1
|
Niu X, You Q, Hou K, Tian Y, Wei P, Zhu Y, Gao B, Ashrafizadeh M, Aref AR, Kalbasi A, Cañadas I, Sethi G, Tergaonkar V, Wang L, Lin Y, Kang D, Klionsky DJ. Autophagy in cancer development, immune evasion, and drug resistance. Drug Resist Updat 2025; 78:101170. [PMID: 39603146 DOI: 10.1016/j.drup.2024.101170] [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: 07/16/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
Macroautophagy/autophagy is a highly conserved evolutionary mechanism involving lysosomes for the degradation of cytoplasmic components including organelles. The constitutive, basal level of autophagy is fundamental for preserving cellular homeostasis; however, alterations in autophagy can cause disease pathogenesis, including cancer. The role of autophagy in cancer is particularly complicated, since this process acts both as a tumor suppressor in precancerous stages but facilitates tumor progression during carcinogenesis and later stages of cancer progression. This shift between anti-tumor and pro-tumor roles may be influenced by genetic and environmental factors modulating key pathways such as those involving autophagy-related proteins, the PI3K-AKT-MTOR axis, and AMPK, which often show dysregulation in tumors. Autophagy regulates various cellular functions, including metabolism of glucose, glutamine, and lipids, cell proliferation, metastasis, and several types of cell death (apoptosis, ferroptosis, necroptosis and immunogenic cell death). These multifaceted roles demonstrate the potential of autophagy to affect DNA damage repair, cell death pathways, proliferation and survival, which are critical in determining cancer cells' response to chemotherapy. Therefore, targeting autophagy pathways presents a promising strategy to combat chemoresistance, as one of the major reasons for the failure in cancer patient treatment. Furthermore, autophagy modulates immune evasion and the function of immune cells such as T cells and dendritic cells, influencing the tumor microenvironment and cancer's biological behavior. However, the therapeutic targeting of autophagy is complex due to its dual role in promoting survival and inducing cell death in cancer cells, highlighting the need for strategies that consider both the beneficial and detrimental effects of autophagy modulation in cancer therapy. Hence, both inducers and inhibitors of autophagy have been introduced for the treatment of cancer. This review emphasizes the intricate interplay between autophagy, tumor biology, and immune responses, offering insights into potential therapeutic approaches that deploy autophagy in the cancer suppression.
Collapse
Affiliation(s)
- Xuegang Niu
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Qi You
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Kaijian Hou
- School of Public Health(Long Hu people hospital), Shantou University, Shantou, 515000, Guangdong, China
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, IL 60532, USA
| | - Penghui Wei
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yang Zhu
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Bin Gao
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
| | - Amir Reza Aref
- VitroVision Department, DeepkinetiX, Inc, Boston, MA, USA
| | - Alireza Kalbasi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Israel Cañadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Gautam Sethi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A⁎STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Lingzhi Wang
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Yuanxiang Lin
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Dezhi Kang
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Li H, Ye Z, Zheng G, Su Z. Polysaccharides targeting autophagy to alleviate metabolic syndrome. Int J Biol Macromol 2024; 283:137393. [PMID: 39521230 DOI: 10.1016/j.ijbiomac.2024.137393] [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: 04/08/2024] [Revised: 10/25/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Metabolic syndrome is a prevalent non-communicable disease characterized by central obesity, insulin resistance, hypertension, hyperglycemia, and hyperlipidemia. Epidemiological statistics indicate that one-third of the world's population is affected by metabolic syndrome. Unfortunately, owing to complicated pathogenesis and limited pharmacological options, the growing prevalence of metabolic syndrome threatens human health worldwide. Autophagy is an intracellular degradation mechanism that involves the degradation of unfolded or aggregated proteins and damaged cellular organelles, thereby maintaining metabolic homeostasis. Increasing evidence indicates that dysfunctional autophagy is closely associated with the development of metabolic syndrome, making it an attractive therapeutic target. Furthermore, a growing number of plant-derived polysaccharides have been shown to regulate autophagy, thereby alleviating metabolic syndrome, such as Astragalus polysaccharides, Laminaria japonica polysaccharides, Ganoderma lucidum polysaccharides and Lycium barbarum polysaccharides. In this review, we summarize recent advances in the discovery of autophagy modulators of plant polysaccharides for the treatment of metabolic syndrome, with the aim of providing precursor compounds for the development of new therapeutic agents. Additionally, we look forward to seeing more diseases being treated with plant polysaccharides by regulating autophagy, as well as the discovery of more intricate mechanisms that govern autophagy.
Collapse
Affiliation(s)
- Hongxia Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zeting Ye
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangjuan Zheng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Zuqing Su
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Pharmacology of Traditional Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
3
|
Li M, Liu X, Li J, Guo H, Xue S, Zhu L, Ma C, Chen D, Wang H, Cai Y, Shen J. Brain glycogen: A key to revealing the pathology of mental diseases. Brain Res 2024; 1844:149194. [PMID: 39182899 DOI: 10.1016/j.brainres.2024.149194] [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: 06/01/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Brain glycogen, which is distinct from muscle glycogen and liver glycogen, has become a crucial node linking metabolism, epigenetics, and autophagy. Recent studies have suggested that brain glycogen governs multiple neurobehavioral processes, such as memory formation and consolidation. However, the changes in brain glycogen levels in mental diseases and the associations of these changes with the disease prognosis are unknown. Here, we review the psychological functions of brain glycogen and the different characteristics of astrocytic glycogen and neuronal glycogen. In addition, we summarize the alterations in brain glycogen levels in depression, schizophrenia and sleep disorders, highlighting that brain glycogen functions as an important metabolite responsible for the development of mental diseases. In summary, brain glycogen is a key to understanding the pathology of mental diseases and deserves more attention in future research.
