagy can shift a cell from a healthy state to a disease state or, conversely, support a return to health. This review delves into the multifaceted role of autophagy during aging and age-related diseases such as cancer, highlighting its significance as a unifying target with promising therapeutic implications. Cancer development is a dynamic process characterized by the acquisition of diverse survival capabilities for proliferating at different stages. This progression unfolds over time, with cancer cells exploiting autophagy to overcome encountered stress conditions during tumor development. Notably, there are several common pathways that utilize the autophagy process during aging and cancer development. This highlights the importance of autophagy as a crucial therapeutic target, holding the potential to not only impede the growth of tumor but also enhance the patient's longevity. This review aims to simplify the intricate relationship between cancer and aging, with a particular focus on the role of autophagy.
Collapse
Affiliation(s)
- Anchala Pandey
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
| | | | | | - Sanjeev Shukla
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
| |
Collapse
|
2
|
Kumar P, Choudhary A, Kinger S, Jagtap YA, Prajapati VK, Chitkara D, Chinnathambi S, Verma RK, Mishra A. Autophagy as a potential therapeutic target in regulating improper cellular proliferation. Front Pharmacol 2025; 16:1579183. [PMID: 40444035 PMCID: PMC12119615 DOI: 10.3389/fphar.2025.1579183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/24/2025] [Indexed: 06/02/2025] Open
Abstract
Autophagy is a degradative process that makes rapid turnover of old and impaired proteins and organelles possible. It is highly instigated by stress signals, like starvation, and contributes to the cell's homeostasis. Autophagy performs a crucial function in keeping cell genomic integrity stable. Impaired autophagic flux is implicated in neurodegenerative diseases, abnormal ageing, and cancerous diseases. In diseases like cancer, autophagy performs a dualistic function; it can have both a tumor-suppressive and supportive role. Autophagy in the initial phases of tumorigenesis maintains the integrity of the genome and, if it fails, leads to cell death, thus having a tumor-suppressive role. Meanwhile, autophagy also imparts the function of the pro-survival mechanism in the latter stages of tumorigenesis and supports the cancerous cells in surviving conditions like hypoxia and increased oxidative stress. Autophagy also helps cancerous cells develop drug resistance in some cases. Thus, modulation of the autophagic mechanism is a possible therapeutic strategy in cancer therapy as its inhibition can sensitise cancer cells to anti-cancerous drugs. The promotion of autophagy, in some cases, can also safeguard cells from toxic protein aggregation and enhanced oxidative stress. Excessive autophagy can result in autophagic cell death. Autophagy also regulates several cellular processes and cell death pathways, like apoptosis. Therefore, an in-depth knowledge of the autophagy process and its regulating molecules is critically important. Pharmaceutical small molecules or cellular target modulation can help modulate the cellular autophagy process in the context of specific disease conditions.
Collapse
Affiliation(s)
- Prashant Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Akash Choudhary
- Department of Biosciences and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Sumit Kinger
- Department of Biosciences and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Yuvraj Anandrao Jagtap
- Department of Biosciences and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | | | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani, Rajasthan, India
| | - Subashchandrabose Chinnathambi
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences, Institute of National Importance, Bangalore, Karnataka, India
| | | | - Amit Mishra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| |
Collapse
|
3
|
Yu JY, Hsieh BY, Tsai SF, Hsieh M. Effects of p53 on the Regulation of Carbonic Anhydrase 8 in Human Colorectal Cancer Cells: Interaction Between p53 and Sp1. Cell Biochem Funct 2025; 43:e70081. [PMID: 40387560 DOI: 10.1002/cbf.70081] [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: 08/28/2024] [Revised: 04/25/2025] [Accepted: 05/01/2025] [Indexed: 05/20/2025]
Abstract
Colorectal cancer ranks among the most common malignancies worldwide. Our previous study indicated Carbonic anhydrase 8 (CA8) is linked to cell proliferation and mobility in colon cancer cells. In the present study, we observed a significant increase in the expression of mutant p53 R273H/P309S in colon cancer cell lines (SW480 and SW620) with stably downregulated CA8. P53, a well-known tumor suppressor gene, is frequently mutated in cancer cells, leading to poor prognosis and drug resistance. Although p53 acts as a transcription factor, the increased mutant p53 did not activate downstream target molecules, suggesting activation defects of mutant p53 R273H/P309S. Furthermore, transient downregulation of CA8 did not alter p53 expression, indicating that the observed increase in mutant p53 in stable cells may be a compensatory effect for cell survival. Given that p53 shares similar consensus sequences at GC-boxes with specific protein 1 (Sp1), a predominant transcription factor for CA8 regulation, we examined the relationship between CA8, p53 and Sp1 in HCT116 and SW620 cells harboring wild-type (WT) or mutant p53, respectively. Notably, transient downregulation of p53 or Sp1 led to a significant decrease in CA8 at both mRNA and protein levels in HCT116 and SW620 cells. Additionally, immunoprecipitation results revealed a protein-protein interaction between Sp1 and p53, suggesting that their interaction may be involved in the regulation of CA8 expression. Although the precise mechanism by which Sp1 and p53 regulate CA8 expression remains unclear, we are the first to report that both Sp1 and p53 are involved in the regulation of the novel hCA8 gene. By further unraveling the interplay among CA8, p53, and Sp1, we hope to pave the way for new therapeutic approaches in colon cancer treatment.
Collapse
Affiliation(s)
- Jia-Yo Yu
- Department of Life Science, Tunghai University, Taichung, Taiwan, Republic of China
| | - Benjamin Y Hsieh
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Shang-Feng Tsai
- Department of Life Science, Tunghai University, Taichung, Taiwan, Republic of China
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
- Department of Postbaccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Mingli Hsieh
- Department of Life Science, Tunghai University, Taichung, Taiwan, Republic of China
- Life Science Research Center, Tunghai University, Taichung, Taiwan, Republic of China
| |
Collapse
|
4
|
Lazovic A, Markovic BS, Corovic I, Markovic T, Andjelkovic M, Stojanovic B, Jovanovic I, Mitrovic M. Unlocking the molecular mechanisms of anticancer and immunomodulatory potentials of cariprazine in triple negative breast cancer. Biomed Pharmacother 2025; 184:117931. [PMID: 39978031 DOI: 10.1016/j.biopha.2025.117931] [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: 11/04/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/22/2025] Open
Abstract
Triple-negative breast cancer (TNBC), a highly invasive type of cancer, is difficult to treat due to insufficient specific targets and low survival rates. Current therapy often encounters drug resistance or relapse; thus, repurposing existing drugs could revolutionize cancer treatment. This study examined the anticancer effects of the antipsychotics Cariprazine (CAR), Olanzapine (OLZ), and Clozapine (CLZ), and the immunomodulatory potential of CAR, in vitro and in vivo in TNBC models. In vitro, CAR, OLZ, and CLZ significantly inhibited the proliferation of TNBC cells. This inhibition occurred via the induction of mitochondrial apoptosis, G0/G1 cell cycle arrest, and the suppression of autophagy, as evidenced by the down-regulation of Bcl-2, p62, and pAKT; the upregulation of Bax and active caspase 3; the decrease of ΔΨM; and the promotion of cytochrome c release. In addition, CAR inhibited MDA-MB-231 cells migration. In vivo, CAR inhibited tumor growth in the 4T1 xenograft model without causing adverse effects and resulted in the mRNA upregulation caspase 9, p53, p21, and Beclin-1. In addition, CAR influenced the immune response by promoting the production of proinflammatory cytokines TNF-α, IFN-γ, IL-17, and IL-1β and increasing the percentage of TNF-α+, IL-17+, IL-1β+, and IFN-γ+ CD3+ splenocytes. In conclusion, compared with other antipsychotics, 5-FU, and cisplatin, CAR exerted the most potent anticancer activity in TNBC in vitro and in vivo. This efficacy may be attributed to its ability to regulate apoptosis and autophagy, promote G0/G1 cell cycle arrest, and modulate antitumor immune response, suggesting its therapeutic potential in breast cancer.
Collapse
Affiliation(s)
- Aleksandar Lazovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia.
| | - Bojana Simovic Markovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia.
| | - Irfan Corovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia; General Hospital of Novi Pazar, Department of Internal Medicine, General Živković 1, Novi Pazar 36300, Serbia.
| | - Tijana Markovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia.
| | - Marija Andjelkovic
- Department of Medical Biochemistry, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića, 69, Kragujevac 34000, Serbia.
| | - Bojan Stojanovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia.
| | - Ivan Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia.
| | - Marina Mitrovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozar Markovic 69, Kragujevac 34000, Serbia; Department of Medical Biochemistry, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića, 69, Kragujevac 34000, Serbia.
| |
Collapse
|
5
|
Torner B, Géczi D, Klekner Á, Balogh I, Penyige A, Birkó Z. Construction of a miRNA Panel for Differentiating Lung Adenocarcinoma Brain Metastases and Glioblastoma. Cancers (Basel) 2025; 17:581. [PMID: 40002176 PMCID: PMC11853152 DOI: 10.3390/cancers17040581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Brain metastases (BM) are the most common type of intracranial malignant tumor and are associated with high mortality. More than 50% of BM cases originate from lung cancer, and lung adenocarcinoma (LUAD) is most commonly associated with the development of BM (25%). The differential diagnosis of solitary BM and glioblastoma (GBM)-one of the most aggressive and fatal malignant brain tumors-remains a considerable challenge. Given the major role of microRNAs (miRNAs) in regulating gene expression, their clinical potential as biomarkers for tumor diagnosis and prognosis offers significant promise. METHODS Next-generation RNA Sequencing (RNA-seq) was used to assess the miRNA expression profiles of 6 LUAD-BM, 6 GBM, and 6 control (non-tumoral brain tissue samples) human brain tissue samples. miRNAs exhibiting the most significant differential expression in LUAD-BM patients in comparison to both control subjects and GBM patients were selected for validation through RT-qPCR. RESULTS The analysis of RNA-seq data revealed the presence of 229 differentially expressed miRNAs in the comparison between LUAD-BM and control samples and 46 in the comparison between LU-AD-BM and GBM samples. Eight miRNAs were selected for further analysis, four of which were upregulated and four downregulated, based on the significant differences in their expression levels observed between the LUAD-BM samples and the other two groups, as confirmed with the Mann-Whitney U test. Functional enrichment analysis was also conducted based on a miRNA-centered target analysis performed using the miRNet tool. To assess the diagnostic potential of these differentially expressed miRNAs, we performed a receiver operating characteristic (ROC) curve analysis. CONCLUSIONS A panel of eight miRNAs was identified in human brain tissue samples, exhibiting high accuracy in distinguishing LUAD-BM from both GBM and control samples.
Collapse
Affiliation(s)
- Bernadett Torner
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (B.T.); (D.G.); (I.B.); (A.P.)
| | - Dóra Géczi
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (B.T.); (D.G.); (I.B.); (A.P.)
| | - Álmos Klekner
- Department of Neurosurgery, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - István Balogh
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (B.T.); (D.G.); (I.B.); (A.P.)
