1
|
Hu X, Chang H, Guo Y, Yu L, Li J, Zhang B, Zhao H, Xu J, Pan G, Zhang K, Lü M, Cui H. Mori Folium ethanol extracts induce ferroptosis and suppress gastric cancer progression by inhibiting the AKT/GSK3β/NRF2 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156789. [PMID: 40344847 DOI: 10.1016/j.phymed.2025.156789] [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: 12/31/2024] [Revised: 04/09/2025] [Accepted: 04/19/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Mori Folium, the leaf of Morus alba L., is a traditional Chinese medicine (TCM) known for its diverse pharmacological activities, including anti-inflammatory and immunomodulatory effects. While the Morus alba itself has been reported to contain various bioactive compounds with anticancer properties, the anticancer activity of Mori Folium and its underlying mechanisms remain insufficiently understood. PURPOSE This study aimed to investigate the effects of Mori Folium ethanol extracts (MFEE) on gastric cancer (GC) and to elucidate its underlying mechanisms. METHODS To investigate the anti-GC properties of MFEE, CCK-8, colony formation, EdU, flow cytometry, and soft agar, scratch, and transwell assays were employed. Western blot was employed to analyze the expression of ferroptotic proteins, while ferroptotic cellular events were also assessed, including iron accumulation, GSH levels, reactive oxygen species (ROS) production, mitochondrial changes, and lipid peroxidation. The chemical profile of MFEE was characterized using a UPLC-ESI-MS/MS system. Additionally, network pharmacology analysis was performed to investigate the potential anti-GC mechanisms of MFEE. Finally, the in vivo anti-cancer effects of MFEE were evaluated using a subcutaneous mouse model, with hematoxylin and eosin (H&E) and immunohistochemistry (IHC) staining to assess histopathological and molecular changes. RESULTS This study demonstrated that MFEE suppresses GC cell proliferation, blocks the G1-S cell cycle transition, and inhibits migration and invasion by promoting Fe²⁺ accumulation, increasing MDA levels and ROS, depleting GSH, and downregulating the expression of xCT and GPX4, thereby inducing ferroptosis. Chemical analysis identified 1596 phytochemicals, including 35 bioactive compounds. The induction of ferroptosis by MFEE was associated with the inhibition of the PI3K/AKT signaling pathway, modulating the AKT/GSK3β/NRF2 axis. Activation of AKT by SC79 was found to mitigate MF-induced ferroptosis. Notably, MFEE enhanced the chemosensitivity of GC cells to cisplatin, potentially through ferroptosis induction. CONCLUSION This study revealed that MFEE induces ferroptosis in GC cells by modulating the PI3K/AKT signaling pathway, enhancing chemosensitivity to cisplatin, and providing a potential therapeutic strategy for GC.
Collapse
Affiliation(s)
- Xin Hu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Hongbo Chang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Yan Guo
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Lang Yu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Jing Li
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Bili Zhang
- School of Life Science, Chongqing University, Chongqing 400044, China
| | - Hui Zhao
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jingyang Xu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Guangzhao Pan
- Center for Innovative Drug Research, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China.