Collapse
Affiliation(s)
- Mingyu Li
- Graduate School, Xi׳an Medical University, Xi'an, Shaanxi, China
| | - Xiaohui Liu
- Department of Psychiatry, Xijing 986 Hospital Department, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jing Li
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haiyun Guo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shanshan Xue
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lei Zhu
- Graduate School, Xi׳an Medical University, Xi'an, Shaanxi, China
| | - Cuicui Ma
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dongyu Chen
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huaning Wang
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yanhui Cai
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Jiangpei Shen
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
4
|
Jin K, Zhao D, Zhou J, Zhang X, Wang Y, Wu Z. Pulsed electromagnetic fields inhibit IL-37 to alleviate CD8 + T cell dysfunction and suppress cervical cancer progression. Apoptosis 2024; 29:2108-2127. [PMID: 39404933 DOI: 10.1007/s10495-024-02006-8] [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] [Accepted: 07/12/2024] [Indexed: 11/10/2024]
Abstract
Pulsed electromagnetic field (PEMF) therapy is a potential non-invasive treatment to modulate immune responses and inhibit tumor growth. Cervical cancer (CC) is influenced by IL-37-mediated immune regulation, making PEMF therapy a potential strategy to impede CC progression. This study aimed to elucidate the effects of PEMF on IL-37 regulation and its molecular mechanisms in CC. CC cell-xenografted mouse models, including IL-37 transgenic (IL-37tg) mice, were used to assess tumor growth through in vivo fluorescence imaging and analyze CC cell apoptosis via flow cytometry. TCGA-CESC transcriptome and clinical data were analyzed to identify key inflammation and immune-related genes. CD8+ T cell models were stimulated with PEMF, and apoptosis, oxidative stress, and inflammatory factor expression were analyzed through RT-qPCR, Western blot, and flow cytometry. PEMF treatment significantly inhibited IL-37 expression (p < 0.05), promoted inflammatory factor release (TNF-α and IL-6), and activated oxidative stress, leading to increased CC cell apoptosis (p < 0.05). IL-37 interaction with SMAD3 impacted the p38/NF-κB signaling pathway, modulating CD8+ T cell activity and cytotoxicity. Co-culture of Hela cells with CD8+ T cells under PEMF treatment showed reduced proliferation (by 40%), migration, and invasion (p < 0.05). In vivo experiments with CC-bearing mice demonstrated that PEMF treatment downregulated IL-37 expression (p < 0.05), enhanced CD8+ T cell function, and inhibited tumor growth (p < 0.05). These molecular mechanisms were validated through RT-qPCR, Western blot, and immunohistochemistry. Thus, PEMF therapy inhibits CC progression by downregulating IL-37 and improving CD8+ T cell function via the SMAD3/p38/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Ke Jin
- Department of Oncology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Dan Zhao
- Department of Oncology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jun Zhou
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xun Zhang
- Department of Obstetrics and Gynecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West 2nd Section, First Ring Road, Qingyang District, Chengdu, 610072, Sichuan Province, China
| | - Yujue Wang
- Department of Obstetrics and Gynecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West 2nd Section, First Ring Road, Qingyang District, Chengdu, 610072, Sichuan Province, China.
| | - Zhao Wu
- Department of Obstetrics and Gynecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West 2nd Section, First Ring Road, Qingyang District, Chengdu, 610072, Sichuan Province, China.
| |
Collapse
|
5
|
Zhang P, Zhang W, Hong Z, Jiang J, Wu N, Lin J, Kang M. Elucidating the role of CYFIP2 in conferring cisplatin resistance in esophageal squamous cell carcinoma. Sci Rep 2024; 14:27130. [PMID: 39511293 PMCID: PMC11544133 DOI: 10.1038/s41598-024-77420-8] [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: 01/24/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024] Open
Abstract
Cisplatin (CDDP) serves as a vital component in the chemotherapeutic approach to treat esophageal squamous cell carcinoma (ESCC). However, prolonged CDDP application frequently culminates in resistance, compromising therapeutic outcomes. Through genome-wide CRISPR library screening, our study elucidates the mechanisms underlying this resistance, pinpointing CYFIP2 as a pivotal mediator. Notably, the involvement CYFIP2 is characterized by pronounced autophagic activity and the modulation of multiple cellular pathways. Empirical validation was achieved by treating ESCC cell lines with CDDP, which resulted in an upsurge of CYFIP2 expression. The functional impact of CYFIP2 was further delineated through knockdown experiments, where a marked suppression in cell proliferation was observed, alongside a discernible decline in reactive oxygen species levels. This was complemented by a suite of assays and microscopic techniques, including GFP-LC3, mRFP-GFP-LC3, electron microscopy and western blot, which collectively affirmed the inhibitory effect of CYFIP2 knockdown on autophagic processes, particularly impeding autophagosome formation and their subsequent fusion with lysosomes. In vivo studies have also confirmed that CYFIP2 knockdown limits tumor progression and increases CDDP efficacy. Conclusively, our findings introduce CYFIP2 as a novel contributor to CDDP resistance in ESCC, underscoring its potential as a therapeutic target. This revelation not only deepens our understanding of resistance mechanisms but also paves the way for novel oncotherapeutic strategies, promising enhanced treatment efficacy against ESCC.
Collapse
Affiliation(s)
- Peipei Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China
| | - Weiguang Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China
| | - Zhinuan Hong
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China
| | - Junfei Jiang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China
| | - Ningzi Wu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China
| | - Jihong Lin
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China.
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China.
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China.
| |
Collapse
|
6
|
Tang S, Chen L. The recent advancements of ferroptosis of gynecological cancer. Cancer Cell Int 2024; 24:351. [PMID: 39462352 PMCID: PMC11520064 DOI: 10.1186/s12935-024-03537-5] [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: 04/07/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024] Open
Abstract
Ovarian, endometrial, and cervical cancer are the most common types of gynecologic tumor in women. Surgery, combined with radiotherapy and chemotherapy, is commonly used to treat these tumors. Unfortunately, difficulties in early diagnosis and acquired drug resistance have resulted in poor outcomes for most patients. Ferroptosis is a form of regulated cell death that depends on iron and is characterized by iron accumulation, reactive oxygen species production, and lipid peroxidation. The strong association between ferroptosis and many diseases, especially tumor diseases, has been confirmed by numerous studies. Many studies have demonstrated that ferroptosis is involved in initiating, progressing and metastasizing gynecologic tumors. This review summarizes the pathogenesis of ferroptosis and its association with the development, treatment, and prognosis of gynecologic tumors, and further explore the potential utility of ferroptosis in treating gynecologic tumors.
Collapse
Affiliation(s)
- Shenglan Tang
- Department of the First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People's Republic of China
| | - Li Chen
- Department of Obstetrics and Gynecology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Road, Shangcheng, Hangzhou, Zhejiang, 310006, People's Republic of China.
| |
Collapse
|
7
|
Wang C, Li M, Huang S, Huang W, He T, Wusiman M, Zhu H, Liu Z. Choline ameliorates tris (2‐chloroisopropyl) phosphate‐induced hepatocellular carcinoma metastasis by inhibiting ROS/Nrf2/Keap1‐mediated autophagy. FOOD FRONTIERS 2024. [DOI: 10.1002/fft2.489] [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] Open
Abstract
AbstractTris (2‐chloroisopropyl) phosphate (TCPP) is an emerging environmental pollutant associated with liver diseases. However, its effects on hepatocellular carcinoma (HCC) remain unknown. Choline, a necessary dietary nutrient, has previously demonstrated inhibitory effects on HCC. Therefore, elucidating the underlying mechanism of TCPP exposure on HCC development and investigating whether choline could mitigate these effects may improve the prognosis of HCC patients. In this study, we examined the tumor‐promoting effects of TCPP on HCC and explored the protective effects of choline. Our findings revealed that choline treatment attenuated the tumor‐promoting effects of TCPP exposure on HCC cells’ epithelial‐mesenchymal transition (EMT) and lung metastasis. Further investigation showed that TCPP exposure induced ROS production via NOX4 upregulation, while choline inhibited ROS generation, thereby mitigating the effects of TCPP on EMT and metastasis in HCC cells. Mechanistic analysis demonstrated that excessive ROS inhibited levels of Keap1, leading to upregulation and nuclear translocation of Nrf2, which promoted autophagy flux and accelerated EMT and metastasis of HCC cells. However, choline treatment significantly impaired TCPP‐induced autophagy by attenuating the ROS/Nrf2/Keap1 pathway. Overall, our data illustrate the adverse effects of TCPP on the malignant progression of HCC and suggest that choline may serve as a potential nutrient to counteract the tumor‐promoting effects of TCPP on HCC.