- Division of Clinical Genetics, Department of Laboratory Medicine, University of Debrecen Clinical Center, 4032 Debrecen, Hungary
| | - András Penyige
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (B.T.); (D.G.); (I.B.); (A.P.)
| | - Zsuzsanna Birkó
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (B.T.); (D.G.); (I.B.); (A.P.)
| |
Collapse
|
6
|
Fan Y, Li Y, Luo X, Xiang S, Hu J, Zhan J, Chang W, Deng R, Ran X, Zhang Y, Cai Y, Zhu W, Wang H, Liu Z, Wang D. PTOV1 exerts pro-oncogenic role in colorectal cancer by modulating SQSTM1-mediated autophagic degradation of p53. J Transl Med 2025; 23:157. [PMID: 39905441 PMCID: PMC11796032 DOI: 10.1186/s12967-025-06179-x] [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: 11/09/2024] [Accepted: 01/25/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Prostate Tumor Overexpressed 1 (PTOV1) is overexpressed and associated with malignant phenotypes in various types of tumors. However, the detailed roles of PTOV1 and its underlying mechanism in CRC remain unclear. METHODS The clinical significance of PTOV1 was assessed in clinical databases and CRC samples. The effects of PTOV1 on the tumor-associated phenotypes of CRC were detected by several in vitro assays and in vivo mouse models. Immunoprecipitation (IP) combined with protein mass spectrometry and Co-Immunoprecipitation (Co-IP) was used to identify p53 interacting with PTOV1. Immunofluorescence assay, western blot and transmission electron microscopy (TEM) analysis were used to evaluated the effects of PTOV1 on autophagy. RESULTS Here, we revealed that PTOV1 was highly expressed in human CRC tissues, especially at advanced stages, and associated with reduced survival time among CRC patients. The upregulated PTOV1 promoted cell proliferation, migration, invasion, tumor growth and metastasis of CRC cells in vitro and in vivo. At the molecular level, PTOV1 destabilized p53 by activating autophagy and recruiting p53 for the cargo receptor SQSTM1 directed autophagic degradation. There was a negative expression correlation between PTOV1 and p53 in CRC tissues. Moreover, p53 overexpression or SQSTM1 knockdown reversed the pro-tumor phenotypes of PTOV1 in CRC. CONCLUSION Our study unveils the oncogenic role of PTOV1 in CRC progression, which was achieved by promoting SQSTM1 directed autophagic degradation of p53. These findings highlight the potential of targeting the PTOV1-SQSTM1-p53 axis as a therapeutic approach for CRC.
Collapse
Affiliation(s)
- Yongli Fan
- Department of Oncology, the First Affiliated Hospital of Henan University, Kaifeng, 475000, China
| | - Yuqin Li
- Department of Medical Laboratory, Tongji Medical College, the Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xia Luo
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shiqi Xiang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Jia Hu
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Jingchun Zhan
- College of Anesthesiology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Weilong Chang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Rui Deng
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xianwen Ran
- Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yize Zhang
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yudie Cai
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Weiwei Zhu
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Huifen Wang
- Gene Hospital of Henan Province, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Zhibo Liu
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Di Wang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
7
|
Hou W, Xiao C, Zhou R, Yao X, Chen Q, Xu T, Cao F, Wang Y, Li X, Yan O, Ai X, Yi C, Cao D, Zhao C. Inhibiting autophagy selectively prunes dysfunctional tumor vessels and optimizes the tumor immune microenvironment. Theranostics 2025; 15:258-276. [PMID: 39744218 PMCID: PMC11667230 DOI: 10.7150/thno.98285] [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: 05/10/2024] [Accepted: 10/30/2024] [Indexed: 03/17/2025] Open
Abstract
Dysfunctional tumor vasculature, hypoxia, and an immunosuppressive microenvironment are significant barriers to effective cancer therapy. Autophagy, which is critical for maintaining cellular homeostasis and apoptosis resistance, is primarily triggered by hypoxia and nutrient deprivation, conditions prevalent in dysfunctional tumor vessels due to poor circulation. However, the role of autophagy in dysfunctional tumor endothelial cells and its impact on treatment and the tumor microenvironment (TME) remain poorly understood. Methods: We used multiplex immunofluorescence and transgene-based imaging to characterize autophagy in endothelial cells from clinical tumor samples, zebrafish xenograft tumors, and murine models. Using a zebrafish xenograft vasculature platform, we analyzed the effects of autophagy inhibitors on the structure and function of the tumor vasculature. In mice, we investigated autophagy inhibition via endothelial-specific autophagy gene knockout (Atg7 iECKO) and the autophagy inhibitor SBI-0206965 and evaluated the synergistic effects of combining SBI-0206965 with low-dose chemotherapy (5-fluorouracil, 5-FU) or PD-1 antibody. Human umbilical vein endothelial cells (HUVECs) were cultured in vitro under hypoxic, glucose-deprived, and serum-free conditions to simulate dysfunctional tumor endothelial cells and to explore the mechanisms by which autophagy inhibition optimizes tumor vasculature. Results: Elevated autophagy was observed in tumor endothelial cells within the dysfunctional vasculature. Autophagy inhibition, through either genetic knockout or pharmacological inhibition, selectively prunes dysfunctional vessels and improves vascular function. It also stimulates the formation of a perivascular immune niche, thereby optimizing the tumor immune microenvironment (TiME). Furthermore, combining the autophagy inhibitor SBI-0206965 with low-dose 5-FU or PD-1 antibody potentiated the anti-tumor effects. Mechanistic studies have indicated that autophagy acts as a protective response to the hypoxic and nutrient-deprived TME, while its inhibition, mediated by p53 activation, promotes tumor endothelial cell apoptosis in dysfunctional tumor vessels, further optimizing the structure and function of the tumor vasculature. Conclusions: Targeting endothelial cell autophagy is a promising strategy for remodeling the dysfunctional tumor vasculature, optimizing the TiME, and boosting the efficacy of chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Wanting Hou
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Chaoxin Xiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, People's Republic of China
| | - Ruihan Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, People's Republic of China
| | - Xiaohong Yao
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Qin Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, People's Republic of China
| | - Tongtong Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, People's Republic of China
| | - Fujun Cao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, People's Republic of China
| | - Yulin Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, People's Republic of China
| | - Xiaoying Li
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Ouying Yan
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Xiaolin Ai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, People's Republic of China
| | - Cheng Yi
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Dan Cao
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Chengjian Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, People's Republic of China
| |
Collapse
|
8
|
Pizzimenti C, Fiorentino V, Ruggeri C, Franchina M, Ercoli A, Tuccari G, Ieni A. Autophagy Involvement in Non-Neoplastic and Neoplastic Endometrial Pathology: The State of the Art with a Focus on Carcinoma. Int J Mol Sci 2024; 25:12118. [PMID: 39596186 PMCID: PMC11594225 DOI: 10.3390/ijms252212118] [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: 09/27/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Autophagy is a cellular process crucial for maintaining homeostasis by degrading damaged proteins and organelles. It is stimulated in response to stress, recycling nutrients and generating energy for cell survival. In normal endometrium, it suppresses tumorigenesis by preventing toxic accumulation and maintaining cellular homeostasis. It is involved in the cyclic remodelling of the endometrium during the menstrual cycle and contributes to decidualisation for successful pregnancy. Such a process is regulated by various signalling pathways, including PI3K/AKT/mTOR, AMPK/mTOR, and p53. Dysregulation of autophagy has been associated with benign conditions like endometriosis and endometrial hyperplasia but also with malignant neoplasms such as endometrial carcinoma. In fact, it has emerged as a crucial player in endometrial carcinoma biology, exhibiting a dual role in both tumour suppression and tumour promotion, providing nutrients during metabolic stress and allowing cancer cell survival. It also regulates cancer stem cells, metastasis and therapy resistance. Targeting autophagy is therefore a promising therapeutic strategy in endometrial carcinoma and potential for overcoming resistance to standard treatments. The aim of this review is to delve into the intricate details of autophagy's role in endometrial pathology, exploring its mechanisms, signalling pathways and potential therapeutic implications.
Collapse
Affiliation(s)
- Cristina Pizzimenti
- Section of Pathology, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.P.); (V.F.); (M.F.)
| | - Vincenzo Fiorentino
- Section of Pathology, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.P.); (V.F.); (M.F.)
| | - Chiara Ruggeri
- Section of Gynecology and Obstetrics, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.R.); (A.E.)
| | - Mariausilia Franchina
- Section of Pathology, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.P.); (V.F.); (M.F.)
| | - Alfredo Ercoli
- Section of Gynecology and Obstetrics, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.R.); (A.E.)
| | - Giovanni Tuccari
- Section of Pathology, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.P.); (V.F.); (M.F.)
| | - Antonio Ieni
- Section of Pathology, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (C.P.); (V.F.); (M.F.)
| |
Collapse
|
9
|
Luo S, Xu J, Mo C, Gong W, Li C, Hou X, Ou M. High-throughput sequencing reveals twelve cell death pattern prognostic target genes as potential drug-response-associated genes in the treatment of colorectal cancer cells with palmatine hydrochloride. ONCOLOGIE 2024. [DOI: 10.1515/oncologie-2024-0378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Abstract
Objectives
Palmatine Hydrochloride (PaH), an isoquinoline alkaloid from Phellodendron amurense and Coptis chinensis, has analgesic, anti-inflammatory, and anticancer properties. This study aimed to assess PaH’s effectiveness against SW480 colorectal cancer (CRC) cells and explore its molecular mechanisms.
Methods
PaH’s effects on SW480 CRC cells were evaluated using MTT assays for proliferation, scratch assays for migration, and flow cytometry for apoptosis. Differentially expressed genes (DEGs) were identified through high-throughput sequencing. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses assessed DEG roles. Prognostic significance related to programmed cell death (PCD) was analyzed using R-Package with TCGA data. RT-qPCR validated key genes identified.
Results
PaH significantly inhibited SW480 cell growth, invasion, and apoptosis. The MTT assay showed inhibition rates increased from 5.49 % at 25 μg/mL to 52.48 % at 400 μg/mL. Scratch assays indicated reduced cell invasion over 24, 48, and 72 h. Apoptosis rose from 12.36 % in controls to 45.54 % at 400 μg/mL. Sequencing identified 3,385 significant DEGs, primarily in cancer pathways (p=0.004). Among 35 PCD-related DEGs, Lasso Cox regression highlighted 12 key genes, including TERT, TGFBR1, WNT4, and TP53. RT-qPCR confirmed TERT and TGFBR1 downregulation (0.614-fold, p=0.008; 0.41-fold, p<0.001) and TP53 and WNT4 upregulation (5.634-fold, p<0.001; 5.124-fold, p=0.002).
Conclusions
PaH inhibits CRC cell proliferation, migration, and invasion by modulating key PCD genes, suggesting its potential as a CRC therapeutic agent.
Collapse
Affiliation(s)
- Sha Luo
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Jiajun Xu
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Chune Mo
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Weiwei Gong
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Chunhong Li
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Xianliang Hou
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Minglin Ou
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| |
Collapse
|
10
|
Mmbando GS. Variation in ultraviolet-B (UV-B)-induced DNA damage repair mechanisms in plants and humans: an avenue for developing protection against skin photoaging. Int J Radiat Biol 2024; 100:1505-1516. [PMID: 39231421 DOI: 10.1080/09553002.2024.2398081] [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: 05/07/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 09/06/2024]
Abstract
PURPOSE The increasing amounts of ultraviolet-B (UV-B) light in our surroundings have sparked worries about the possible effects on humans and plants. The detrimental effects of heightened UV-B exposure on these two vital elements of terrestrial life are different due to their unique and concurrent nature. Understanding common vulnerabilities and distinctive adaptations of UV-B radiation by exploring the physiological and biochemical responses of plants and the effects on human health is of huge importance. The comparative effects of UV-B radiation on plants and animals, however, are poorly studied. This review sheds light on the sophisticated web of UV-B radiation effects by navigating the complex interaction between botanical and medical perspectives, drawing upon current findings. CONCLUSION By providing a comprehensive understanding of the complex effects of heightened UV-B radiation on plants and humans, this study summarizes relevant adaptation strategies to the heightened UV-B radiation stress, which offer new approaches for improving human cellular resilience to environmental stressors.