| | - Kui Zhang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Muhan Lü
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
2
|
Tang L, Zhang S, Zhai Y, Wu K, Wang P, Liu Y, Zhang C, Yin H, Tian Y, Zhao B, Lu H. Endoplasmic reticulum stress regulates swainsonine-induced the autophagy in renal tubular epithelial cells through UPR signaling pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 981:179616. [PMID: 40354703 DOI: 10.1016/j.scitotenv.2025.179616] [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: 01/12/2025] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Swainsonine (SW), the primary toxic component of locoweed, induces toxic response in grazing livestock. The swainsonine induced toxicity is characterized by symptoms including head tremors, ataxia, and limb paralysis. The mechanisms of the toxicity remained to be investigated. The unfolded protein response (UPR) plays a key role in alleviating endoplasmic reticulum stress (ERS) by reducing protein synthesis and promoting the degradation of misfolded proteins. ERS is closely associated with both the UPR and autophagy activation. However, the involvement of the UPR signaling pathway in SW-induced ERS and autophagy remains unclear. In this study, we demonstrate that SW up-regulates the expression of GRP78, XBP1s, LC3-II/I, and ATG5 in both in vitro and in vivo models, suggesting activation of ERS, UPR, and autophagy. To investigate the molecular mechanisms by which the UPR regulates autophagy under ERS in primary rat renal tubular epithelial cells (RTECs), we observed that inhibiting PERK led to increased levels of p62. Inhibition of ATF6 significantly reduced the up-regulation of LC3-II/I, p62, and ATG5. Furthermore, inhibiting IRE1α significantly decreased the expression of LC3-II/I and p62. These findings suggest that PERK and ATF6 regulate autophagy mainly by modulating the expression of autophagy-related genes, while IRE1α likely regulates these genes through the IRE1α-XBP1 pathway. Additionally, autophagy is directly regulated through the IRE1α-JNK signaling pathway.
Collapse
Affiliation(s)
- Lihui Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Shuhang Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yichao Zhai
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Kexin Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Pan Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yiling Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Congcheng Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Hai Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yanan Tian
- School of Medicine, Hainan Vocational University of Science and Technology, Haikou 570100, Hainan, China
| | - Baoyu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Hao Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
3
|
Wang M, Zhao JH, Tang MX, Li M, Zhao H, Li ZY, Liu AD. Cell Death Modalities in Therapy of Melanoma. Int J Mol Sci 2025; 26:3475. [PMID: 40331942 PMCID: PMC12026598 DOI: 10.3390/ijms26083475] [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: 03/03/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Melanoma, one of the most lethal cancers, demands urgent and effective treatment strategies. However, a successful therapeutic approach requires a precise understanding of the mechanisms underlying melanoma initiation and progression. This review provides an overview of melanoma pathogenesis, identifies current pathogenic factors contributing to mortality, and explores targeted therapy and checkpoint inhibitor therapy. Furthermore, we examine melanoma classification and corresponding therapies, along with advancements in various cell death mechanisms for melanoma treatment. We also discuss the current treatment status along with some drawbacks encountered during research stages such as resistance and metastasis.
Collapse
Affiliation(s)
- Meng Wang
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.W.); (J.-H.Z.); (M.-X.T.); (M.L.); (H.Z.)
| | - Jia-Hui Zhao
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.W.); (J.-H.Z.); (M.-X.T.); (M.L.); (H.Z.)
| | - Ming-Xuan Tang
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.W.); (J.-H.Z.); (M.-X.T.); (M.L.); (H.Z.)
| | - Meng Li
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.W.); (J.-H.Z.); (M.-X.T.); (M.L.); (H.Z.)
| | - Hu Zhao
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.W.); (J.-H.Z.); (M.-X.T.); (M.L.); (H.Z.)
- National Demonstration Center for Experimental Basic Medical Education, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhong-Yu Li
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.W.); (J.-H.Z.); (M.-X.T.); (M.L.); (H.Z.)
- National Demonstration Center for Experimental Basic Medical Education, Huazhong University of Science and Technology, Wuhan 430030, China
| | - An-Dong Liu
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.W.); (J.-H.Z.); (M.-X.T.); (M.L.); (H.Z.)