Collapse
Affiliation(s)
- Chen Wang
- Department of Nutrition, School of Public Health Sun Yat‐sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health Sun Yat‐sen University Guangzhou China
| | - Meng‐chu Li
- Department of Nutrition, School of Public Health Sun Yat‐sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health Sun Yat‐sen University Guangzhou China
| | - Si‐yu Huang
- Department of Nutrition, School of Public Health Sun Yat‐sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health Sun Yat‐sen University Guangzhou China
| | - Wen‐ge Huang
- Center of Experimental Animals Sun Yat‐sen University Guangzhou Guangdong Province China
| | - Tong‐tong He
- Department of Nutrition, School of Public Health Sun Yat‐sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health Sun Yat‐sen University Guangzhou China
| | - Maierhaba Wusiman
- Department of Nutrition, School of Public Health Sun Yat‐sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health Sun Yat‐sen University Guangzhou China
| | - Hui‐lian Zhu
- Department of Nutrition, School of Public Health Sun Yat‐sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health Sun Yat‐sen University Guangzhou China
| | - Zhao‐yan Liu
- Department of Nutrition, School of Public Health Sun Yat‐sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health Sun Yat‐sen University Guangzhou China
| |
Collapse
|
8
|
Peng Y, Yang Q. Targeting KRAS in gynecological malignancies. FASEB J 2024; 38:e70089. [PMID: 39377766 DOI: 10.1096/fj.202401734r] [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: 07/28/2024] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024]
Abstract
Cervical, endometrial, and ovarian cancers stand prominently as the leading gynecological malignancies of the female reproductive system. The conventional therapeutic modalities for gynecological malignancies have predominantly encompassed surgery, chemotherapy, and radiotherapy. However, efficacy of these approaches remains limited in cases of relapse or drug resistance. KRAS is one of the most frequently mutated oncogenes in human cancers. The KRAS gene encodes a small guanosine triphosphatase protein that acts as a molecular switch for crucial intracellular signaling pathways. KRAS mutations are deeply involved in the occurrence and development of gynecological malignancies. The present review aims to expound upon the role of oncogenic KRAS as a biomarker, elucidating various therapeutic approaches under investigation targeting the KRAS pathway in gynecological tumors.
Collapse
Affiliation(s)
- Yuanyuan Peng
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
9
|
Ma X, Huang T, Li X, Zhou X, Pan H, Du A, Zeng Y, Yuan K, Wang Z. Exploration of the link between COVID-19 and gastric cancer from the perspective of bioinformatics and systems biology. Front Med (Lausanne) 2024; 11:1428973. [PMID: 39371335 PMCID: PMC11449776 DOI: 10.3389/fmed.2024.1428973] [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: 05/07/2024] [Accepted: 09/04/2024] [Indexed: 10/08/2024] Open
Abstract
Background Coronavirus disease 2019 (COVID-19), an infectious disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has caused a global pandemic. Gastric cancer (GC) poses a great threat to people's health, which is a high-risk factor for COVID-19. Previous studies have found some associations between GC and COVID-19, whereas the underlying molecular mechanisms are not well understood. Methods We employed bioinformatics and systems biology to explore these links between GC and COVID-19. Gene expression profiles of COVID-19 (GSE196822) and GC (GSE179252) were obtained from the Gene Expression Omnibus (GEO) database. After identifying the shared differentially expressed genes (DEGs) for GC and COVID-19, functional annotation, protein-protein interaction (PPI) network, hub genes, transcriptional regulatory networks and candidate drugs were analyzed. Results We identified 209 shared DEGs between COVID-19 and GC. Functional analyses highlighted immune-related pathways as key players in both diseases. Ten hub genes (CDK1, KIF20A, TPX2, UBE2C, HJURP, CENPA, PLK1, MKI67, IFI6, IFIT2) were identified. The transcription factor/gene and miRNA/gene interaction networks identified 38 transcription factors (TFs) and 234 miRNAs. More importantly, we identified ten potential therapeutic agents, including ciclopirox, resveratrol, etoposide, methotrexate, trifluridine, enterolactone, troglitazone, calcitriol, dasatinib and deferoxamine, some of which have been reported to improve and treat GC and COVID-19. Conclusion This research offer valuable insights into the molecular interplay between COVID-19 and GC, potentially guiding future therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhen Wang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Koutsifeli P, Daniels LJ, Neale J, Fong S, Varma U, Annandale M, Li X, Nursalim Y, Bell JR, Weeks KL, Stotland A, Taylor DJ, Gottlieb RA, Delbridge LMD, Mellor KM. Methods for detection of cardiac glycogen-autophagy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.11.607511. [PMID: 39211188 PMCID: PMC11361060 DOI: 10.1101/2024.08.11.607511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Glycogen-autophagy ('glycophagy') is a selective autophagy process involved in delivering glycogen to the lysosome for bulk degradation. Glycophagy protein intermediaries include STBD1 as a glycogen tagging receptor, delivering the glycogen cargo into the forming phagosome by partnering with the Atg8 homolog, GABARAPL1. Glycophagy is emerging as a key process of energy metabolism and development of reliable tools for assessment of glycophagy activity is an important priority. Here we show that antibodies raised against the N-terminus of the GABARAPL1 protein (but not the full-length protein) detected a specific endogenous GABARAPL1 immunoblot band at 18kDa. A stable GFP-GABARAPL1 cardiac cell line was used to quantify GABARAPL1 lysosomal flux via measurement of GFP puncta in response to lysosomal inhibition with bafilomycin. Endogenous glycophagy flux was quantified in primary rat ventricular myocytes by the extent of glycogen accumulation with bafilomycin combined with chloroquine treatment (no effect observed with bafilomycin or chloroquine alone). In wild-type isolated mouse hearts, bafilomycin alone and bafilomycin combined with chloroquine (but not chloroquine alone) elicited a significant increase in glycogen content signifying basal glycophagy flux. Collectively, these methodologies provide a comprehensive toolbox for tracking cardiac glycophagy activity to advance research into the role of glycophagy in health and disease.