Collapse
Affiliation(s)
- Gideon Sadikiel Mmbando
- College of Natural and Mathematical Sciences, Department of Biology, The University of Dodoma, Dodoma, Tanzania
| |
Collapse
|
11
|
Yu CL, Huang KY, Chen JJ, Lai CT, Chen GW, Huang CC, Yeh YH, Lee CH, Lee JJ, Huang DM, Wang SW. Hernandonine-mediated autophagic cell death in hepatocellular carcinoma: Interplay of p53 and YAP signaling pathways. Free Radic Biol Med 2024; 222:456-466. [PMID: 38950659 DOI: 10.1016/j.freeradbiomed.2024.06.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 07/03/2024]
Abstract
Hepatocellular carcinoma (HCC), the primary form of liver cancer, is the third leading cause of cancer-related death globally. Hernandonine is a natural alkaloid derived from Hernandia nymphaeifolia that has been shown to exert various biological functions. In a previous study, hernandonine was shown to suppress the proliferation of several solid tumor cell lines without affecting normal human cell lines. However, little is known about the effect of hernandonine on HCC. Therefore, this study aimed to investigate the effect and mechanism of hernandonine on HCC in relation to autophagy. We found that hernandonine inhibited HCC cell growth in vitro and in vivo. In addition, hernandonine elicited autophagic cell death and DNA damage in HCC cells. RNA-seq analysis revealed that hernandonine upregulated p53 and Hippo signaling pathway-related genes in HCC cells. Small RNA interference of p53 resulted in hernandonine-induced autophagic cell death attenuation. However, inhibition of YAP sensitized HCC cells to hernandonine by increasing the autophagy induction. This is the first study to illustrate the complex involvement of p53 and YAP in the hernandonine-induced autophagic cell death in human HCC cells. Our findings provide novel evidence for the potential of hernandonine as a therapeutic agent for HCC treatment.
Collapse
Affiliation(s)
- Chen-Lin Yu
- Institute of Biomedical Science, MacKay Medical College, New Taipei City, Taiwan
| | - Kai-Yao Huang
- Department of Medical Research, Hsinchu MacKay Memorial Hospital, Hsinchu City, Taiwan
| | - Jih-Jung Chen
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Cheng-Ta Lai
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Guang-Wei Chen
- Department of Chinese Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chen-Chen Huang
- Institute of Biomedical Science, MacKay Medical College, New Taipei City, Taiwan
| | - Yen-Hsiu Yeh
- Department and Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Hsing Lee
- Department of Pharmacology, School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Taiwan
| | - Jie-Jen Lee
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Dong-Ming Huang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan.
| | - Shih-Wei Wang
- Institute of Biomedical Science, MacKay Medical College, New Taipei City, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, Taiwan; Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
12
|
Fares R, Elasmer SM, A. AK, Shaker OG, El-Tahlawi SM, Sabri A, Yaseen SM. Molecular Signature of miR-34a/NEAT-1/p53 Axis in Mycosis Fungoides. Dermatol Res Pract 2024; 2024:3163839. [PMID: 39184920 PMCID: PMC11343631 DOI: 10.1155/2024/3163839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024] Open
Abstract
Background Mycosis fungoides (MF) is a type of cutaneous T-cell lymphoma where red rash exists on the skin. Understanding the role of miRNAs and ncRNAs in p53-response has become an open discussion, as they can regulate p53 or its downstream targets, and ncRNAs themselves. Objectives To evaluate the serum levels of NEAT-1, miR-34a, and p53 in MF patients and its relation to healthy controls to indicate whether it has a potential role in the pathogenesis of the disease. Subjects and Methods. This prospective case-control study was carried out on 75 subjects subdivided into two groups, 35 MF patients (stages 1 and II) and 40 matched healthy controls. Their clinical investigations and serum biomarkers (NEAT-1, miR-34a, and p53) were measured. Results There were significant elevations in the expression levels of both NEAT-1 (5.10 ± 1.16) and p53 (277.28 ± 62.02) in the serum of MF patients in comparison with controls (1.01 ± 0.031) and (194.29 ± 16.039), respectively, while the level of miR-34a tends to decrease in MF patients (0.24 ± 0.15). There are no significant difference between MF stages and the level of miR-34a, while in NEAT-1 and p53, there are significant differences with p value <0.05 between the stages and the biomarkers. There is a positive correlation between the %BSA and miR-34a and a slightly positive correlation between NEAT-1 and P53 with (r = 0.353, p=0.037) and (r = 0112, p=0.05), respectively. There were also negative correlations between disease duration and NEAT-1 with (r = -0.341, p=0.045) and between B2 microglobulin level and p53 (r = -0.373, p=0.027). Conclusion The combination of miR-34a, NEAT-1, and p53 may be considered as potential biomarkers that play an active role in the disease process of MF for helping in its early diagnosis and stage identification as well.
Collapse
Affiliation(s)
- Reham Fares
- Department of Medical Biochemistry and Molecular BiologyFaculty of MedicineFayoum University, Fayoum, Egypt
| | - Shimaa M. Elasmer
- Department of Clinical and Chemical PathologyFaculty of MedicineFayoum University, Fayoum, Egypt
| | - Abeer Khalefa A.
- Department of PhysiologyFaculty of MedicineZagazig University, Zagazig, Egypt
| | - Olfat G. Shaker
- Department of Medical Biochemistry and Molecular BiologyFaculty of MedicineCairo University, Cairo, Egypt
| | | | | | - Sara M. Yaseen
- Department of Dermatology, STDs & AndrologyFaculty of MedicineFayoum University, Fayoum, Egypt
| |
Collapse
|
13
|
Rahman MA, Rakib-Uz-Zaman SM, Chakraborti S, Bhajan SK, Gupta RD, Jalouli M, Parvez MAK, Shaikh MH, Hoque Apu E, Harrath AH, Moon S, Kim B. Advancements in Utilizing Natural Compounds for Modulating Autophagy in Liver Cancer: Molecular Mechanisms and Therapeutic Targets. Cells 2024; 13:1186. [PMID: 39056768 PMCID: PMC11274515 DOI: 10.3390/cells13141186] [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: 04/16/2024] [Revised: 06/30/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Autophagy, an intrinsic catabolic mechanism that eliminates misfolded proteins, dysfunctional organelles, and lipid droplets, plays a vital function in energy balance and cytoplasmic quality control, in addition to maintaining cellular homeostasis. Liver cancer such as hepatocellular carcinoma (HCC) is one of the most common causes of cancer deaths globally and shows resistance to several anticancer drugs. Despite the rising incidence and poor prognosis of malignant HCC, the underlying molecular mechanisms driving this aggressive cancer remain unclear. Several natural compounds, such as phytochemicals of dietary and non-dietary origin, affect hepatocarcinogenesis signaling pathways in vitro and in vivo, which may help prevent and treat HCC cells. Current HCC cells treatments include chemotherapy, radiation, and surgery. However, these standard therapies have substantial side effects, and combination therapy enhances side effects for an acceptable therapeutic benefit. Therefore, there is a need to develop treatment strategies for HCC cells that are more efficacious and have fewer adverse effects. Multiple genetic and epigenetic factors are responsible for the HCC cells to become resistant to standard treatment. Autophagy contributes to maintain cellular homeostasis, which activates autophagy for biosynthesis and mitochondrial regulation and recycling. Therefore, modifying autophagic signaling would present a promising opportunity to identify novel therapies to treat HCC cells resistant to current standard treatments. This comprehensive review illustrates how natural compounds demonstrate their anti-hepatocellular carcinoma function through autophagy.
Collapse
Affiliation(s)
- Md Ataur Rahman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - S M Rakib-Uz-Zaman
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (S.M.R.-U.-Z.); (S.C.)
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, BRAC University, Dhaka 1212, Bangladesh
| | - Somdeepa Chakraborti
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (S.M.R.-U.-Z.); (S.C.)
| | - Sujay Kumar Bhajan
- Department of Biotechnology & Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science & Technology University, Gopalganj 8100, Bangladesh;
| | - Rajat Das Gupta
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA;
| | - Maroua Jalouli
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia;
| | | | - Mushfiq H. Shaikh
- Department of Otolaryngology-Head & Neck Surgery, Western University, London, ON N6A 4V2, Canada;
| | - Ehsanul Hoque Apu
- Department of Biomedical Sciences, College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA;
- DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN 37752, USA
- Division of Hematology and Oncology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Abdel Halim Harrath
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Seungjoon Moon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 1–5 Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea;
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 1–5 Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea;
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
14
|
Villagomez FR, Lang J, Rosario FJ, Nunez-Avellaneda D, Webb P, Neville M, Woodruff ER, Bitler BG. Claudin-4 Modulates Autophagy via SLC1A5/LAT1 as a Mechanism to Regulate Micronuclei. CANCER RESEARCH COMMUNICATIONS 2024; 4:1625-1642. [PMID: 38867360 PMCID: PMC11218812 DOI: 10.1158/2767-9764.crc-24-0240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/21/2024] [Accepted: 06/07/2024] [Indexed: 06/14/2024]
Abstract
Genome instability is a hallmark of cancer crucial for tumor heterogeneity and is often a result of defects in cell division and DNA damage repair. Tumors tolerate genomic instability, but the accumulation of genetic aberrations is regulated to avoid catastrophic chromosomal alterations and cell death. In ovarian cancer tumors, claudin-4 is frequently upregulated and closely associated with genome instability and worse patient outcomes. However, its biological association with regulating genomic instability is poorly understood. Here, we used CRISPR interference and a claudin mimic peptide to modulate the claudin-4 expression and its function in vitro and in vivo. We found that claudin-4 promotes a tolerance mechanism for genomic instability through micronuclei generation in tumor cells. Disruption of claudin-4 increased autophagy and was associated with the engulfment of cytoplasm-localized DNA. Mechanistically, we observed that claudin-4 establishes a biological axis with the amino acid transporters SLC1A5 and LAT1, which regulate autophagy upstream of mTOR. Furthermore, the claudin-4/SLC1A5/LAT1 axis was linked to the transport of amino acids across the plasma membrane as one of the potential cellular processes that significantly decreased survival in ovarian cancer patients. Together, our results show that the upregulation of claudin-4 contributes to increasing the threshold of tolerance for genomic instability in ovarian tumor cells by limiting its accumulation through autophagy. SIGNIFICANCE Autophagy regulation via claudin-4/SLC1A5/LAT1 has the potential to be a targetable mechanism to interfere with genomic instability in ovarian tumor cells.
Collapse
Affiliation(s)
- Fabian R. Villagomez
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Julie Lang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Fredrick J. Rosario
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Daniel Nunez-Avellaneda
- Deputy Directorate of Technological Development, Linkage, and Innovation, National Council of Humanities, Sciences, and Technologies, Mexico City, Mexico.
| | - Patricia Webb
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Margaret Neville
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Elizabeth R. Woodruff
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Benjamin G. Bitler
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
15
|
Liu W, Ma Y, He Y, Liu Y, Guo Z, He J, Han X, Hu Y, Li M, Jiang R, Wang S. Discovery of Novel p53-MDM2 Inhibitor (RG7388)-Conjugated Platinum IV Complexes as Potent Antitumor Agents. J Med Chem 2024; 67:9645-9661. [PMID: 38776419 DOI: 10.1021/acs.jmedchem.4c00784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
While a number of p53-MDM2 inhibitors have progressed into clinical trials for the treatment of cancer, their progression has been hampered by a variety of problems, including acquired drug resistance, dose-dependent toxicity, and limited clinical efficiency. To make more progress, we integrated the advantages of MDM2 inhibitors and platinum drugs to construct novel PtIV-RG7388 (a selective MDM2 inhibitor) complexes. Most complexes, especially 5a and 5b, displayed greatly improved antiproliferative activity against both wild-type and mutated p53 cancer cells. Remarkably, 5a exhibited potent in vivo tumor growth inhibition in the A549 xenograft model (66.5%) without apparent toxicity. It arrested the cell cycle at both the S phase and the G2/M phase and efficiently induced apoptosis via the synergistic effects of RG7388 and cisplatin. Altogether, PtIV-RG7388 complex 5a exhibited excellent in vitro and in vivo antitumor activities, highlighting the therapeutic potential of PtIV-RG7388 complexes as antitumor agents.