- National Demonstration Center for Experimental Basic Medical Education, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
4
|
Li Y, Dong M, Qin H, An G, Cen L, Deng L, Cui H. Mulberrin suppresses gastric cancer progression and enhances chemosensitivity to oxaliplatin through HSP90AA1/PI3K/AKT axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156441. [PMID: 39938178 DOI: 10.1016/j.phymed.2025.156441] [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: 10/28/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Mulberrin, a natural flavonoid featured with two isopentenyl groups, is derived from the mulberry family. Although previous researches have uncovered various properties of mulberrin, including the antioxidant, hypoglycemic, antibacterial, food-preserving, skin-whitening and life-extending effects in nematode, its potential therapeutic application and the underlying mechanism in gastric cancer (GC) remains largely unexplored. PURPOSE This research intends to examine the anti-tumoral effects of mulberrin and reveal the molecular mechanism through which mulberrin inhibits the GC progression. METHODS To detect the proliferative of GC cells co-incubated with mulberrin, MTT, EdU, and colony formation assays were conducted. The migration and invasion capabilities of GC cells co-incubated with mulberrin were assessed using wound healing and Transwell assay experiments. Flow cytometry and western blot were utilized to depict changes in the cell cycle and protein expression of associated cyclins. The interaction between mulberrin and HSP90AA1 was accomplished through auto-dock molecular docking, CETSA and DARTS assays. The subcutaneous tumor model was constructed with NOD/SCID mice to detect the tumorigenic potential of GC cells. The drug synergetic effect of mulberrin and oxaliplatin was analyzed using the Jin's formula. RESULTS Mulberrin effectively inhibited GC cell proliferation by inducing cell cycle arrest at G0/G1 phase. In addition, mulberrin significantly repressed the migration and invasion capacities of GC cells via reducing HSP90AA1 expression, leading to inhibition of the PI3K/AKT pathway and EMT process, hence curtailing proliferation, migration and invasion capacities of GC cells. Furthermore, mulberrin diminished the tumorigenic potential of GC cells, an effect mitigated by HSP90AA1 restoration. Notably, combining mulberrin with oxaliplatin yielded a synergistic inhibitory effect on GC cells, surpassing the efficacy of either agent used alone. CONCLUSION Mulberrin suppresses proliferation, migration and invasion of GC cells by directly targeting and reducing HSP90AA1 to inhibit the PI3K/AKT pathway and EMT process. Additionally, mulberrin increases the sensitivity of GC cells to oxaliplatin. Together, these findings uncover a latent use of mulberrin as a promising therapeutic drug that may be included in anti-GC strategies for the treatment of GC patients.
Collapse
Affiliation(s)
- Yongsen Li
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China
| | - Mengyao Dong
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China
| | - Hanghang Qin
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Guozhen An
- Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China
| | - Liang Cen
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China
| | - Longfei Deng
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China.
| |
Collapse
|
5
|
Xing Y, Lv X, Chen X, Du J, Hu D, He R, Liang X, Yang Y. Maackiain induces apoptosis and autophagy via ROS-mediated endoplasmic reticulum stress in endometrial cancer. Int Immunopharmacol 2025; 147:113935. [PMID: 39756166 DOI: 10.1016/j.intimp.2024.113935] [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: 09/13/2024] [Revised: 12/09/2024] [Accepted: 12/21/2024] [Indexed: 01/07/2025]
Abstract
Endometrial cancer (EC) is a common gynecological cancer, characterized by increasing incidence and mortality rates. Maackiain (MA), a natural flavonoid compound, has multiple biological activities, but little is known about how it affects EC cells. In the present study, CCK-8, EdU, colony formation, and flow cytometry assays were used to evaluate the effects of MA on EC cell proliferation, apoptosis, and reactive oxygen species (ROS) levels. The effect of MA on autophagy in EC cells were examined through the observation of cell morphology and ultrastructure, and cells were transfected with AdPlus-mCherry-GFP-LC3B for further analysis. Transcriptomic and western blot analyses revealed the underlying mechanism. To evaluate the anti-EC effect of MA in vivo, a xenograft model was established. The results demonstrated that MA inhibited KLE and Ishikawa cell growth in a dose-dependent manner. Furthermore, MA significantly suppressed EC xenograft tumor growth in vivo while exhibiting low toxicity. In addition, EC cells treated with MA exhibited pro-apoptotic and pro-autophagic responses, with the latter exhibiting cytoprotective properties. MA also induced the accumulation of ROS, which promoted endoplasmic reticulum (ER) stress. Notably, the use of the N-acetyl-L-cysteine (NAC) ROS scavenger and the 4-phenylbutyric acid (4-PBA) ER stress inhibitor effectively mitigated the autophagy and apoptosis induced by MA. These results collectively implied that MA triggers autophagy and apoptosis in EC cells through ROS-mediated ER stress, highlighting its potential as a therapeutic agent against EC.