Collapse
|
11
|
Jia Y, Li Y, Du N, Zhao W, Liu Y. LIMK1 promotes the development of cervical cancer by up-regulating the ROS/Src-FAK/cofilin signaling pathway. Aging (Albany NY) 2024; 16:11090-11102. [PMID: 38975937 PMCID: PMC11272116 DOI: 10.18632/aging.206007] [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: 09/04/2023] [Accepted: 06/10/2024] [Indexed: 07/09/2024]
Abstract
OBJECTIVE In this study, we investigated the mechanism of action of LIMK1 in cervical cancer progression. METHODS The biological role of LIMK1 in regulating the growth, invasion, and metastasis of cervical cancer was studied in SiHa, CaSki cells and nude mice tumor models. The role of LIMK1 in the growth of cervical cancer was evaluated by HE staining. The role of LIMK1 in the invasion, metastasis, and proliferation of cervical cancer was evaluated by cell scratch, Transwell, and monoclonal experiments. The interaction among LIMK1, ROS, and Src was evaluated by Western blotting. The effects of regulating ROS and p-Src expression on LIMK1 in the migration/invasion and proliferation of cervical cancer cells were evaluated through cellular functional assays. RESULTS Overexpression of LIMK1 promoted tumor growth in nude mice. Cell scratch, Transwell, and monoclonal experiments suggested that LIMK1 promoted the invasion, metastasis, and proliferation of cervical cancer cells. Western blotting suggested that LIMK1 can promote the expression of ROS-related proteins NOX2, NOX4, p-Src, and downstream proteins p-FAK, p-ROCK1/2, p-Cofilin-1, F-actin and inhibit the expression of p-SHP2 protein. Correction experiments showed that LIMK1 regulated the expression of p-FAK and p-Cofilin-1 proteins by regulating ROS and p-Src. Through the detection of cervical cancer cell functions, it was found that the activation of ROS and p-Src induced by LIMK1 is an early event that promotes the migration, proliferation, and invasion of cervical cancer cells. CONCLUSIONS LIMK1 promotes the expression of F-actin and promotes the development of cervical cancer by regulating the oxidative stress/Src-mediated p-FAK/p-ROCK1/2/p-Cofilin-1 pathway.
Collapse
Affiliation(s)
- Yajing Jia
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Yongping Li
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Naiyi Du
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Wei Zhao
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Yakun Liu
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| |
Collapse
|
12
|
Li C, Xue Y, Wu J, Zhang L, Yang T, Ai M, Han J, Zheng X, Wang R, Boldogh I, Ba X. MTH1 inhibition synergizes with ROS-inducing agents to trigger cervical cancer cells undergoing parthanatos. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167190. [PMID: 38657912 DOI: 10.1016/j.bbadis.2024.167190] [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: 09/21/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Cervical cancer cells possess high levels of reactive oxygen species (ROS); thus, increasing oxidative stress above the toxicity threshold to induce cell death is a promising chemotherapeutic strategy. However, the underlying mechanisms of cell death are elusive, and efficacy and toxicity issues remain. Within DNA, 8-oxo-7,8-dihydroguanine (8-oxoG) is the most frequent base lesion repaired by 8-oxoguanine glycosylase 1 (OGG1)-initiated base excision repair. Cancer cells also express high levels of MutT homolog 1 (MTH1), which prevents DNA replication-induced incorporation of 8-oxoG into the genome by hydrolyzing 8-oxo-7,8-dihydro-2'-deoxyguanosine 5'-triphosphate (8-oxo-dGTP). Here, we revealed that ROS-inducing agents triggered cervical cancer to undergo parthanatos, which was mainly induced by massive DNA strand breaks resulting from overwhelming 8-oxoG excision by OGG1. Furthermore, the MTH1 inhibitor synergized with a relatively low dose of ROS-inducing agents by enhancing 8-oxoG loading in the DNA. In vivo, this drug combination suppressed the growth of tumor xenografts, and this inhibitory effect was significantly decreased in the absence of OGG1. Hence, the present study highlights the roles of base repair enzymes in cell death induction and suggests that the combination of lower doses of ROS-inducing agents with MTH1 inhibitors may be a more selective and safer strategy for cervical cancer chemotherapy.
Collapse
Affiliation(s)
- Chunshuang Li
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Yaoyao Xue
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Jiaxin Wu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Lihong Zhang
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Tianming Yang
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Mengtao Ai
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Jinling Han
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Xu Zheng
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Ruoxi Wang
- Key Laboratory of Animal Resistance Biology of Shandong Province, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Xueqing Ba
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China.
| |
Collapse
|
13
|
Qian Z, Wu F, Feng G, Lin W, Cai X, Wu J, Ke K, Ye Z, Xu G. A prognostic risk model based on lactate metabolism and transport-related lncRNAs for gastric adenocarcinoma. Biomarkers 2024; 29:211-221. [PMID: 38629165 DOI: 10.1080/1354750x.2024.2341411] [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: 09/17/2023] [Accepted: 04/04/2024] [Indexed: 05/15/2024]
Abstract
BACKGROUND Increased lactate levels and metastasis in tumours are strongly associated with dismal outcomes. But prognostic value of lactate metabolism and transport-related lncRNAs in gastric adenocarcinoma (GA) patients remains unaddressed. METHODS Gene expression data of GA were provided by The Cancer Genome Atlas. Lactate metabolism and transport-related gene data were accessed from GSEA. LncRNAs related to lactate metabolism and transport were identified by correlation analysis. A prognostic model was built by regression analysis. Validity of prognostic model was confirmed through survival analysis and receiver operating characteristic (ROC) curve. Immunity of each risk group was evaluated by immune correlation analysis .LncRNA-mRNA network was built by correlation analysis using Cytoscape software. RESULTS A 12-gene prognostic model based on lactate metabolism and transport-related lncRNAs was built in GA. Median riskscore was utilized to classify GA samples into high- and low-risk groups. Survival analysis and ROC curves demonstrated validity of prognostic model. Most immune checkpoint molecules and TIDE scores were lower in the low-risk group. LINC01303 and LINC01545 may be the key prognostic factors in patients with GA. CONCLUSION This study successfully built a prognostic model of lactate metabolism and transport-related lncRNAs in GA. The findings guide prognostic management of GA patients.
Collapse
Affiliation(s)
- Zhenyuan Qian
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Fang Wu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Guoqing Feng
- Department of General Surgery, Haining Traditional Chinese Medicine Hospital, Haining, Jiaxing, Zhejiang, China
| | - Wenfa Lin
- Zhejiang Chinese Medicine University, Hangzhou, Zhejiang, China
| | - Xufan Cai
- Zhejiang Chinese Medicine University, Hangzhou, Zhejiang, China
| | - Jianzhang Wu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kun Ke
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zaiyuan Ye
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Guoxi Xu
- Department of Gastrointestinal Surgery, Jinjiang Municipal Hospital, Jinjiang, Quanzhou, Fujian, China
| |
Collapse
|
14
|
Liu Y, Sun Y, Yang J, Wu D, Yu S, Liu J, Hu T, Luo J, Zhou H. DNMT1-targeting remodeling global DNA hypomethylation for enhanced tumor suppression and circumvented toxicity in oral squamous cell carcinoma. Mol Cancer 2024; 23:104. [PMID: 38755637 PMCID: PMC11097543 DOI: 10.1186/s12943-024-01993-1] [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: 12/19/2023] [Accepted: 04/03/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND The faithful maintenance of DNA methylation homeostasis indispensably requires DNA methyltransferase 1 (DNMT1) in cancer progression. We previously identified DNMT1 as a potential candidate target for oral squamous cell carcinoma (OSCC). However, how the DNMT1- associated global DNA methylation is exploited to regulate OSCC remains unclear. METHODS The shRNA-specific DNMT1 knockdown was employed to target DNMT1 on oral cancer cells in vitro, as was the use of DNMT1 inhibitors. A xenografted OSCC mouse model was established to determine the effect on tumor suppression. High-throughput microarrays of DNA methylation, bulk and single-cell RNA sequencing analysis, multiplex immunohistochemistry, functional sphere formation and protein immunoblotting were utilized to explore the molecular mechanism involved. Analysis of human samples revealed associations between DNMT1 expression, global DNA methylation and collaborative molecular signaling with oral malignant transformation. RESULTS We investigated DNMT1 expression boosted steadily during oral malignant transformation in human samples, and its inhibition considerably minimized the tumorigenicity in vitro and in a xenografted OSCC model. DNMT1 overexpression was accompanied by the accumulation of cancer-specific DNA hypomethylation during oral carcinogenesis; conversely, DNMT1 knockdown caused atypically extensive genome-wide DNA hypomethylation in cancer cells and xenografted tumors. This novel DNMT1-remodeled DNA hypomethylation pattern hampered the dual activation of PI3K-AKT and CDK2-Rb and inactivated GSK3β collaboratively. When treating OSCC mice, targeting DNMT1 achieved greater anticancer efficacy than the PI3K inhibitor, and reduced the toxicity of blood glucose changes caused by the PI3K inhibitor or combination of PI3K and CDK inhibitors as well as adverse insulin feedback. CONCLUSIONS Targeting DNMT1 remodels a novel global DNA hypomethylation pattern to facilitate anticancer efficacy and minimize potential toxic effects via balanced signaling synergia. Our study suggests DNMT1 is a crucial gatekeeper regarding OSCC destiny and treatment outcome.