Collapse
Affiliation(s)
- Wei Liu
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Yi Ma
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Youyou He
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Yanhong Liu
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Zhongjie Guo
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jin He
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xiaodong Han
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yujiao Hu
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Muqiong Li
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ru Jiang
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Shengzheng Wang
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
16
|
Zamanian MY, Golmohammadi M, Yumashev A, Hjazi A, Toama MA, AbdRabou MA, Gehlot A, Alwaily ER, Shirsalimi N, Yadav PK, Moriasi G. Effects of metformin on cancers in experimental and clinical studies: Focusing on autophagy and AMPK/mTOR signaling pathways. Cell Biochem Funct 2024; 42:e4071. [PMID: 38863255 DOI: 10.1002/cbf.4071] [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: 03/23/2024] [Revised: 05/25/2024] [Accepted: 06/02/2024] [Indexed: 06/13/2024]
Abstract
Metformin (MET) is a preferred drug for the treatment of type 2 diabetes mellitus. Recent studies show that apart from its blood glucose-lowering effects, it also inhibits the development of various tumours, by inducing autophagy. Various studies have confirmed the inhibitory effects of MET on cancer cell lines' propagation, migration, and invasion. The objective of the study was to comprehensively review the potential of MET as an anticancer agent, particularly focusing on its ability to induce autophagy and inhibit the development and progression of various tumors. The study aimed to explore the inhibitory effects of MET on cancer cell proliferation, migration, and invasion, and its impact on key signaling pathways such as adenosine monophosphate-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR), and PI3K. This review noted that MET exerts its anticancer effects by regulating key signalling pathways such as phosphoinositide 3-kinase (PI3K), LC3-I and LC3-II, Beclin-1, p53, and the autophagy-related gene (ATG), inhibiting the mTOR protein, downregulating the expression of p62/SQSTM1, and blockage of the cell cycle at the G0/G1. Moreover, MET can stimulate autophagy through pathways associated with the 5' AMPK, thereby inhibiting he development and progression of various human cancers, including hepatocellular carcinoma, prostate cancer, pancreatic cancer, osteosarcoma, myeloma, and non-small cell lung cancer. In summary, this detailed review provides a framework for further investigations that may appraise the autophagy-induced anticancer potential of MET and its repurposing for cancer treatment.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mariam Alaa Toama
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | | | - Anita Gehlot
- Department of Electronics & Communication Engineering, Uttaranchal Institute of Technology, Uttaranchal University, Dehradun, India
| | - Enas R Alwaily
- Microbiology Research Group, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Niyousha Shirsalimi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Pankaj Kumar Yadav
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, India
| | - Gervason Moriasi
- Department of Medical Biochemistry, School of Medicine, College of Health Sciences, Mount Kenya University, Thika, Kenya
| |
Collapse
|
17
|
Ranjbar-Niavol F, Rezaei N, Zhao Y, Mirzaei H, Hassan M, Vosough M. P53/NANOG balance; the leading switch between poorly to well differentiated status in liver cancer cells. Front Oncol 2024; 14:1377761. [PMID: 38846985 PMCID: PMC11153735 DOI: 10.3389/fonc.2024.1377761] [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: 01/28/2024] [Accepted: 04/22/2024] [Indexed: 06/09/2024] Open
Abstract
Enforcing a well-differentiated state on cells requires tumor suppressor p53 activation as a key player in apoptosis induction and well differentiation. In addition, recent investigations showed a significant correlation between poorly differentiated status and higher expression of NANOG. Inducing the expression of NANOG and decreasing p53 level switch the status of liver cancer cells from well differentiated to poorly status. In this review, we highlighted p53 and NANOG cross-talk in hepatocellular carcinoma (HCC) which is regulated through mitophagy and makes it a novel molecular target to attenuate cancerous phenotype in the management of this tumor.
Collapse
Affiliation(s)
- Fazeleh Ranjbar-Niavol
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Niloufar Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Ying Zhao
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute and Karolinska University Hospital-Huddinge, Huddinge, Sweden
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute and Karolinska University Hospital-Huddinge, Huddinge, Sweden
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute and Karolinska University Hospital-Huddinge, Huddinge, Sweden
| |
Collapse
|
18
|
Wu P, Wang X, Yin M, Zhu W, Chen Z, Zhang Y, Jiang Z, Shi L, Zhu Q. ULK1 Mediated Autophagy-Promoting Effects of Rutin-Loaded Chitosan Nanoparticles Contribute to the Activation of NF-κB Signaling Besides Inhibiting EMT in Hep3B Hepatoma Cells. Int J Nanomedicine 2024; 19:4465-4493. [PMID: 38779103 PMCID: PMC11110815 DOI: 10.2147/ijn.s443117] [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] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Background Liver cancer remains to be one of the leading causes of cancer worldwide. The treatment options face several challenges and nanomaterials have proven to improve the bioavailability of several drug candidates and their applications in nanomedicine. Specifically, chitosan nanoparticles (CNPs) are extremely biodegradable, pose enhanced biocompatibility and are considered safe for use in medicine. Methods CNPs were synthesized by ionic gelation, loaded with rutin (rCNPs) and characterized by ultraviolet-visible spectroscopy (UV-Vis), Fourier-transform infrared spectroscopy (FTIR), dynamic light scattering (DLS) and transmission electron microscopy (TEM). The rCNPs were tested for their cytotoxic effects on human hepatoma Hep3B cells, and experiments were conducted to determine the mechanism of such effects. Further, the biocompatibility of the rCNPs was tested on L929 fibroblasts, and their hemocompatibility was determined. Results Initially, UV-vis and FTIR analyses indicated the possible loading of rutin on rCNPs. Further, the rutin load was quantitatively measured using Ultra-Performance Liquid Chromatography (UPLC) and the concentration was 88 µg/mL for 0.22 micron filtered rCNPs. The drug loading capacity (LC%) of the rCNPs was observed to be 13.29 ± 0.68%, and encapsulation efficiency (EE%) was 19.55 ± 1.01%. The drug release was pH-responsive as 88.58% of the drug was released after 24 hrs at the lysosomal pH 5.5, whereas 91.44% of the drug was released at physiological pH 7.4 after 102 hrs. The cytotoxic effects were prominent in 0.22 micron filtered samples of 5 mg/mL rutin precursor. The particle size for the rCNPs at this concentration was 144.1 nm and the polydispersity index (PDI) was 0.244, which is deemed to be ideal for tumor targeting. A zeta potential (ζ-potential) value of 16.4 mV indicated rCNPs with good stability. The IC50 value for the cytotoxic effects of rCNPs on human hepatoma Hep3B cells was 9.7 ± 0.19 μg/mL of rutin load. In addition, the increased production of reactive oxygen species (ROS) and changes in mitochondrial membrane potential (MMP) were observed. Gene expression studies indicated that the mechanism for cytotoxic effects of rCNPs on Hep3B cells was due to the activation of Unc-51-like autophagy-activating kinase (ULK1) mediated autophagy and nuclear factor kappa B (NF-κB) signaling besides inhibiting the epithelial-mesenchymal Transition (EMT). In addition, the rCNPs were less toxic on NCTC clone 929 (L929) fibroblasts in comparison to the Hep3B cells and possessed excellent hemocompatibility (less than 2% of hemolysis). Conclusion The synthesized rCNPs were pH-responsive and possessed the physicochemical properties suitable for tumor targeting. The particles were effectively cytotoxic on Hep3B cells in comparison to normal cells and possessed excellent hemocompatibility. The very low hemolytic profile of rCNPs indicates that the drug could be administered intravenously for cancer therapy.
Collapse
Affiliation(s)
- Peng Wu
- Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Xiaoyong Wang
- The People’s Hospital of Rugao, Nantong, People’s Republic of China
| | - Min Yin
- Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Wenjie Zhu
- Kangda College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Zheng Chen
- Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Yang Zhang
- Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Ziyu Jiang
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, People’s Republic of China
| | - Longqing Shi
- Department of Hepatobiliary and Pancreatic Surgery, Third Affiliated Hospital of Soochow University, Jiangsu, People’s Republic of China
| | - Qiang Zhu
- Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
19
|
Muttiah B, Ng SL, Lokanathan Y, Ng MH, Law JX. Extracellular Vesicles in Breast Cancer: From Intercellular Communication to Therapeutic Opportunities. Pharmaceutics 2024; 16:654. [PMID: 38794316 PMCID: PMC11125876 DOI: 10.3390/pharmaceutics16050654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Breast cancer, a multifaceted and heterogeneous disease, poses significant challenges in terms of understanding its intricate resistance mechanisms and devising effective therapeutic strategies. This review provides a comprehensive overview of the intricate landscape of extracellular vesicles (EVs) in the context of breast cancer, highlighting their diverse subtypes, biogenesis, and roles in intercellular communication within the tumour microenvironment (TME). The discussion spans various aspects, from EVs and stromal cells in breast cancer to their influence on angiogenesis, immune response, and chemoresistance. The impact of EV production in different culture systems, including two dimensional (2D), three dimensional (3D), and organoid models, is explored. Furthermore, this review delves into the therapeutic potential of EVs in breast cancer, presenting emerging strategies such as engineered EVs for gene delivery, nanoplatforms for targeted chemotherapy, and disrupting tumour derived EVs as a treatment approach. Understanding these complex interactions of EV within the breast cancer milieu is crucial for identifying resistance mechanisms and developing new therapeutic targets.
Collapse
Affiliation(s)
- Barathan Muttiah
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Sook Luan Ng
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| |
Collapse
|
20
|
Chauhan S, Jaiswal S, Jakhmola V, Singh B, Bhattacharya S, Garg M, Sengupta S. Potential role of p53 deregulation in modulating immune responses in human malignancies: A paradigm to develop immunotherapy. Cancer Lett 2024; 588:216766. [PMID: 38408603 PMCID: PMC7615729 DOI: 10.1016/j.canlet.2024.216766] [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: 01/19/2024] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
The crucial role played by the oncogenic expression of TP53, stemming from mutation or amyloid formation, in various human malignancies has been extensively studied over the past two decades. Interestingly, the potential role of TP53 as a crucial player in modulating immune responses has provided new insight into the field of cancer biology. The loss of p53's transcriptional functions and/or the acquisition of tumorigenic properties can efficiently modulate the recruitment and functions of myeloid and lymphoid cells, ultimately leading to the evasion of immune responses in human tumors. Consequently, the oncogenic nature of the tumor suppressor p53 can dynamically alter the function of immune cells, providing support for tumor progression and metastasis. This review comprehensively explores the dual role of p53 as both the guardian of the genome and an oncogenic driver, especially in the context of regulation of autophagy, apoptosis, the tumor microenvironment, immune cells, innate immunity, and adaptive immune responses. Additionally, the focus of this review centers on how p53 status in the immune response can be harnessed for the development of tailored therapeutic strategies and their potential application in immunotherapy against human malignancies.