Collapse
Affiliation(s)
- Yijuan Xing
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, 730000 Gansu, China
| | - Xiao Lv
- Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, 730000 Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Lanzhou, 730000 Gansu, China
| | - Xi Chen
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, 730000 Gansu, China
| | - Junhong Du
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, 730000 Gansu, China
| | - Dan Hu
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, 730000 Gansu, China
| | - Ruifen He
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, 730000 Gansu, China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, 730000 Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Lanzhou, 730000 Gansu, China.
| | - Yongxiu Yang
- Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, 730000 Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Lanzhou, 730000 Gansu, China.
| |
Collapse
|
6
|
Song W, Zhang Q, Cao Z, Jing G, Zhan T, Yuan Y, Kang N, Zhang Q. Targeting SERCA2 in Anti-Tumor Drug Discovery. Curr Drug Targets 2025; 26:1-16. [PMID: 39323343 DOI: 10.2174/0113894501325497240918042654] [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: 04/23/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
SERCA2, a P-type ATPase located on the endoplasmic reticulum of cells, plays an important role in maintaining calcium balance within cells by transporting calcium from the cytoplasm to the endoplasmic reticulum against its concentration gradient. A multitude of studies have demonstrated that the expression of SERCA2 is abnormal in a wide variety of tumor cells. Consequently, research exploring compounds that target SERCA2 may offer a promising avenue for the development of novel anti-tumor drugs. This review has summarized the anti-tumor compounds targeting SERCA2, including thapsigargin, dihydroartemisinin, curcumin, galangin, etc. These compounds interact with SERCA2 on the endoplasmic reticulum membrane, disrupting intracellular calcium ion homeostasis, leading to tumor cell apoptosis, autophagy and cell cycle arrest, ultimately producing anti-tumor effects. Additionally, several potential research directions for compounds targeting SERCA2 as clinical anti-cancer drugs have been proposed in the review. In summary, SERCA2 is a promising anti-tumor target for drug discovery and development.
Collapse
Affiliation(s)
- Wanqian Song
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qiuju Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhiyong Cao
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Guo Jing
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tiancheng Zhan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yongkang Yuan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ning Kang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qiang Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
7
|
Li X, Xu J, Yan L, Tang S, Zhang Y, Shi M, Liu P. Targeting Disulfidptosis with Potentially Bioactive Natural Products in Metabolic Cancer Therapy. Metabolites 2024; 14:604. [PMID: 39590840 PMCID: PMC11596291 DOI: 10.3390/metabo14110604] [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: 08/31/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Metabolic cancers are defined by metabolic reprogramming. Although this reprograming drives rapid tumour growth and invasion, it also reveals specific metabolic vulnerabilities that can be therapeutically exploited in cancer therapy. A novel form of programmed cell death, known as disulfidptosis, was identified last year; tumour cells with high SLC7A11 expression undergo disulfidptosis when deprived of glucose. Natural products have attracted increasing attention and have shown potential to treat metabolic cancers through diverse mechanisms. METHODS We systematically searched electronic databases involving PubMed, Web of Science, Gooale Scholar. To ensue comprehensive exploration, keywords including metabolic reprogramming, metabolic cancer, disulfidptosis, natural products and some other words were employed. RESULTS In this review, we focus on the shared characteristics and metabolic vulnerabilities of metabolic cancers. Additionally, we discuss the molecular mechanisms underlying disulfidptosis and highlight key regulatory genes. Furthermore, we predict bioactive natural products that target disulfidptosis-related genes, offering new perspectives for anticancer strategies through the modulation of disulfidptosis. CONCLUSIONS By summarizing current research progress, this review mainly analyzed the potential mechanisms of natural products in the treatment of metabolic cancer.