Collapse
Affiliation(s)
- Yangfan Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yu Sun
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- School of Stomatology, Hainan Medical University, Haikou, 571199, Hainan, China
| | - Jin Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Deyang Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Shuang Yu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Junjiang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Tao Hu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jingjing Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Hongmei Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
15
|
Liu H, Wang X, He K, Chen Z, Li X, Ren J, Zhao X, Liu S, Zhou T, Chen H. Oxidized DJ-1 activates the p-IKK/NF-κB/Beclin1 pathway by binding PTEN to induce autophagy and exacerbate myocardial ischemia-reperfusion injury. Eur J Pharmacol 2024; 971:176496. [PMID: 38508437 DOI: 10.1016/j.ejphar.2024.176496] [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: 01/21/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/22/2024]
Abstract
Patients with myocardial infarction have a much worse prognosis when they have myocardial ischemia-reperfusion (I/R) injury. Further research into the molecular basis of myocardial I/R injury is therefore urgently needed, as well as the identification of novel therapeutic targets and linkages to interventions. Three cysteine residues are present in DJ-1 at amino acids 46, 53, and 106 sites, with the cysteine at position 106 being the most oxidation-prone. This study sought to understand how oxidized DJ-1(C106) contributes to myocardial I/R damage. Rats' left anterior descending branches were tied off to establish a myocardial I/R model in vivo. A myocardial I/R model in vitro was established via anoxia/reoxygenation (A/R) of H9c2 cells. The results showed that autophagy increased after I/R, accompanied by the increased expression of oxidized DJ-1 (ox-DJ-1). In contrast, after pretreatment with NAC (N-acetylcysteine, a ROS scavenger) or Comp-23 (Compound-23, a specific antioxidant binding to the C106 site of DJ-1), the levels of ox-DJ-1, autophagy and LDH release decreased, and cell survival rate increased. Furthermore, the inhibition of interaction between ox-DJ-1 and PTEN could increase PTEN phosphatase activity, inhibit the p-IKK/NF-κB/Beclin1 pathway, reduce injurious autophagy, and alleviate A/R injury. However, BA (Betulinic acid, a NF-κB agonist) was able to reverse the protective effects produced by Comp-23 pretreatment. In conclusion, ox-DJ-1 could activate detrimental autophagy through the PTEN/p-IKK/NF-κB/Beclin1 pathway and exacerbate myocardial I/R injury.
Collapse
Affiliation(s)
- Huiru Liu
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Xueying Wang
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330004, PR China
| | - Kang He
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Zihan Chen
- Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Xiaoqi Li
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Jianmin Ren
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Xiaoyan Zhao
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, PR China
| | - Song Liu
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Tingting Zhou
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, PR China
| | - Heping Chen
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China.
| |
Collapse
|
16
|
Yang Y, Liu L, Tian Y, Gu M, Wang Y, Ashrafizadeh M, Reza Aref A, Cañadas I, Klionsky DJ, Goel A, Reiter RJ, Wang Y, Tambuwala M, Zou J. Autophagy-driven regulation of cisplatin response in human cancers: Exploring molecular and cell death dynamics. Cancer Lett 2024; 587:216659. [PMID: 38367897 DOI: 10.1016/j.canlet.2024.216659] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/29/2023] [Accepted: 01/17/2024] [Indexed: 02/19/2024]
Abstract
Despite the challenges posed by drug resistance and side effects, chemotherapy remains a pivotal strategy in cancer treatment. A key issue in this context is macroautophagy (commonly known as autophagy), a dysregulated cell death mechanism often observed during chemotherapy. Autophagy plays a cytoprotective role by maintaining cellular homeostasis and recycling organelles, and emerging evidence points to its significant role in promoting cancer progression. Cisplatin, a DNA-intercalating agent known for inducing cell death and cell cycle arrest, often encounters resistance in chemotherapy treatments. Recent studies have shown that autophagy can contribute to cisplatin resistance or insensitivity in tumor cells through various mechanisms. This resistance can be mediated by protective autophagy, which suppresses apoptosis. Additionally, autophagy-related changes in tumor cell metastasis, particularly the induction of Epithelial-Mesenchymal Transition (EMT), can also lead to cisplatin resistance. Nevertheless, pharmacological strategies targeting the regulation of autophagy and apoptosis offer promising avenues to enhance cisplatin sensitivity in cancer therapy. Notably, numerous non-coding RNAs have been identified as regulators of autophagy in the context of cisplatin chemotherapy. Thus, therapeutic targeting of autophagy or its associated pathways holds potential for restoring cisplatin sensitivity, highlighting an important direction for future clinical research.