Collapse
Affiliation(s)
- Shivi Chauhan
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India
| | - Shivani Jaiswal
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India
| | - Vibhuti Jakhmola
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India
| | - Bhavana Singh
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India
| | - Sujata Bhattacharya
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India.
| | - Shinjinee Sengupta
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noda, 201313, India.
| |
Collapse
|
21
|
Ortega MA, Fraile-Martinez O, de Leon-Oliva D, Boaru DL, Lopez-Gonzalez L, García-Montero C, Alvarez-Mon MA, Guijarro LG, Torres-Carranza D, Saez MA, Diaz-Pedrero R, Albillos A, Alvarez-Mon M. Autophagy in Its (Proper) Context: Molecular Basis, Biological Relevance, Pharmacological Modulation, and Lifestyle Medicine. Int J Biol Sci 2024; 20:2532-2554. [PMID: 38725847 PMCID: PMC11077378 DOI: 10.7150/ijbs.95122] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/04/2024] [Indexed: 05/12/2024] Open
Abstract
Autophagy plays a critical role in maintaining cellular homeostasis and responding to various stress conditions by the degradation of intracellular components. In this narrative review, we provide a comprehensive overview of autophagy's cellular and molecular basis, biological significance, pharmacological modulation, and its relevance in lifestyle medicine. We delve into the intricate molecular mechanisms that govern autophagy, including macroautophagy, microautophagy and chaperone-mediated autophagy. Moreover, we highlight the biological significance of autophagy in aging, immunity, metabolism, apoptosis, tissue differentiation and systemic diseases, such as neurodegenerative or cardiovascular diseases and cancer. We also discuss the latest advancements in pharmacological modulation of autophagy and their potential implications in clinical settings. Finally, we explore the intimate connection between lifestyle factors and autophagy, emphasizing how nutrition, exercise, sleep patterns and environmental factors can significantly impact the autophagic process. The integration of lifestyle medicine into autophagy research opens new avenues for promoting health and longevity through personalized interventions.
Collapse
Affiliation(s)
- Miguel A Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Diego de Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Miguel Angel Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Luis G Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain
| | - Diego Torres-Carranza
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Miguel A Saez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-UAH Madrid, 28801 Alcala de Henares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Department of General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, 28805 Alcala de Henares, Spain
| | - Agustin Albillos
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), Príncipe de Asturias University Hospital, 28806 Alcala de Henares, Spain
| |
Collapse
|
22
|
Jiang M, Wu W, Xiong Z, Yu X, Ye Z, Wu Z. Targeting autophagy drug discovery: Targets, indications and development trends. Eur J Med Chem 2024; 267:116117. [PMID: 38295689 DOI: 10.1016/j.ejmech.2023.116117] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/30/2023] [Accepted: 12/31/2023] [Indexed: 02/25/2024]
Abstract
Autophagy plays a vital role in sustaining cellular homeostasis and its alterations have been implicated in the etiology of many diseases. Drugs development targeting autophagy began decades ago and hundreds of agents were developed, some of which are licensed for the clinical usage. However, no existing intervention specifically aimed at modulating autophagy is available. The obstacles that prevent drug developments come from the complexity of the actual impact of autophagy regulators in disease scenarios. With the development and application of new technologies, several promising categories of compounds for autophagy-based therapy have emerged in recent years. In this paper, the autophagy-targeted drugs based on their targets at various hierarchical sites of the autophagic signaling network, e.g., the upstream and downstream of the autophagosome and the autophagic components with enzyme activities are reviewed and analyzed respectively, with special attention paid to those at preclinical or clinical trials. The drugs tailored to specific autophagy alone and combination with drugs/adjuvant therapies widely used in clinical for various diseases treatments are also emphasized. The emerging drug design and development targeting selective autophagy receptors (SARs) and their related proteins, which would be expected to arrest or reverse the progression of disease in various cancers, inflammation, neurodegeneration, and metabolic disorders, are critically reviewed. And the challenges and perspective in clinically developing autophagy-targeted drugs and possible combinations with other medicine are considered in the review.
Collapse
Affiliation(s)
- Mengjia Jiang
- Department of Pharmacology and Pharmacy, China Jiliang University, China
| | - Wayne Wu
- College of Osteopathic Medicine, New York Institute of Technology, USA
| | - Zijie Xiong
- Department of Pharmacology and Pharmacy, China Jiliang University, China
| | - Xiaoping Yu
- Department of Biology, China Jiliang University, China
| | - Zihong Ye
- Department of Biology, China Jiliang University, China
| | - Zhiping Wu
- Department of Pharmacology and Pharmacy, China Jiliang University, China.
| |
Collapse
|
23
|
Jasim SA, Almajidi YQ, Al-Rashidi RR, Hjazi A, Ahmad I, Alawadi AHR, Alwaily ER, Alsaab HO, Haslany A, Hameed M. The interaction between lncRNAs and transcription factors regulating autophagy in human cancers: A comprehensive and therapeutical survey. Cell Biochem Funct 2024; 42:e3971. [PMID: 38509767 DOI: 10.1002/cbf.3971] [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: 12/31/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/22/2024]
Abstract
Autophagy, as a highly conserved cellular process, participates in cellular homeostasis by degradation and recycling of damaged organelles and proteins. Besides, autophagy has been evidenced to play a dual role through cancer initiation and progression. In the early stage, it may have a tumor-suppressive function through inducing apoptosis and removing damaged cells and organelles. However, late stages promote tumor progression by maintaining stemness features and induction of chemoresistance. Therefore, identifying and targeting molecular mechanisms involved in autophagy is a potential therapeutic strategy for human cancers. Multiple transcription factors (TFs) are involved in the regulation of autophagy by modulating the expression of autophagy-related genes (ATGs). In addition, a wide array of long noncoding RNAs (lncRNAs), a group of regulatory ncRNAs, have been evidenced to regulate the function of these autophagy-related TFs through tumorigenesis. Subsequently, the lncRNAs/TFs/ATGs axis shows great potential as a therapeutic target for human cancers. Therefore, this review aimed to summarize new findings about the role of lncRNAs in regulating autophagy-related TFs with therapeutic perspectives.
Collapse
Affiliation(s)
| | - Yasir Qasim Almajidi
- Department of Pharmacy (Pharmaceutics), Baghdad College of Medical Sciences, Baghdad, Iraq
| | | | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Ahmed Hussien Radie Alawadi
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| | - Enas R Alwaily
- Microbiology Research Group, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Hashem O Alsaab
- Pharmaceutics and Pharmaceutical Technology, Taif University, Taif, Saudi Arabia
| | - Ali Haslany
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Mohamood Hameed
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
24
|
Price K, Yang WH, Cardoso L, Wang CM, Yang RH, Yang WH. Jun Dimerization Protein 2 (JDP2) Increases p53 Transactivation by Decreasing MDM2. Cancers (Basel) 2024; 16:1000. [PMID: 38473360 DOI: 10.3390/cancers16051000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
The AP-1 protein complex primarily consists of several proteins from the c-Fos, c-Jun, activating transcription factor (ATF), and Jun dimerization protein (JDP) families. JDP2 has been shown to interact with the cAMP response element (CRE) site present in many cis-elements of downstream target genes. JDP2 has also demonstrates important roles in cell-cycle regulation, cancer development and progression, inhibition of adipocyte differentiation, and the regulation of antibacterial immunity and bone homeostasis. JDP2 and ATF3 exhibit significant similarity in their C-terminal domains, sharing 60-65% identities. Previous studies have demonstrated that ATF3 is able to influence both the transcriptional activity and p53 stability via a p53-ATF3 interaction. While some studies have shown that JDP2 suppresses p53 transcriptional activity and in turn, p53 represses JDP2 promoter activity, the direct interaction between JDP2 and p53 and the regulatory role of JDP2 in p53 transactivation have not been explored. In the current study, we provide evidence, for the first time, that JDP2 interacts with p53 and regulates p53 transactivation. First, we demonstrated that JDP2 binds to p53 and the C-terminal domain of JDP2 is crucial for the interaction. Second, in p53-null H1299 cells, JDP2 shows a robust increase of p53 transactivation in the presence of p53 using p53 (14X)RE-Luc. Furthermore, JDP2 and ATF3 together additively enhance p53 transactivation in the presence of p53. While JDP2 can increase p53 transactivation in the presence of WT p53, JDP2 fails to enhance transactivation of hotspot mutant p53. Moreover, in CHX chase experiments, we showed that JDP2 slightly enhances p53 stability. Finally, our findings indicate that JDP2 has the ability to reverse MDM2-induced p53 repression, likely due to decreased levels of MDM2 by JDP2. In summary, our results provide evidence that JDP2 directly interacts with p53 and decreases MDM2 levels to enhance p53 transactivation, suggesting that JDP2 is a novel regulator of p53 and MDM2.
Collapse
Affiliation(s)
- Kasey Price
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA 31404, USA
| | - William H Yang
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA 31404, USA
| | - Leticia Cardoso
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA 31404, USA
| | - Chiung-Min Wang
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA 31404, USA
| | - Richard H Yang
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA 31404, USA
| | - Wei-Hsiung Yang
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA 31404, USA
| |
Collapse
|
25
|
Jianpraphat N, Supsavhad W, Ngernmeesri P, Siripattarapravat K, Soontararak S, Akrimajirachoote N, Phaochoosak N, Jermnak U. A New Benzo[6,7]oxepino[3,2-b] Pyridine Derivative Induces Apoptosis in Canine Mammary Cancer Cell Lines. Animals (Basel) 2024; 14:386. [PMID: 38338029 PMCID: PMC10854894 DOI: 10.3390/ani14030386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
CMC is the most frequently diagnosed cancer and one of the leading causes of death in non-spayed female dogs. Exploring novel therapeutic agents is necessary to increase the survival rate of dogs with CMC. MPOBA is a BZOP derivative that has a significant anticancer effect in a human cell line. The main goal of this study was to investigate the anticancer properties of MPOBA against two CMC cell lines (REM134 and CMGT071020) using a 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay, a wound healing assay, a transwell migration assay, an Annexin V-FITC apoptosis assay with a flow cytometry analysis, a mRNA expression analysis using quantitative real-time PCR (qRT-PCR), and an immunohistochemistry (IHC). According to the accumulated studies, MPOBA caused significant concentration- and time-dependent reductions in cell proliferation and cell migration and induced apoptosis in both CMC cell lines. In gene expression analysis, nine canine genes, including TP53, BCL-2, BAX, epidermal growth factor receptor (EGFR), snail transcription factor (SNAIL), snail-related zinc-finger transcription factor (SLUG), TWIST, E-cadherin, and N-cadherin, were investigated. The mRNA expression results revealed that MPOBA induced upregulation of TP53 and overexpression of the pro-apoptotic gene BAX, together with an inhibition of BCL-2. Moreover, MPOBA also suppressed the mRNA expression levels of SNAIL, EGFR, and N-cadherin and induced upregulation of E-cadherin, crucial genes related to the epithelial-to-mesenchymal transition (EMT). However, there was no significant difference in the IHC results of the expression patterns of vimentin (VT) and cytokeratin (CK) between MPOBA-treated and control CMC cells. In conclusion, the results of the present study suggested that MPOBA exhibited significant anticancer activity by inducing apoptosis in both CMCs via upregulation of TP53 and BAX and downregulation of BCL-2 relative mRNA expression. MPOBA may prove to be a potential candidate drug to be further investigated as a therapeutic agent for CMC.