Collapse
Affiliation(s)
- Xinyan Li
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China;
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
| | - Jiayi Xu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
| | - Liangwen Yan
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
| | - Shenkang Tang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
- Department of Oncology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - Yinggang Zhang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
| | - Mengjiao Shi
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China;
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Pengfei Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education of China, Xi’an 710061, China
| |
Collapse
|
8
|
Trocchia M, Ventrici A, Modestino L, Cristinziano L, Ferrara AL, Palestra F, Loffredo S, Capone M, Madonna G, Romanelli M, Ascierto PA, Galdiero MR. Innate Immune Cells in Melanoma: Implications for Immunotherapy. Int J Mol Sci 2024; 25:8523. [PMID: 39126091 PMCID: PMC11313504 DOI: 10.3390/ijms25158523] [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: 07/12/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
The innate immune system, composed of neutrophils, basophils, eosinophils, myeloid-derived suppressor cells (MDSCs), macrophages, dendritic cells (DCs), mast cells (MCs), and innate lymphoid cells (ILCs), is the first line of defense. Growing evidence demonstrates the crucial role of innate immunity in tumor initiation and progression. Several studies support the idea that innate immunity, through the release of pro- and/or anti-inflammatory cytokines and tumor growth factors, plays a significant role in the pathogenesis, progression, and prognosis of cutaneous malignant melanoma (MM). Cutaneous melanoma is the most common skin cancer, with an incidence that rapidly increased in recent decades. Melanoma is a highly immunogenic tumor, due to its high mutational burden. The metastatic form retains a high mortality. The advent of immunotherapy revolutionized the therapeutic approach to this tumor and significantly ameliorated the patients' clinical outcome. In this review, we will recapitulate the multiple roles of innate immune cells in melanoma and the related implications for immunotherapy.
Collapse
Affiliation(s)
- Marialuisa Trocchia
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, 80138 Naples, Italy; (M.T.); (A.V.); (A.L.F.); (F.P.); (S.L.)
| | - Annagioia Ventrici
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, 80138 Naples, Italy; (M.T.); (A.V.); (A.L.F.); (F.P.); (S.L.)
| | - Luca Modestino
- Department of Internal Medicine and Clinical Immunology, University Hospital of Naples Federico II, 80138 Naples, Italy;
| | - Leonardo Cristinziano
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80138 Naples, Italy;
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, 80138 Naples, Italy; (M.T.); (A.V.); (A.L.F.); (F.P.); (S.L.)
| | - Francesco Palestra
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, 80138 Naples, Italy; (M.T.); (A.V.); (A.L.F.); (F.P.); (S.L.)
| | - Stefania Loffredo
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, 80138 Naples, Italy; (M.T.); (A.V.); (A.L.F.); (F.P.); (S.L.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80138 Naples, Italy;
| | - Mariaelena Capone
- Melanoma, Cancer Immunotherapy, and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, 80138 Naples, Italy; (M.C.); (G.M.); (M.R.); (P.A.A.)
| | - Gabriele Madonna
- Melanoma, Cancer Immunotherapy, and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, 80138 Naples, Italy; (M.C.); (G.M.); (M.R.); (P.A.A.)
| | - Marilena Romanelli
- Melanoma, Cancer Immunotherapy, and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, 80138 Naples, Italy; (M.C.); (G.M.); (M.R.); (P.A.A.)
| | - Paolo Antonio Ascierto
- Melanoma, Cancer Immunotherapy, and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, 80138 Naples, Italy; (M.C.); (G.M.); (M.R.); (P.A.A.)
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, 80138 Naples, Italy; (M.T.); (A.V.); (A.L.F.); (F.P.); (S.L.)