Collapse
Affiliation(s)
- Yang Yang
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Lixia Liu
- Department of Ultrasound, Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, IL, USA
| | - Miaomiao Gu
- Department of Ultrasound, Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yanan Wang
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | - Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440 Ji Yan Road, Jinan, Shandong, China
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Translational Sciences, Xsphera Biosciences Inc, 6, Tide Street, Boston, MA, 02210, USA
| | - Israel Cañadas
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA, USA; Nuclear Dynamics and Cancer Program, Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Arul Goel
- University of California Santa Barbara, Santa Barbara, CA, USA
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health, Long School of Medicine, San Antonio, TX, 78229, USA
| | - Yuzhuo Wang
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Murtaza Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln, LN6 7TS, UK.
| | - Jianyong Zou
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, China.
| |
Collapse
|
17
|
Li J, Jiang H, Zhu Y, Ma Z, Li B, Dong J, Xiao C, Hu A. Fine particulate matter (PM 2.5) induces the stem cell-like properties of hepatocellular carcinoma by activating ROS/Nrf2/Keap1-mediated autophagy. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116052. [PMID: 38325274 DOI: 10.1016/j.ecoenv.2024.116052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/18/2024] [Accepted: 01/27/2024] [Indexed: 02/09/2024]
Abstract
Exposure to fine particulate matter (PM2.5) has been linked to an increased incidence and mortality of hepatocellular carcinoma (HCC). However, the impact of PM2.5 exposure on HCC progression and the underlying mechanisms remain largely unknown. This study aimed to investigate the effects of PM2.5 exposure on the stem cell-like properties of HCC cells. Our findings indicate that PM2.5 exposure significantly enhances the stemness of HCC cells (p < 0.01). Subsequently, male nude mice were divided into two groups (n = 8/group for tumor-bearing assay, n = 5/group for metastasis assay) for control and PM2.5 exposure. In vivo assays revealed that exposure to PM2.5 promoted the growth, metastasis, and epithelial-mesenchymal transition (EMT) of HCC cells (p < 0.01). Further exploration demonstrated that PM2.5 enhances the stemness of HCC cells by inducing cellular reactive oxygen species (ROS) generation (p < 0.05). Mechanistic investigation indicated that elevated intracellular ROS inhibited kelch-like ECH-associated protein 1 (Keap1) levels, promoting the upregulation and nucleus translocation of NFE2-like bZIP transcription factor 2 (Nrf2). This, in turn, induced autophagy activation, thereby promoting the stemness of HCC cells (p < 0.01). Our present study demonstrates the adverse effects of PM2.5 exposure on HCC development and highlights the mechanism of ROS/Nrf2/Keap1-mediated autophagy. For the first time, we reveal the impact of PM2.5 exposure on the poor prognosis-associated cellular phenotype of HCC and its underlying mechanism, which is expected to provide new theoretical basis for the improvement of public health.
Collapse
Affiliation(s)
- Jiujiu Li
- Hefei Center for Disease Control and Prevention, Hefei 230032, China
| | - Haoqi Jiang
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Yu Zhu
- Hefei Center for Disease Control and Prevention, Hefei 230032, China
| | - Zijian Ma
- Hefei Center for Disease Control and Prevention, Hefei 230032, China
| | - Bin Li
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Jun Dong
- Hefei Center for Disease Control and Prevention, Hefei 230032, China
| | - Changchun Xiao
- Hefei Center for Disease Control and Prevention, Hefei 230032, China.
| | - Anla Hu
- School of Public Health, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
18
|
Meng Y, Liu Y, Zhu Y, Qiu C, Ma A, Liu D, Zhang S, Gu L, Zhang J, Wang J. Insights into the time-course cellular effects triggered by iron oxide nanoparticles by combining proteomics with the traditional pharmacology strategy. J Mater Chem B 2024; 12:1892-1904. [PMID: 38305086 DOI: 10.1039/d3tb02476h] [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: 02/03/2024]
Abstract
In recent years, a number of initially approved magnetic iron oxide nanoparticle (IONP)-based nano-medicines have been withdrawn due to the obscure nano-bio effects. Therefore, there is an urgent need to study the cellular effects triggered by IONPs on cells. In this study, we investigate the time-course cellular effects on the response of RAW 264.7 cells caused by Si-IONPs via pharmacological and mass spectrometry-based proteomics techniques. Our results revealed that Si-IONPs were internalized by clathrin-mediated endocytosis within 1 hour, and gradually degraded in endolysosomes over time, which might influence autophagy, oxidative stress, innate immune response, and inflammatory response after 12 hours. Our research provides a necessary assessment of Si-IONPs for further clinical treatment.
Collapse
Affiliation(s)
- Yuqing Meng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Yanqing Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Yongping Zhu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Chong Qiu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Ang Ma
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Dandan Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Shujie Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Liwei Gu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Jigang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| |
Collapse
|
19
|
Zhang C, Liu L, Li W, Li M, Zhang X, Zhang C, Yang H, Xie J, Pan W, Guo X, She P, Zhong L, Li T. Upregulation of FAM83F by c-Myc promotes cervical cancer growth and aerobic glycolysis via Wnt/β-catenin signaling activation. Cell Death Dis 2023; 14:837. [PMID: 38104106 PMCID: PMC10725447 DOI: 10.1038/s41419-023-06377-9] [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: 07/13/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
Cervical cancer (CC) seriously affects women's health. Therefore, elucidation of the exact mechanisms and identification of novel therapeutic targets are urgently needed. In this study, we identified FAM83F, which was highly expressed in CC cells and tissues, as a potential target. Our clinical data revealed that FAM83F protein expression was markedly elevated in CC tissues and was positively correlated with poor prognosis. Moreover, we observed that FAM83F knockdown significantly inhibited cell proliferation, induced apoptosis, and suppressed glycolysis in CC cells, while its overexpression displayed opposite effects. Mechanistically, FAM83F regulated CC cell growth and glycolysis by the modulation of Wnt/β-catenin pathway. The enhancing effects of FAM83F overexpression on CC cell proliferation and glycolysis could be impaired by the Wnt/β-catenin inhibitor XAV939. Moreover, we found that c-Myc bound to the FAM83F promoter and activated the transcription of FAM83F. Notably, knockdown of FAM83F impaired the enhancement of cell proliferation and glycolysis induced by ectopic c-Myc. Consistent with in vitro findings, results from a xenograft mouse model confirmed the promoting role of FAM83F. In summary, our study demonstrated that FAM83F promoted CC growth and glycolysis through regulating the Wnt/β-catenin pathway, suggesting that FAM83F may be a potential molecular target for CC treatment. Schematic summary of c-Myc-activated FAM83F transcription to promote cervical cancer growth and glycolysis by targeting the Wnt/β-catenin signal pathway.
Collapse
Affiliation(s)
- Changlin Zhang
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, China
| | - Lixiang Liu
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Weizhao Li
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Mengxiong Li
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xunzhi Zhang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Chi Zhang
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Huan Yang
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jiayuan Xie
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Wei Pan
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xue Guo
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Peng She
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Li Zhong
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Tian Li
- Department of Gynecology, Pelvic Floor Disorders Center, Department of Orthopedics, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, China.
| |
Collapse
|
20
|
Mancini MC, Noland RC, Collier JJ, Burke SJ, Stadler K, Heden TD. Lysosomal glucose sensing and glycophagy in metabolism. Trends Endocrinol Metab 2023; 34:764-777. [PMID: 37633800 PMCID: PMC10592240 DOI: 10.1016/j.tem.2023.07.008] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/28/2023]
Abstract
Lysosomes are cellular organelles that function to catabolize both extra- and intracellular cargo, act as a platform for nutrient sensing, and represent a core signaling node integrating bioenergetic cues to changes in cellular metabolism. Although lysosomal amino acid and lipid sensing in metabolism has been well characterized, lysosomal glucose sensing and the role of lysosomes in glucose metabolism is unrefined. This review will highlight the role of the lysosome in glucose metabolism with a focus on lysosomal glucose and glycogen sensing, glycophagy, and lysosomal glucose transport and how these processes impact autophagy and energy metabolism. Additionally, the role of lysosomal glucose metabolism in genetic and metabolic diseases will be briefly discussed.