Collapse
Affiliation(s)
- Natamon Jianpraphat
- Department of Pharmacology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand; (N.J.); (N.P.)
| | - Wachiraphan Supsavhad
- Department of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand; (W.S.); (K.S.)
| | - Paiboon Ngernmeesri
- Department of Chemistry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand;
| | - Kannika Siripattarapravat
- Department of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand; (W.S.); (K.S.)
| | - Sirikul Soontararak
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand;
| | | | - Napasorn Phaochoosak
- Department of Pharmacology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand; (N.J.); (N.P.)
| | - Usuma Jermnak
- Department of Pharmacology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand; (N.J.); (N.P.)
| |
Collapse
|
26
|
Marras E, Balacchi CJ, Orlandi V, Caruso E, Brivio MF, Bolognese F, Mastore M, Malacarne MC, Rossi M, Caruso F, Vivona V, Ferrario N, Gariboldi MB. Ruthenium(II)-Arene Curcuminoid Complexes as Photosensitizer Agents for Antineoplastic and Antimicrobial Photodynamic Therapy: In Vitro and In Vivo Insights. Molecules 2023; 28:7537. [PMID: 38005258 PMCID: PMC10673066 DOI: 10.3390/molecules28227537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Photodynamic therapy (PDT) is an anticancer/antibacterial strategy in which photosensitizers (PSs), light, and molecular oxygen generate reactive oxygen species and induce cell death. PDT presents greater selectivity towards tumor cells than conventional chemotherapy; however, PSs have limitations that have prompted the search for new molecules featuring more favorable chemical-physical characteristics. Curcumin and its derivatives have been used in PDT. However, low water solubility, rapid metabolism, interference with other drugs, and low stability limit curcumin use. Chemical modifications have been proposed to improve curcumin activity, and metal-based PSs, especially ruthenium(II) complexes, have attracted considerable attention. This study aimed to characterize six Ru(II)-arene curcuminoids for anticancer and/or antibacterial PDT. The hydrophilicity, photodegradation rates, and singlet oxygen generation of the compounds were evaluated. The photodynamic effects on human colorectal cancer cell lines were also assessed, along with the ability of the compounds to induce ROS production, apoptotic, necrotic, and/or autophagic cell death. Overall, our encouraging results indicate that the Ru(II)-arene curcuminoid derivatives are worthy of further investigation and could represent an interesting option for cancer PDT. Additionally, the lack of significant in vivo toxicity on the larvae of Galleria mellonella is an important finding. Finally, the photoantimicrobial activity of HCurc I against Gram-positive bacteria is indeed promising.
Collapse
Affiliation(s)
- Emanuela Marras
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy; (E.M.); (C.J.B.); (V.O.); (E.C.); (F.B.); (M.C.M.); (V.V.); (N.F.)
| | - Camilla J. Balacchi
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy; (E.M.); (C.J.B.); (V.O.); (E.C.); (F.B.); (M.C.M.); (V.V.); (N.F.)
| | - Viviana Orlandi
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy; (E.M.); (C.J.B.); (V.O.); (E.C.); (F.B.); (M.C.M.); (V.V.); (N.F.)
| | - Enrico Caruso
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy; (E.M.); (C.J.B.); (V.O.); (E.C.); (F.B.); (M.C.M.); (V.V.); (N.F.)
| | - Maurizio F. Brivio
- Department of Theoretical and Applied Sciences (DiSTA), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy; (M.F.B.); (M.M.)
| | - Fabrizio Bolognese
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy; (E.M.); (C.J.B.); (V.O.); (E.C.); (F.B.); (M.C.M.); (V.V.); (N.F.)
| | - Maristella Mastore
- Department of Theoretical and Applied Sciences (DiSTA), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy; (M.F.B.); (M.M.)
| | - Miryam C. Malacarne
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy; (E.M.); (C.J.B.); (V.O.); (E.C.); (F.B.); (M.C.M.); (V.V.); (N.F.)
| | - Miriam Rossi
- Department of Chemistry, Vassar College, Poughkeepsie, NY 12604, USA; (M.R.)
| | - Francesco Caruso
- Department of Chemistry, Vassar College, Poughkeepsie, NY 12604, USA; (M.R.)
| | - Veronica Vivona
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy; (E.M.); (C.J.B.); (V.O.); (E.C.); (F.B.); (M.C.M.); (V.V.); (N.F.)
| | - Nicole Ferrario
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy; (E.M.); (C.J.B.); (V.O.); (E.C.); (F.B.); (M.C.M.); (V.V.); (N.F.)
| | - Marzia B. Gariboldi
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy; (E.M.); (C.J.B.); (V.O.); (E.C.); (F.B.); (M.C.M.); (V.V.); (N.F.)
| |
Collapse
|
27
|
Khilwani R, Singh S. Systems Biology and Cytokines Potential Role in Lung Cancer Immunotherapy Targeting Autophagic Axis. Biomedicines 2023; 11:2706. [PMID: 37893079 PMCID: PMC10604646 DOI: 10.3390/biomedicines11102706] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 10/29/2023] Open
Abstract
Lung cancer accounts for the highest number of deaths among men and women worldwide. Although extensive therapies, either alone or in conjunction with some specific drugs, continue to be the principal regimen for evolving lung cancer, significant improvements are still needed to understand the inherent biology behind progressive inflammation and its detection. Unfortunately, despite every advancement in its treatment, lung cancer patients display different growth mechanisms and continue to die at significant rates. Autophagy, which is a physiological defense mechanism, serves to meet the energy demands of nutrient-deprived cancer cells and sustain the tumor cells under stressed conditions. In contrast, autophagy is believed to play a dual role during different stages of tumorigenesis. During early stages, it acts as a tumor suppressor, degrading oncogenic proteins; however, during later stages, autophagy supports tumor cell survival by minimizing stress in the tumor microenvironment. The pivotal role of the IL6-IL17-IL23 signaling axis has been observed to trigger autophagic events in lung cancer patients. Since the obvious roles of autophagy are a result of different immune signaling cascades, systems biology can be an effective tool to understand these interconnections and enhance cancer treatment and immunotherapy. In this review, we focus on how systems biology can be exploited to target autophagic processes that resolve inflammatory responses and contribute to better treatment in carcinogenesis.
Collapse
Affiliation(s)
| | - Shailza Singh
- Systems Medicine Laboratory, National Centre for Cell Science, SPPU Campus, Ganeshkhind Road, Pune 411007, India;
| |
Collapse
|
28
|
Elshazly AM, Gewirtz DA. The Cytoprotective Role of Autophagy in Response to BRAF-Targeted Therapies. Int J Mol Sci 2023; 24:14774. [PMID: 37834222 PMCID: PMC10572960 DOI: 10.3390/ijms241914774] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
BRAF-targeted therapies are widely used for the treatment of melanoma patients with BRAF V600 mutations. Vemurafenib, dabrafenib as well as encorafenib have demonstrated substantial therapeutic activity; however, as is the case with other chemotherapeutic agents, the frequent development of resistance limits their efficacy. Autophagy is one tumor survival mechanism that could contribute to BRAF inhibitor resistance, and multiple studies support an association between vemurafenib-induced and dabrafenib-induced autophagy and tumor cell survival. Clinical trials have also demonstrated a potential benefit from the inclusion of autophagy inhibition as an adjuvant therapy. This review of the scientific literature relating to the role of autophagy that is induced in response to BRAF-inhibitors supports the premise that autophagy targeting or modulation could be an effective adjuvant therapy.
Collapse
Affiliation(s)
- Ahmed M. Elshazly
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
| |
Collapse
|
29
|
Elshazly AM, Gewirtz DA. Cytoprotective, Cytotoxic and Cytostatic Roles of Autophagy in Response to BET Inhibitors. Int J Mol Sci 2023; 24:12669. [PMID: 37628849 PMCID: PMC10454099 DOI: 10.3390/ijms241612669] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/28/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The bromodomain and extra-terminal domain (BET) family inhibitors are small molecules that target the dysregulated epigenetic readers, BRD2, BRD3, BRD4 and BRDT, at various transcription-related sites, including super-enhancers. BET inhibitors are currently under investigation both in pre-clinical cell culture and tumor-bearing animal models, as well as in clinical trials. However, as is the case with other chemotherapeutic modalities, the development of resistance is likely to constrain the therapeutic benefits of this strategy. One tumor cell survival mechanism that has been studied for decades is autophagy. Although four different functions of autophagy have been identified in the literature (cytoprotective, cytotoxic, cytostatic and non-protective), primarily the cytoprotective and cytotoxic forms appear to function in different experimental models exposed to BET inhibitors (with some evidence for the cytostatic form). This review provides an overview of the cytoprotective, cytotoxic and cytostatic functions of autophagy in response to BET inhibitors in various tumor models. Our aim is to determine whether autophagy targeting or modulation could represent an effective therapeutic strategy to enhance the response to these modalities and also potentially overcome resistance to BET inhibition.
Collapse
Affiliation(s)
- Ahmed M. Elshazly
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
| |
Collapse
|
30
|
Lee S, Hwang N, Seok BG, Lee S, Lee SJ, Chung SW. Autophagy mediates an amplification loop during ferroptosis. Cell Death Dis 2023; 14:464. [PMID: 37491375 PMCID: PMC10368698 DOI: 10.1038/s41419-023-05978-8] [Citation(s) in RCA: 106] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/09/2023] [Accepted: 07/11/2023] [Indexed: 07/27/2023]
Abstract
Ferroptosis, a programmed cell death, has been identified and associated with cancer and various other diseases. Ferroptosis is defined as a reactive oxygen species (ROS)-dependent cell death related to iron accumulation and lipid peroxidation, which is different from apoptosis, necrosis, autophagy, and other forms of cell death. However, accumulating evidence has revealed a link between autophagy and ferroptosis at the molecular level and has suggested that autophagy is involved in regulating the accumulation of iron-dependent lipid peroxidation and ROS during ferroptosis. Understanding the roles and pathophysiological processes of autophagy during ferroptosis may provide effective strategies for the treatment of ferroptosis-related diseases. In this review, we summarize the current knowledge regarding the regulatory mechanisms underlying ferroptosis, including iron and lipid metabolism, and its association with the autophagy pathway. In addition, we discuss the contribution of autophagy to ferroptosis and elucidate the role of autophagy as a ferroptosis enhancer during ROS-dependent ferroptosis.
Collapse
Affiliation(s)
- Seunghee Lee
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, 93 Daehak-ro, Nam-gu, Ulsan, 44610, South Korea
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, VA Palo Alto Health Care System and Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| | - Narae Hwang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Byeong Geun Seok
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, 93 Daehak-ro, Nam-gu, Ulsan, 44610, South Korea
| | - Sangguk Lee
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, 93 Daehak-ro, Nam-gu, Ulsan, 44610, South Korea
| | - Seon-Jin Lee
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, 34141, South Korea
| | - Su Wol Chung
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, 93 Daehak-ro, Nam-gu, Ulsan, 44610, South Korea.
- Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan, 44610, South Korea.
| |
Collapse
|
31
|
Lei Y, Klionsky DJ. Transcriptional regulation of autophagy and its implications in human disease. Cell Death Differ 2023; 30:1416-1429. [PMID: 37045910 PMCID: PMC10244319 DOI: 10.1038/s41418-023-01162-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
Macroautophagy/autophagy is a conserved catabolic pathway that is vital for maintaining cell homeostasis and promoting cell survival under stressful conditions. Dysregulation of autophagy is associated with a variety of human diseases, such as cancer, neurodegenerative diseases, and metabolic disorders. Therefore, this pathway must be precisely regulated at multiple levels, involving epigenetic, transcriptional, post-transcriptional, translational, and post-translational mechanisms, to prevent inappropriate autophagy activity. In this review, we focus on autophagy regulation at the transcriptional level, summarizing the transcription factors that control autophagy gene expression in both yeast and mammalian cells. Because the expression and/or subcellular localization of some autophagy transcription factors are altered in certain diseases, we also discuss how changes in transcriptional regulation of autophagy are associated with human pathophysiologies.
Collapse
Affiliation(s)
- Yuchen Lei
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
32
|
Belyaeva E, Loginova N, Schroeder BA, Goldlust IS, Acharya A, Kumar S, Timashev P, Ulasov I. The spectrum of cell death in sarcoma. Biomed Pharmacother 2023; 162:114683. [PMID: 37031493 DOI: 10.1016/j.biopha.2023.114683] [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: 03/03/2023] [Revised: 04/01/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023] Open
Abstract
The balance between cell death and cell survival is a highly coordinated process by which cells break down and remove unnecessary or harmful materials in a controlled, highly regulated, and compartmentalized manner. Cell exposure to various stresses, such as oxygen starvation, a lack of nutrients, or exposure to radiation, can initiate autophagy. Autophagy is a carefully orchestrated process with multiple steps, each regulated by specific genes and proteins. Autophagy proteins impact cellular maintenance and cell fate in response to stress, and targeting this process is one of the most promising methods of anti-tumor therapy. It is currently not fully understood how autophagy affects different types of tumor cells, which makes it challenging to predict outcomes when this process is manipulated. In this review, we will explore the mechanisms of autophagy and investigate it as a potential and promising therapeutic target for aggressive sarcomas.