- Department of Internal Medicine and Clinical Immunology, University Hospital of Naples Federico II, 80138 Naples, Italy;
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80138 Naples, Italy;
| |
Collapse
|
9
|
Zhong X, Ke X, Yang H, Ye X, Li C, Pan J, Ran W, Wang F, Cui H. Moracin D suppresses cell growth and induces apoptosis via targeting the XIAP/PARP1 axis in pancreatic cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155527. [PMID: 38489888 DOI: 10.1016/j.phymed.2024.155527] [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: 12/01/2023] [Revised: 02/02/2024] [Accepted: 03/08/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Pancreatic cancer, a tumor with a high metastasis rate and poor prognosis, is among the deadliest human malignancies. Investigating effective drugs for their treatment is imperative. Moracin D, a natural benzofuran compound isolated from Morus alba L., shows anti-inflammation and anti-breast cancer properties and is effective against Alzheimer's disease. However, the effect and mechanism of Moracin D action in pancreatic cancer remain obscure. PURPOSE To investigate the function and molecular mechanism of Moracin D action in repressing the malignant progression of pancreatic cancer. METHODS Pancreatic cancer cells were treated with Moracin D, and cell proliferation was evaluated by cell counting kit-8 (CCK-8) and immunofluorescence assays. The clonogenicity of pancreatic cancer cells was assessed based on plate colony formation and soft agar assay. Flow cytometry was used to detect cell apoptosis. The expression of proteins related to the apoptosis pathway was determined by Western blot analysis. Moracin D and XIAP were subjected to docking by auto-dock molecular docking analysis. Ubiquitination levels of XIAP and the interaction of XIAP and PARP1 were assessed by co-immunoprecipitation analysis. Moracin D's effects on tumorigenicity were assessed by a tumor xenograft assay. RESULTS Moracin D inhibited cell proliferation, induced cell apoptosis, and regulated the protein expression of molecules involved in caspase-dependent apoptosis pathways. Moracin D suppressed clonogenicity and tumorigenesis of pancreatic cancer cells. Mechanistically, XIAP could interact with PARP1 and stabilize PARP1 by controlling its ubiquitination levels. Moracin D diminished the stability of XIAP and decreased the expression of XIAP by promoting proteasome-dependent XIAP degradation, further blocking the XIAP/PARP1 axis and repressing the progression of pancreatic cancer. Moracin D could dramatically improve the chemosensitivity of gemcitabine in pancreatic cancer cells. CONCLUSION Moracin D repressed cell growth and tumorigenesis, induced cell apoptosis, and enhanced the chemosensitivity of gemcitabine through the XIAP/PARP1 axis in pancreatic cancer. Moracin D is a potential therapeutic agent or adjuvant for pancreatic cancer.
Collapse
Affiliation(s)
- Xi Zhong
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China
| | - Xiaoxue Ke
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China
| | - He Yang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China
| | - Xiang Ye
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China
| | - Can Li
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China
| | - Jun Pan
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China
| | - Wenhao Ran
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China
| | - Feng Wang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, China.
| |
Collapse
|
10
|
Wen W, Ertas YN, Erdem A, Zhang Y. Dysregulation of autophagy in gastric carcinoma: Pathways to tumor progression and resistance to therapy. Cancer Lett 2024; 591:216857. [PMID: 38583648 DOI: 10.1016/j.canlet.2024.216857] [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: 02/06/2024] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
The considerable death rates and lack of symptoms in early stages of gastric cancer (GC) make it a major health problem worldwide. One of the most prominent risk factors is infection with Helicobacter pylori. Many biological processes, including those linked with cell death, are disrupted in GC. The cellular "self-digestion" mechanism necessary for regular balance maintenance, autophagy, is at the center of this disturbance. Misregulation of autophagy, however, plays a role in the development of GC. In this review, we will examine how autophagy interacts with other cell death processes, such as apoptosis and ferroptosis, and how it affects the progression of GC. In addition to wonderful its role in the epithelial-mesenchymal transition, it is engaged in GC metastasis. The role of autophagy in GC in promoting drug resistance stands out. There is growing interest in modulating autophagy for GC treatment, with research focusing on natural compounds, small-molecule inhibitors, and nanoparticles. These approaches could lead to breakthroughs in GC therapy, offering new hope in the fight against this challenging disease.