Collapse
Affiliation(s)
- Melina C Mancini
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Robert C Noland
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - J Jason Collier
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Susan J Burke
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | | | - Timothy D Heden
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| |
Collapse
|
21
|
Tang Q, Liu M, Zhao H, Chen L. Glycogen-binding protein STBD1: Molecule and role in pathophysiology. J Cell Physiol 2023; 238:2010-2025. [PMID: 37435888 DOI: 10.1002/jcp.31078] [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: 02/21/2023] [Revised: 06/19/2023] [Accepted: 06/23/2023] [Indexed: 07/13/2023]
Abstract
Starch-binding domain-containing protein 1 (STBD1) is a glycogen-binding protein discovered in skeletal muscle gene differential expression that is pivotal to cellular energy metabolism. Recent studies have indicated that STBD1 is involved in many physiological processes, such as glycophagy, glycogen accumulation, and lipid droplet formation. Moreover, dysregulation of STBD1 causes multiple diseases, including cardiovascular disease, metabolic disease, and even cancer. Deletions and/or mutations in STBD1 promote tumorigenesis. Therefore, STBD1 has garnered considerable interest in the pathology community. In this review, we first summarized the current understanding of STBD1, including its structure, subcellular localization, tissue distribution, and biological functions. Next, we examined the roles and molecular mechanisms of STBD1 in related diseases. Based on available research, we discussed the novel function and future of STBD1, including its potential application as a therapeutic target in glycogen-related diseases. Given the significance of STBD1 in energy metabolism, an in-depth understanding of the protein is crucial for understanding physiological processes and developing therapeutic strategies for related diseases.
Collapse
Affiliation(s)
- Qiannan Tang
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, China
| | - Meiqing Liu
- Key Laboratory of Cardiovascular Diseases of Yunnan Province, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Central Laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Hong Zhao
- Nursing College, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Linxi Chen
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
22
|
Qin Y, Ashrafizadeh M, Mongiardini V, Grimaldi B, Crea F, Rietdorf K, Győrffy B, Klionsky DJ, Ren J, Zhang W, Zhang X. Autophagy and cancer drug resistance in dialogue: Pre-clinical and clinical evidence. Cancer Lett 2023; 570:216307. [PMID: 37451426 DOI: 10.1016/j.canlet.2023.216307] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
The emergence of drug resistance is a major challenge for oncologists. Resistance can be categorized as acquired or intrinsic; the alteration of several biological mechanisms contributes to both intrinsic and acquired resistance. Macroautophagy/autophagy is the primary process in eukaryotes for the degradation of macromolecules and organelles. This process is critical in maintaining cellular homeostasis. Given its function as either a pro-survival or a pro-death phenomenon, autophagy has a complex physio-pathological role. In some circumstances, autophagy can confer chemoresistance and promote cell survival, whereas in others it can promote chemosensitivity and contribute to cell death. The role of autophagy in the modulation of cancer drug resistance reflects its impact on apoptosis and metastasis. The regulation of autophagy in cancer is mediated by various factors including AMP-activated protein kinase (AMPK), MAPK, phosphoinositide 3-kinase (PI3K)-AKT, BECN1 and ATG proteins. Non-coding RNAs are among the main regulators of autophagy, e.g., via the modulation of chemoresistance pathways. Due to the significant contribution of autophagy in cancer drug resistance, small molecule modulators and natural compounds targeting autophagy have been introduced to alter the response of cancer cells to chemotherapy. Furthermore, nanotherapeutic approaches based on autophagy regulation have been introduced in pre-clinical cancer therapy. In this review we consider the potential for using autophagy regulators for the clinical treatment of malignancies.
Collapse
Affiliation(s)
- Yi Qin
- Department of Lab, Chifeng Cancer Hospital (The 2nd Affliated Hospital of Chifeng University), Chifeng University, Chifeng City, Inner Mongolia Autonomous Region, 024000, China.
| | - Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Vera Mongiardini
- Molecular Medicine Research Line, Fondazione Istituto Italiano di Tecnologia (IIT), Genoa, 16163, Italy
| | - Benedetto Grimaldi
- Molecular Medicine Research Line, Fondazione Istituto Italiano di Tecnologia (IIT), Genoa, 16163, Italy
| | - Francesco Crea
- Cancer Research Group-School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Katja Rietdorf
- Cancer Research Group-School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Tüzoltó u. 7-9, 1094, Budapest, Hungary; Department of Pediatrics, Semmelweis University, Tüzoltó u. 7-9, 1094, Budapest, Hungary; Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, Magyar tudosok korutja 2, 1117, Budapest, Hungary
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wei Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Xianbin Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
23
|
Zhu H, Gao X, Wang B, Niu B, Liu D, Zhang J, Jin Y, Yang X. A biodegradable hollow nanoagent enables a boosted chemodynamic therapy by simultaneous autophagy inhibition and macrophage reeducation. Int J Pharm 2023; 643:123248. [PMID: 37467817 DOI: 10.1016/j.ijpharm.2023.123248] [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: 01/30/2023] [Revised: 06/20/2023] [Accepted: 07/17/2023] [Indexed: 07/21/2023]
Abstract
Various therapeutic strategies, including chemotherapy, radiotherapy, photothermal therapy (PTT), and immunotherapy have been applied in cancer therapy. However, intrinsic or acquired therapeutic resistance is the main obstacle that attenuates the treatment effect of the therapeutic reagents used in these strategies. Studies have shown that autophagy and immunosuppressive tumor-associated macrophages (TAMs), as internal and external resistance mechanisms, would significantly compromise the effectiveness of cancer treatment. Therefore, selectively blocking the autophagy and repolarizing TAMs to anti-tumor phenotype (M1) will be effective for cancer treatments. Herein, an ambidextrous strategy that simultaneously inhibited autophagy and reeducated TAMs to promote anti-tumor therapy meditated by the iron-based nanocarriers was reported. The released Fe (II) ion reacted with the released artemisinin (ART) to produce ROS for chemodynamic therapy (CDT). The chloroquine (CQ) was used to inhibit autophagy in cancer cells and reset TAMs from the M2 phenotype to the M1 phenotype, eliminating the resistance of cancer cells and realizing an augmented therapeutic effect. This work provides a promising way for augmenting therapeutic efficiency by simultaneously interfering with two critical therapeutic resistance mechanisms.