Collapse
Affiliation(s)
- Elizaveta Belyaeva
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Nina Loginova
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Brett A Schroeder
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Ian S Goldlust
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Arbind Acharya
- Laboratory of Cancer Immunology, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Sandeep Kumar
- Laboratory of Cancer Immunology, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Peter Timashev
- World-Class Research Centre "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia.
| |
Collapse
|
33
|
Elshazly AM, Gewirtz DA. Is Autophagy Inhibition in Combination with Temozolomide a Therapeutically Viable Strategy? Cells 2023; 12:535. [PMID: 36831202 PMCID: PMC9954434 DOI: 10.3390/cells12040535] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Temozolomide is an oral alkylating agent that is used as the first line treatment for glioblastoma multiform, and in recurrent anaplastic astrocytoma, as well as having demonstrable activity in patients with metastatic melanoma. However, as the case with other chemotherapeutic agents, the development of resistance often limits the therapeutic benefit of temozolomide, particularly in the case of glioblastoma. A number of resistance mechanisms have been proposed including the development of cytoprotective autophagy. Cytoprotective autophagy is a survival mechanism that confers upon tumor cells the ability to survive in a nutrient deficient environment as well as under external stresses, such as cancer chemotherapeutic drugs and radiation, in part through the suppression of apoptotic cell death. In this review/commentary, we explore the available literature and provide an overview of the evidence for the promotion of protective autophagy in response to temozolomide, highlighting the possibility of targeting autophagy as an adjuvant therapy to potentially increase the effectiveness of temozolomide and to overcome the development of resistance.
Collapse
Affiliation(s)
- Ahmed M. Elshazly
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA
| |
Collapse
|
34
|
Yang J, Xu Y, Fu Z, Chen J, Fan W, Wu X. Progress in research and development of temozolomide brain-targeted preparations: a review. J Drug Target 2023; 31:119-133. [PMID: 36039767 DOI: 10.1080/1061186x.2022.2119243] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gliomas are a heterogeneous group of brain tumours with high malignancy, for which surgical resection remains the mainstay of treatment at present. However, the overall prognosis of gliomas remains poor because of their aggressiveness and high recurrence. Temozolomide (TMZ) has anti-proliferative and cytotoxic effects and is indicated for glioblastoma multiforme and recurrent mesenchymal astrocytoma. However, TMZ is disadvantaged by low efficacy and drug resistance, and therefore it is necessary to enhance the brain drug concentration of TMZ to improve its effectiveness and reduce the toxic and adverse effects from systemic administration. There have been many nano-formulations developed for the delivery of TMZ to gliomas that overcome the limitations of TMZ penetration to tumours and increase brain targeting. In this paper, we review the research progress of TMZ nano-formulations, and also discuss challenges and opportunities in the research and development of drug delivery systems, hoping that the data and information summarised herein could provide assistance for the clinical treatment of gliomas.
Collapse
Affiliation(s)
- Jiefen Yang
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Youfa Xu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Department of Pharmacy, Shanghai Wei Er Biopharmaceutical Technology Co., Ltd, Shanghai, China
| | - Zhiqin Fu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Department of Pharmacy, Shanghai Wei Er Biopharmaceutical Technology Co., Ltd, Shanghai, China
| | - Jianming Chen
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Wei Fan
- Department of Pharmacy, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Wu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Department of Pharmacy, Shanghai Wei Er Biopharmaceutical Technology Co., Ltd, Shanghai, China
| |
Collapse
|
35
|
Yang WH, George AP, Wang CM, Yang RH, Duncan AM, Patel D, Neil ZD, Yang WH. Tumor Suppressor p53 Down-Regulates Programmed Cell Death Protein 4 (PDCD4) Expression. Curr Oncol 2023; 30:1614-1625. [PMID: 36826085 PMCID: PMC9955764 DOI: 10.3390/curroncol30020124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/19/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
The programmed cell death protein 4 (PDCD4), a well-known tumor suppressor, inhibits translation initiation and cap-dependent translation by inhibiting the helicase activity of EIF4A. The EIF4A tends to target mRNAs with a structured 5'-UTR. In addition, PDCD4 can also prevent tumorigenesis by inhibiting tumor promoter-induced neoplastic transformation, and studies indicate that PDCD4 binding to certain mRNAs inhibits those mRNAs' translation. A previous study demonstrated that PDCD4 inhibits the translation of p53 mRNA and that treatment with DNA-damaging agents down-regulates PDCD4 expression but activates p53 expression. The study further demonstrated that treatment with DNA-damaging agents resulted in the downregulation of PDCD4 expression and an increase in p53 expression, suggesting a potential mechanism by which p53 regulates the expression of PDCD4. However, whether p53 directly regulates PDCD4 remains unknown. Herein, we demonstrate for the first time that p53 regulates PDCD4 expression. Firstly, we found that overexpression of p53 in p53-null cells (H1299 and Saos2 cells) decreased the PDCD4 protein level. Secondly, p53 decreased PDCD4 promoter activity in gene reporter assays. Moreover, we demonstrated that mutations in p53 (R273H: contact hotspot mutation, and R175H: conformational hotspot mutation) abolished p53-mediated PDCD4 repression. Furthermore, mutations in the DNA-binding domain, but not in the C-terminal regulatory domain, of p53 disrupted p53-mediated PDCD4 repression. Finally, the C-terminal regulatory domain truncation study showed that the region between aa374 and aa370 is critical for p53-mediated PDCD4 repression. Taken together, our results suggest that p53 functions as a novel regulator of PDCD4, and the relationship between p53 and PDCD4 may be involved in tumor development and progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wei-Hsiung Yang
- Correspondence: ; Tel.: +1-912-721-8203; Fax: +1-912-721-8268
| |
Collapse
|
36
|
Saberi F, Dehghan Z, Noori E, Taheri Z, Sameni M, Zali H. Identification of Critical Molecular Factors and Side Effects Underlying the Response to Thalicthuberine in Prostate Cancer: A Systems Biology Approach. Avicenna J Med Biotechnol 2023; 15:53-64. [PMID: 36789117 PMCID: PMC9895985 DOI: 10.18502/ajmb.v15i1.11425] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/05/2022] [Indexed: 12/27/2022] Open
Abstract
Background Uncontrolled mitosis of cancer cells and resistance cells to chemotherapy drugs are the challenges of prostate cancer. Thalicthuberine causes a mitotic arrest and a reduction of the effects of drug resistance, resulting in cell death. In this study, we applied bioinformatics and computational biology methods to identify functional pathways and side effects in response to Thalicthuberine in prostate cancer patients. Methods Microarray data were retrieved from Gene Expression Omnibus (GEO), and protein-protein interactions and gene regulatory networks were constructed, using the Cytoscape software. The critical genes and molecular mechanisms in response to Thalicthuberine and its side effects were identified, using the Cytoscape software and WebGestalt server, respectively. Finally, GEPIA2 was used to predict the relationship between critical genes and prostate cancer. Results The POLQ, EGR1, CDKN1A, FOS, MDM2, CDC20, CCNB1, and CCNB2 were identified as critical genes in response to this drug. The functional mechanisms of Thalicthuberine include a response to oxygen levels, toxic substances and immobilization stress, cell cycle regulation, regeneration, the p53 signaling pathway, the action of the parathyroid hormone, and the FoxO signaling pathway. Besides, the drug has side effects including muscle cramping, abdominal pains, paresthesia, and metabolic diseases. Conclusion Our model suggested newly predicted crucial genes, molecular mechanisms, and possible side effects of this drug. However, further studies are required.
Collapse
Affiliation(s)
- Fatemeh Saberi
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Dehghan
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Effat Noori
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Taheri
- Department of Biology and Biotechnology, Pavia University, Pavia, Italy
| | - Marzieh Sameni
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hakimeh Zali
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Chang HH, Lin YH, Chen TM, Tsai YL, Lai CR, Tsai WC, Cheng YC, Chen Y. ONX-0914 Induces Apoptosis and Autophagy with p53 Regulation in Human Glioblastoma Cells. Cancers (Basel) 2022; 14:cancers14225712. [PMID: 36428804 PMCID: PMC9688407 DOI: 10.3390/cancers14225712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Glioblastoma is believed to be one of the most aggressive brain tumors in the world. ONX-0914 (PR957) is a selective inhibitor of proteasome subunit beta type-8 (PSMB8). Previous studies have shown that inhibiting PSMB8 expression in glioblastoma reduces tumor progression. Therefore, this study aimed to determine whether ONX-0914 has antitumor effects on human glioblastoma. The results indicated that ONX-0914 treatment inhibited survival in LN229, GBM8401, and U87MG glioblastoma cells. Cell cycle analysis showed that ONX-0914 treatment caused cell cycle arrest at the G1 phase and apoptosis in glioblastoma cells. The protein expression of BCL-2 was reduced and PARP was cleaved after ONX-0914 treatment. Furthermore, the levels of p53 and phosphorylated p53 were increased by ONX-0914 treatment in glioblastoma cells. ONX-0914 also induced autophagy in glioblastoma cells. Furthermore, the p53 inhibitor pifithrin attenuated apoptosis but enhanced autophagy caused by ONX-0914. In an orthotopic mouse model, TMZ plus ONX-0914 reduced tumor progression better than the control or TMZ alone. These data suggest that ONX-0914 is a novel therapeutic drug for glioblastoma.
Collapse
Affiliation(s)
- Hsin-Han Chang
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114201, Taiwan
- Department of Nursing, Ching Kuo Institute of Management and Health, Keelung 203301, Taiwan
| | - Yi-Hsuan Lin
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114201, Taiwan
| | - Tzu-Min Chen
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114201, Taiwan
| | - Yu-Ling Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Chien-Rui Lai
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114201, Taiwan
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Yu-Chen Cheng
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114201, Taiwan
- Correspondence: (Y.-C.C.); (Y.C.); Tel.: +886-2-8792-3100 (ext. 18739) (Y.C.)
| | - Ying Chen
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114201, Taiwan
- Correspondence: (Y.-C.C.); (Y.C.); Tel.: +886-2-8792-3100 (ext. 18739) (Y.C.)
| |
Collapse
|
38
|
Wang Y, Goh KY, Chen Z, Lee WX, Choy SM, Fong JX, Wong YK, Li D, Hu F, Tang HW. A Novel TP53 Gene Mutation Sustains Non-Small Cell Lung Cancer through Mitophagy. Cells 2022; 11:3587. [PMID: 36429016 PMCID: PMC9688643 DOI: 10.3390/cells11223587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Lung cancer is the leading cause of cancer death in the world. In particular, non-small-cell lung cancer (NSCLC) represents the majority of the lung cancer population. Advances in DNA sequencing technologies have significantly contributed to revealing the roles, functions and mechanisms of gene mutations. However, the driver mutations that cause cancers and their pathologies remain to be explored. Here, we performed next-generation sequencing (NGS) on tumor tissues isolated from 314 Chinese NSCLC patients and established the mutational landscape in NSCLC. Among 656 mutations, we identified TP53-p.Glu358Val as a driver mutation in lung cancer and found that it activates mitophagy to sustain cancer cell growth. In support of this finding, mice subcutaneously implanted with NSCLC cells expressing TP53-p.Glu358Val developed larger tumors compared to wild-type cells. The pharmaceutical inhibition of autophagy/mitophagy selectively suppresses the cell proliferation of TP53-null or TP53-p.Glu358Val-expressing lung cancer cells. Together, our study characterizes a new TP53 mutation identified from Chinese lung cancer patients and uncovers its roles in regulating mitophagy, providing a new insight into NSCLC treatment.