Collapse
Affiliation(s)
- Wen Wen
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey.
| | - Ahmet Erdem
- Institute for Quantitative Health Science and Engineering (IQ), Department of Biomedical Engineering, College of Engineering and Human Medicine, Michigan State University, East Lansing, MI, 48824, USA; Department of Biomedical Engineering, Kocaeli University, Umuttepe Campus, Kocaeli, 41001 Turkey.
| | - Yao Zhang
- Department of Gynaecology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
11
|
Hu W, Zhao J, Hu Y, Song S, Chen X, Sun Y. Huangqi Jiuni decoction prevents acute kidney injury induced by severe burns by inhibiting activation of the TNF/NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117344. [PMID: 37949330 DOI: 10.1016/j.jep.2023.117344] [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: 08/10/2023] [Revised: 10/15/2023] [Accepted: 10/22/2023] [Indexed: 11/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangqi Jiuni decoction (HQJND) is a prescription for the treatment of severe burns provided based on traditional Chinese and Western medicine, which is created by the First Affiliated Hospital of Anhui Medical University. It consists of 12 herbs and has been used clinically for decades. It has greatly shortened the course of the disease, but the mechanism by which HQJND treats the disease still remains unclear. AIM OF THE STUDY Hence, the objective of this investigation was to utilize modern pharmacological tools to demonstrate the efficacy and mechanism of HQJND in the treatment of acute kidney injury (AKI) caused by severe burns. MATERIALS AND METHODS In this study, the chemical constituents in HQJND were first examined using liquid chromatography tandem mass spectrometry (LC-MS/MS). Then, by using network pharmacology, we screened the targets of drug and disease action, and predicted the signaling pathways acting in the course of drug treatment of disease. Finally, we attempted to verify the efficacy of the drug and explored its therapeutic mechanism after the establishment of an animal model, herbal gavage treatment, collection of rat kidneys and serum for renal function, quantitative real-time Polymerase Chain Reaction (RT-qPCR), Western Blotting (WB), Hematoxylin and eosin (HE) staining and Immunohistochemistry (IHC). RESULTS The 14 important active ingredients in HQJND was analyzed by liquid chromatography tandem mass spectrometry, while network pharmacology screening was performed to identify 353 disease-associated marker genes and 286 drug targets, finally identifying the TNF/NF-κB (tumor necrosis factor/nuclear factor kappa-B) signaling site: the key pathway of burn-induced acute kidney injury when HQJND intervened. The serum renal function and histopathology of rats demonstrated that the use of HQJND significantly improved the renal function in severe burns. RT-qPCR and WB confirmed that the TNF/NF-κB signaling pathway was activated in the Model group of rats, and HQJND could curb the signaling pathway because it moderated the expressions of key proteins in the process. CONCLUSION Based on modern pharmacology, we explored an effective herbal preparation to ameliorate the impairment of renal function after severe burns, which is most likely to function through the TNF/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Wanxuan Hu
- Department of Burn, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, PR China
| | - Jie Zhao
- Department of Chinese Medicine, The First Affiliated Hospital of Anhui Medical University, No.218, Jixi Road, Shushan District, Hefei, Anhui, 230032, PR China; Department of Chinese Integrative Medicine, Anhui Medical University, No. 80, Meishan Road, Shushan District, Hefei, Anhui, 230032, PR China
| | - Yuxin Hu
- Department of Chinese Integrative Medicine, Anhui Medical University, No. 80, Meishan Road, Shushan District, Hefei, Anhui, 230032, PR China
| | - Shuai Song
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China
| | - Xulin Chen
- Department of Burn, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, PR China
| | - Yexiang Sun
- Department of Burn, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, PR China.