Collapse
Affiliation(s)
- Han Zhu
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, PR China
| | - Xin Gao
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, PR China
| | - Bei Wang
- College of Basic Medical Science, Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-autoimmune Diseases of Hebei Province, Hebei University, Baoding 071002, PR China
| | - Biao Niu
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, PR China
| | - Dandan Liu
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, PR China
| | - Jinchao Zhang
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, PR China
| | - Yi Jin
- College of Basic Medical Science, Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-autoimmune Diseases of Hebei Province, Hebei University, Baoding 071002, PR China.
| | - Xinjian Yang
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, PR China.
| |
Collapse
|
24
|
Exogenous H 2S Attenuates Hypertension by Regulating Renin Exocytosis under Hyperglycaemic and Hyperlipidaemic Conditions. Int J Mol Sci 2023; 24:ijms24021690. [PMID: 36675205 PMCID: PMC9860892 DOI: 10.3390/ijms24021690] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Obesity, along with type 2 diabetes mellitus (T2DM), is a major contributor to hypertension. The renin-angiotensin-aldosterone system is involved in the occurrence of diabetes and hypertension. However, the mechanism by which obesity is related to T2DM induced hypertension is unclear. In this study, we observed that blood pressure and serum renin content were increased in patients with diabetes and hypertension. Hydrogen sulfide (H2S), as an endogenous bioactive molecule, has been shown to be a vasodilator. Db/db mice, characterized by obesity and T2DM, and juxtaglomerular (JG) cells, which line the afferent arterioles at the entrance of the glomeruli to produce renin, treated with glucose, palmitic acid (PA) and oleic acid (OA), were used as animal and cellular models. NaHS, the H2S donor, was administered to db/db mice through intraperitoneal injection. NaHS significantly alleviated blood pressure in db/db mice, decreased the renin content in the serum of db/db mice and reduced renin secretion from JG cells. NaHS modulated renin release via cAMP and soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs), including synaptosome-associated protein 23 (SNAP23) and vesicle-associated membrane protein 2 (VAMP2), which mediate renin exocytosis. Furthermore, NaHS increased the levels of autophagy-related proteins and colocalization with EGFP-LC3 puncta with renin-containing granules and VAMP2 to consume excessive renin to maintain intracellular homeostasis. Therefore, exogenous H2S attenuates renin release and promotes renin-vesicular autophagy to relieve diabetes-induced hypertension.
Collapse
|
25
|
Wan X, Xiang J, Fan H, Jiang Y, Lu Y, Zhang C, Zhang Y, Chen Q, Lei Y. Ciclopirox Olamine Induces Proliferation Inhibition and Protective Autophagy in Hepatocellular Carcinoma. Pharmaceuticals (Basel) 2023; 16:ph16010113. [PMID: 36678610 PMCID: PMC9863056 DOI: 10.3390/ph16010113] [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: 12/13/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Hepatocellular carcinoma is one of the most common fatal malignancies worldwide. Thus far, the hepatocellular carcinoma prognosis has been bleak due to deficiencies in the identification and diagnosis of early hepatocellular carcinoma. Ciclopirox olamine (CPX) is a synthetic antifungal agent and has been considered as an anti-cancer candidate drug recently, though the detailed mechanisms related to its anti-cancer effect in hepatocellular carcinoma have not yet been revealed. Here, we found that CPX could inhibit proliferation in HCC cells but not in intrahepatic cholangiocarcinoma cells by arresting the cell cycle. Moreover, the anti-cancer effects of CPX in HCC cells were also attributed to CPX-triggered ROS accumulation and DJ-1 downregulation. Additionally, CPX could promote complete autophagic flux, which alleviated the anti-cancer effect of CPX in HCC cells, whereas the ROS scavenger (NAC) would attenuate CPX-induced protective autophagy. Interestingly, CPX could also induce glycogen clustering in HCC cells. Altogether, this study provides a new insight into the detailed molecular mechanisms of CPX as an anti-cancer therapy and a strategy for treating hepatocellular carcinoma.
Collapse
|
26
|
Ciclopirox drives growth arrest and autophagic cell death through STAT3 in gastric cancer cells. Cell Death Dis 2022; 13:1007. [PMID: 36443287 PMCID: PMC9705325 DOI: 10.1038/s41419-022-05456-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022]
Abstract
Ciclopirox (CPX), an antifungal drug, has recently been identified as a promising agent for cancer treatment. However, the effects and underlying mechanism of CPX as an antitumor agent of gastric cancer (GC) remain largely unknown. Here, we found that CPX dramatically suppresses GC xenograft growth in vitro via inhibiting proliferation and stimulating autophagic cell death rather than apoptosis. Moreover, CPX (20 mg/kg, intraperitoneally) substantially inhibits GC xenograft tumor growth in vivo. Mechanistically, CPX promotes growth arrest and autophagic cell death through suppressing the phosphorylation of signal transducers and activators of transcription 3 (STAT3) at tyrosine 705 (Tyr705) and serine 727 (Ser727) sites, respectively. Additionally, CPX induces STAT3 ubiquitination, which subsequently leads to a decrease in the p-STAT3 (Ser727) level. On the other hand, CPX represses the p-STAT3 (Tyr705) level via p-Src (Tyr416) inhibition. Collectively, our findings unmask a novel mechanism by which CPX regulates growth and autophagic cell death in GC cells via regulating the phosphorylation of STAT3 both at Tyr705 and Ser727 residues, and suggest that CPX may be a potential treatment for GC.
Collapse
|
27
|
Glycogen overload transforms the liver. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1939-1941. [PMID: 36514217 PMCID: PMC10157623 DOI: 10.3724/abbs.2022172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
28
|
Ye L, Ma RH, Zhang XX, Thakur K, Zhang JG, Khan MR, Busquets R, Wei ZJ. Isorhamnetin Induces Apoptosis and Suppresses Metastasis of Human Endometrial Carcinoma Ishikawa Cells via Endoplasmic Reticulum Stress Promotion and Matrix Metalloproteinase-2/9 Inhibition In Vitro and In Vivo. Foods 2022; 11:3415. [PMID: 36360027 PMCID: PMC9654916 DOI: 10.3390/foods11213415] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 09/08/2024] Open
Abstract
Endometrial cancer (EC) is a very common female cancer which has attracted more and more attention. According to the individual patient's condition, the current treatment of EC patients is mainly based on surgery, which is supplemented by chemotherapy, radiotherapy, and endocrine intervention. However, these existing treatment strategies also have some inevitable limitations. Therefore, it is particularly important to find an active ingredient with low toxicity and a high safety profile against EC. Isorhamnetin is a flavonoid known to be present in a variety of plants, such as sea buckthorn, dry willow, and wolfberry. In recent years, the anti-tumor effects of isorhamnetin have been reported. In our study, isorhamnetin was shown to induce apoptosis in Ishikawa cells by inducing the endogenous mitochondrial apoptotic pathway and exogenous death receptor pathway, promoting the endoplasmic reticulum stress-related pathway, and activating the corresponding markers of UPR response. In addition, isorhamnetin affected the expression of MMP2 and MMP9-related proteins in vitro and in vivo and eventually repressed metastasis. Therefore, isorhamnetin can be used as a promising medicinal material for the treatment of EC.
Collapse
Affiliation(s)
- Lei Ye
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Run-Hui Ma
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Xiu-Xiu Zhang
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Kiran Thakur
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China
- School of Biological Science and Engineering, North Minzu University, Yinchuan 750021, China
| | - Jian-Guo Zhang
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China
- School of Biological Science and Engineering, North Minzu University, Yinchuan 750021, China
| | - Mohammad Rizwan Khan
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Rosa Busquets
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, London KT1 2EE, UK
| | - Zhao-Jun Wei
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China
- School of Biological Science and Engineering, North Minzu University, Yinchuan 750021, China
| |
Collapse
|