Collapse
Affiliation(s)
- Yuanli Wang
- School of Life and Environmental Sciences, Guilin University of Electronic Technology, Guilin 541014, China
- Precision Medicine Laboratory, The First People’s Hospital of Qinzhou, Qinzhou 535000, China
| | - Kah Yong Goh
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Zhencheng Chen
- School of Life and Environmental Sciences, Guilin University of Electronic Technology, Guilin 541014, China
| | - Wen Xing Lee
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Sze Mun Choy
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Jia Xin Fong
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Yun Ka Wong
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Dongxia Li
- School of Electronic Engineering and Automation, Guilin University of Electronic Technology, Guilin 541004, China
| | - Fangrong Hu
- School of Electronic Engineering and Automation, Guilin University of Electronic Technology, Guilin 541004, China
| | - Hong-Wen Tang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Division of Cellular & Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore
| |
Collapse
|
39
|
Yao J, Ma C, Feng K, Tan G, Wen Q. Focusing on the Role of Natural Products in Overcoming Cancer Drug Resistance: An Autophagy-Based Perspective. Biomolecules 2022; 12:1565. [PMID: 36358919 PMCID: PMC9687214 DOI: 10.3390/biom12111565] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 10/15/2023] Open
Abstract
Autophagy is a critical cellular adaptive response in tumor formation. Nutritional deficiency and hypoxia exacerbate autophagic flux in established malignancies, promoting tumor cell proliferation, migration, metastasis, and resistance to therapeutic interventions. Pro-survival autophagy inhibition may be a promising treatment option for advanced cancer. Furthermore, excessive or persistent autophagy is cytotoxic, resulting in tumor cell death. Targeted autophagy activation has also shown significant promise in the fight against tumor drug resistance. Several research groups have examined the ability of natural products (NPs) such as alkaloids, terpenoids, polyphenols, and anthraquinones to serve as autophagy inhibitors or activators. The data support the capacity of NPs that promote lethal autophagy or inhibit pro-survival autophagy from being employed against tumor drug resistance. This paper discusses the potential applications of NPs that regulate autophagy in the fight against tumor drug resistance, some limitations of the current studies, and future research needs and priorities.
Collapse
Affiliation(s)
- Jiaqi Yao
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Chi Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Kaixuan Feng
- Department of Anesthesiology, The Affiliated Xinhua Hospital of Dalian University, Dalian 116021, China
| | - Guang Tan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Qingping Wen
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
40
|
Rahman MA, Ahmed KR, Rahman MDH, Parvez MAK, Lee IS, Kim B. Therapeutic Aspects and Molecular Targets of Autophagy to Control Pancreatic Cancer Management. Biomedicines 2022; 10:1459. [PMID: 35740481 PMCID: PMC9220066 DOI: 10.3390/biomedicines10061459] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/17/2022] [Accepted: 06/17/2022] [Indexed: 12/29/2022] Open
Abstract
Pancreatic cancer (PC) begins within the organ of the pancreas, which produces digestive enzymes, and is one of the formidable cancers for which appropriate treatment strategies are urgently needed. Autophagy occurs in the many chambers of PC tissue, including cancer cells, cancer-related fibroblasts, and immune cells, and can be fine-tuned by various promotive and suppressive signals. Consequently, the impacts of autophagy on pancreatic carcinogenesis and progression depend greatly on its stage and conditions. Autophagy inhibits the progress of preneoplastic damage during the initial phase. However, autophagy encourages tumor formation during the development phase. Several studies have reported that both a tumor-promoting and a tumor-suppressing function of autophagy in cancer that is likely cell-type dependent. However, autophagy is dispensable for pancreatic ductal adenocarcinoma (PDAC) growth, and clinical trials with autophagy inhibitors, either alone or in combination with other therapies, have had limited success. Autophagy's dual mode of action makes it therapeutically challenging despite autophagy inhibitors providing increased longevity in medical studies, highlighting the need for a more rigorous review of current findings and more precise targeting strategies. Indeed, the role of autophagy in PC is complicated, and numerous factors must be considered when transitioning from bench to bedside. In this review, we summarize the evidence for the tumorigenic and protective role of autophagy in PC tumorigenesis and describe recent advances in the understanding of how autophagy may be regulated and controlled in PDAC.
Collapse
Affiliation(s)
- Md. Ataur Rahman
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 02447, Korea; (K.R.A.); (M.H.R.)
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
- Global Biotechnology & Biomedical Research Network (GBBRN), Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh
| | - Kazi Rejvee Ahmed
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 02447, Korea; (K.R.A.); (M.H.R.)
| | - MD. Hasanur Rahman
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 02447, Korea; (K.R.A.); (M.H.R.)
| | | | - In-Seon Lee
- Acupuncture & Meridian Science Research Center, Kyung Hee University, Seoul 02447, Korea;
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 02447, Korea; (K.R.A.); (M.H.R.)
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
41
|
Li X, Xiong K, Bi D, Zhao C. A Novel CRISPR/Cas9 Screening Potential Index for Prognostic and Immunological Prediction in Low-Grade Glioma. Front Genet 2022; 13:839884. [PMID: 35586564 PMCID: PMC9109250 DOI: 10.3389/fgene.2022.839884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 03/18/2022] [Indexed: 12/05/2022] Open
Abstract
Glioma is a malignancy with the highest mortality in central nervous system disorders. Here, we implemented the computational tools based on CRISPR/Cas9 to predict the clinical outcomes and biological characteristics of low-grade glioma (LGG). The transcriptional expression profiles and clinical phenotypes of LGG patients were retrieved from The Cancer Genome Atlas and Chinese Glioma Genome Atlas. The CERES algorithm was used to screen for LGG-lethal genes. Cox regression and random survival forest were adopted for survival-related gene selection. Nonnegative matrix factorization distinguished patients into different clusters. Single-sample gene set enrichment analysis was employed to create a novel CRISPR/Cas9 screening potential index (CCSPI), and patients were stratified into low- and high-CCSPI groups. Survival analysis, area under the curve values (AUCs), nomogram, and tumor microenvironment exploration were included for the model validation. A total of 20 essential genes in LGG were used to classify patients into two clusters and construct the CCSPI system. High-CCSPI patients were associated with a worse prognosis of both training and validation set (p < 0.0001) and higher immune fractions than low-CCSPI individuals. The CCSPI system had a promising performance with 1-, 3-, and 5-year AUCs of 0.816, 0.779, 0.724, respectively, and the C-index of the nomogram model reached 0.743 (95% CI = 0.725–0.760). Immune-infiltrating cells and immune checkpoints such as PD-1/PD-L1 and POLD3 were positively associated with CCSPI. In conclusion, the CCSPI had prognostic value in LGG, and the model will deepen our cognition of the interaction between the CNS and immune system in different LGG subtypes.
Collapse
Affiliation(s)
- Xiangpan Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Kewei Xiong
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China.,School of Mathematics and Statistics, Central China Normal University, Wuhan, China
| | - Dong Bi
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chen Zhao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
42
|
Lagunas-Martínez A, Madrid-Marina V, Gómez-Cerón C, Deas J, Peralta-Zaragoza O. The Autophagy Process in Cervical Carcinogenesis: Role of Non-Coding-RNAs, Molecular Mechanisms, and Therapeutic Targets. Cells 2022; 11:cells11081323. [PMID: 35456001 PMCID: PMC9028856 DOI: 10.3390/cells11081323] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 02/01/2023] Open
Abstract
Autophagy is a highly conserved multistep lysosomal degradation process in which cellular components are localized to autophagosomes, which subsequently fuse with lysosomes to degrade the sequestered contents. Autophagy serves to maintain cellular homeostasis. There is a close relationship between autophagy and tumor progression, which provides opportunities for the development of anticancer therapeutics that target the autophagy pathway. In this review, we analyze the effects of human papillomavirus (HPV) E5, E6, and E7 oncoproteins on autophagy processes in cervical cancer development. Inhibition of the expression or the activity of E5, E6, and E7 can induce autophagy in cells expressing HPV oncogenes. Thus, E5, E6, and E7 oncoproteins target autophagy during HPV-associated carcinogenesis. Furthermore, noncoding RNA (ncRNA) expression profiling in cervical cancer has allowed the identification of autophagy-related ncRNAs associated with HPV. Autophagy-related genes are essential drivers of autophagy and are regulated by ncRNAs. We review the existing evidence regarding the role of autophagy-related proteins, the function of HPV E5, E6, and E7 oncoproteins, and the effects of noncoding RNA on autophagy regulation in the setting of cervical carcinogenesis. By characterizing the mechanisms behind the dysregulation of these critical factors and their impact on host cell autophagy, we advance understanding of the relationship between autophagy and progression from HPV infection to cervical cancer, and highlight pathways that can be targeted in preventive and therapeutic strategies against cervical cancer.
Collapse
Affiliation(s)
- Alfredo Lagunas-Martínez
- Direction of Chronic Infections and Cancer, Research Center in Infection Diseases, National Institute of Public Health, Av. Universidad No. 655, Cerrada los Pinos y Caminera, Colonia Santa María Ahuacatitlán, Cuernavaca 62100, Morelos, Mexico; (A.L.-M.); (V.M.-M.); (J.D.)
| | - Vicente Madrid-Marina
- Direction of Chronic Infections and Cancer, Research Center in Infection Diseases, National Institute of Public Health, Av. Universidad No. 655, Cerrada los Pinos y Caminera, Colonia Santa María Ahuacatitlán, Cuernavaca 62100, Morelos, Mexico; (A.L.-M.); (V.M.-M.); (J.D.)
| | - Claudia Gómez-Cerón
- Research Center in Population Health, Department of Cancer Epidemiology, National Institute of Public Health, Av. Universidad No. 655, Cerrada los Pinos y Caminera, Colonia Santa María Ahuacatitlán, Cuernavaca 62100, Morelos, Mexico;
| | - Jessica Deas
- Direction of Chronic Infections and Cancer, Research Center in Infection Diseases, National Institute of Public Health, Av. Universidad No. 655, Cerrada los Pinos y Caminera, Colonia Santa María Ahuacatitlán, Cuernavaca 62100, Morelos, Mexico; (A.L.-M.); (V.M.-M.); (J.D.)
| | - Oscar Peralta-Zaragoza
- Direction of Chronic Infections and Cancer, Research Center in Infection Diseases, National Institute of Public Health, Av. Universidad No. 655, Cerrada los Pinos y Caminera, Colonia Santa María Ahuacatitlán, Cuernavaca 62100, Morelos, Mexico; (A.L.-M.); (V.M.-M.); (J.D.)
- Correspondence: ; Tel.: +52-777-3293000
| |
Collapse
|
43
|
Feng H, Wang N, Zhang N, Liao HH. Alternative autophagy: mechanisms and roles in different diseases. Cell Commun Signal 2022; 20:43. [PMID: 35361231 PMCID: PMC8973741 DOI: 10.1186/s12964-022-00851-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/01/2022] [Indexed: 12/11/2022] Open
Abstract
As an important mechanism to maintain cellular homeostasis, autophagy exerts critical functions via degrading misfolded proteins and damaged organelles. Recent years, alternative autophagy, a new type of autophagy has been revealed, which shares similar morphology with canonical autophagy but is independent of Atg5/Atg7. Investigations on different diseases showed the pivotal role of alternative autophagy during their physio-pathological processes, including heart diseases, neurodegenerative diseases, oncogenesis, inflammatory bowel disease (IBD), and bacterial infection. However, the studies are limited and the precise roles and mechanisms of alternative autophagy are far from clear. It is necessary to review current research on alternative autophagy and get some hint in order to provide new insight for further study. Video Abstract.
Collapse
Affiliation(s)
- Hong Feng
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Nian Wang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Nan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Hai-Han Liao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.
| |
Collapse
|