| |
Collapse
|
12
|
Zhang K, Hu X, Su J, Li D, Thakur A, Gujar V, Cui H. Gastrointestinal Cancer Therapeutics via Triggering Unfolded Protein Response and Endoplasmic Reticulum Stress by 2-Arylbenzofuran. Int J Mol Sci 2024; 25:999. [PMID: 38256073 PMCID: PMC10816499 DOI: 10.3390/ijms25020999] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Gastrointestinal cancers are a major global health challenge, with high mortality rates. This study investigated the anti-cancer activities of 30 monomers extracted from Morus alba L. (mulberry) against gastrointestinal cancers. Toxicological assessments revealed that most of the compounds, particularly immunotoxicity, exhibit some level of toxicity, but it is generally not life-threatening under normal conditions. Among these components, Sanggenol L, Sanggenon C, Kuwanon H, 3'-Geranyl-3-prenyl-5,7,2',4'-tetrahydroxyflavone, Morusinol, Mulberrin, Moracin P, Kuwanon E, and Kuwanon A demonstrate significant anti-cancer properties against various gastrointestinal cancers, including colon, pancreatic, and gastric cancers. The anti-cancer mechanism of these chemical components was explored in gastric cancer cells, revealing that they inhibit cell cycle and DNA replication-related gene expression, leading to the effective suppression of tumor cell growth. Additionally, they induced unfolded protein response (UPR) and endoplasmic reticulum (ER) stress, potentially resulting in DNA damage, autophagy, and cell death. Moracin P, an active monomer characterized as a 2-arylbenzofuran, was found to induce ER stress and promote apoptosis in gastric cancer cells, confirming its potential to inhibit tumor cell growth in vitro and in vivo. These findings highlight the therapeutic potential of Morus alba L. monomers in gastrointestinal cancers, especially focusing on Moracin P as a potent inducer of ER stress and apoptosis.
Collapse
Affiliation(s)
- Kui Zhang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Xin Hu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Jingjing Su
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Dong Li
- State Key Laboratory of Resource Insects, Institute of Sericulture and Systems Biology, Southwest University, Chongqing 400715, China
| | - Abhimanyu Thakur
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vikramsingh Gujar
- Department of Anatomy and Cell Biology, Okhlahoma State University Center for Health Sciences, Tulsa, OK 74107, USA
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| |
Collapse
|
13
|
Zhang K, Man X, Hu X, Tan P, Su J, Abbas MN, Cui H. GATA binding protein 6 regulates apoptosis in silkworms through interaction with poly (ADP-ribose) polymerase. Int J Biol Macromol 2024; 256:128515. [PMID: 38040165 DOI: 10.1016/j.ijbiomac.2023.128515] [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: 10/08/2023] [Revised: 11/26/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
The GATA family of genes plays various roles in crucial biological processes, such as development, cell differentiation, and disease progression. However, the roles of GATA in insects have not been thoroughly explored. In this study, a genome-wide characterization of the GATA gene family in the silkworm, Bombyx mori, was conducted, revealing lineage-specific expression profiles. Notably, GATA6 is ubiquitously expressed across various developmental stages and tissues, with predominant expression in the midgut, ovaries, and Malpighian tubules. Overexpression of GATA6 inhibits cell growth and promotes apoptosis, whereas, in contrast, knockdown of PARP mitigates the apoptotic effects driven by GATA6 overexpression. Co-immunoprecipitation (co-IP) has demonstrated that GATA6 can interact with Poly (ADP-ribose) polymerase (PARP), suggesting that GATA6 may induce cell apoptosis by activating the enzyme's activity. These findings reveal a dynamic and regulatory relationship between GATA6 and PARP, suggesting a potential role for GATA6 as a key regulator in apoptosis through its interaction with PARP. This research deepens the understanding of the diverse roles of the GATA family in insects, shedding light on new avenues for studies in sericulture and pest management.
Collapse
Affiliation(s)
- Kui Zhang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China.
| | - Xu Man
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Xin Hu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Peng Tan
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Jingjing Su
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Muhammad Nadeem Abbas
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China.
| |
Collapse
|