1
|
Holoubek A, Strachotová D, Wolfová K, Otevřelova P, Belejová S, Röselová P, Benda A, Brodská B, Herman P. Correlation of p53 oligomeric status and its subcellular localization in the presence of the AML-associated NPM mutant. PLoS One 2025; 20:e0322096. [PMID: 40334261 PMCID: PMC12058200 DOI: 10.1371/journal.pone.0322096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/17/2025] [Indexed: 05/09/2025] Open
Abstract
Tumor suppressor p53 is a key player in the cell response to DNA damage that suffers by frequent inactivating aberrations. Some of them disturb p53 oligomerization and influence cell decision between proliferation, growth arrest and apoptosis. Active p53 resides mostly in the nucleus, degradation occurs in the cytoplasm. Acute myeloid leukemia (AML)-related mutation of NPM (NPMmut) induces massive mislocalization of p53 to the cytoplasm, which might be related to leukemia initiation. Since both proteins interact and execute their function as oligomers, we investigated the role of perturbed p53 oligomerization in the p53 mislocalization process in live cells by FLIM (fluorescence lifetime imaging microscopy), fluorescence anisotropy imaging (FAIM), fluorescence cross-correlation spectroscopy (FCCS) and immunochemical methods. On a set of fluorescently labeled p53 variants, monomeric R337G and L344P, dimeric L344A, and multimeric D352G and A353S, we correlated their cellular localization, oligomerization and interaction with NPMmut. Interplay between nuclear export signal (NES) and nuclear localization signal (NLS) of p53 was investigated as well. While NLS was found critical for the nuclear p53 localization, NES plays less significant role. We observed cytoplasmic translocation only for multimeric A353S variant with sufficient stability and strong interaction with NPMmut. Less stable multimer D352G and L344A dimer were not translocated, monomeric p53 variants always resided in the nucleus independently of the presence of NPMmut and NES intactness. Oligomeric state of NPMmut is not required for p53 translocation, which happens also in the presence of the nonoligomerizing NPMmut variant. The prominent structural and functional role of the R337 residue is shown.
Collapse
Affiliation(s)
- Aleš Holoubek
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Dita Strachotová
- Faculty of Mathematics and Physics, Institute of Physics, Charles University, Prague, Czech Republic
| | - Kateřina Wolfová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Petra Otevřelova
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Sára Belejová
- Faculty of Mathematics and Physics, Institute of Physics, Charles University, Prague, Czech Republic
| | - Pavla Röselová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Aleš Benda
- Imaging Methods Core Facility at BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Barbora Brodská
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Petr Herman
- Faculty of Mathematics and Physics, Institute of Physics, Charles University, Prague, Czech Republic
| |
Collapse
|
2
|
Dashti A, Hosseini HM, Mirhosseini SA. Epsilon toxin induces cytotoxicity by mediating autophagy and apoptosis via the PI3K/AKT/mTOR signaling pathway in A549 cells. Mol Biol Rep 2025; 52:403. [PMID: 40252125 DOI: 10.1007/s11033-025-10439-w] [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: 04/17/2024] [Accepted: 03/14/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Epsilon toxin, which is synthesized by Clostridium perfringens, is a type of pore-forming protein that is associated with the development of enterotoxemia in ruminants. As toxins are agents of bioterrorism, exposure to toxin aerosols causes endothelial cell damage and cytotoxicity in human lung cells. However, little information is available regarding the cytotoxicity and mechanisms associated with lung cancer cell lines. The aim of the present study was to explore the cytotoxic effects of epsilon toxin on the human lung cell line A549 and its involvement in the PI3K/AKT/mTOR signaling pathway to clarify the underlying molecular mechanism involved. METHODS AND RESULTS A549 cells were treated with epsilon toxin, and cytotoxicity was assessed via MTT and LDH assays. Flow cytometry evaluated ROS levels, cell cycle arrest, and apoptosis, while Hoechst 33,258 staining confirmed apoptotic morphology. qRT‒PCR and Western blotting measured apoptosis-, autophagy-, and PI3K/AKT/mTOR-related markers. Epsilon toxin reduced cell viability and increased membrane leakage in a concentration-dependent manner, accompanied by ROS overproduction. It upregulated autophagy markers (beclin-1, LC3 II/I, p62) and suppressed PI3K/AKT/mTOR signaling. Cell cycle arrest at the sub-G1 phase and apoptosis were induced via p53 activation, Bax/Bcl-2 imbalance, and caspase-3 cleavage, as confirmed by annexin V/PI and Hoechst 33,258 staining. CONCLUSIONS Epsilon toxin triggers cytotoxicity in A549 cells by activating apoptosis and autophagy through PI3K/AKT/mTOR pathway inhibition. These findings elucidate molecular mechanisms underlying epsilon toxin's action in lung cancer cells, highlighting its dual role in programmed cell death and potential therapeutic relevance.
Collapse
Affiliation(s)
- Ayat Dashti
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hamideh Mahmoodzadeh Hosseini
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mirhosseini
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Li A, Miao X, Han Z, Lin J, Huang J, Zheng X. Poly(glycerol succinate) hydrogel promotes spinal cord repair by regulating bio-energetic activity in severe injury. Mater Today Bio 2025; 31:101624. [PMID: 40124342 PMCID: PMC11929082 DOI: 10.1016/j.mtbio.2025.101624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/09/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Traumatic spinal cord injury (SCI) is a major clinical challenge, imposing a significant burden on both patients and healthcare systems. The complexity of SCI stems from its multifactorial pathogenesis, incorporating a variety of regulating factors. Despite the exploration of mechanisms of SCI pathophysiology and the development of biomedical therapies, current clinical interventions are still limited to surgical interventions and rehabilitative care. This study introduces an approach to protect mitochondria-a pivotal factor in SCI pathogenesis-through the use of poly(glycerol succinate)-based hydrogel. To regulate the process, the PEGylated poly(glycerol succinate) (PPGS), was designed and synthesized via a novel method, combined with recent findings that emphasize the roles of glycerol-based hydrogel in soft tissue regeneration. Building on these, an innovative, bioenergetically-active hydrogel, acrylated PEGylated poly(glycerol succinate) (APPGS), which improves mitochondrial function after injury, targeting SCI treatment, was developed. The evidence, supported by both in vivo and in vitro assays, affirms the therapeutic efficacy of the APPGS hydrogel in SCI contexts. The APPGS hydrogel represents a significant advancement with substantial potential for clinical application in SCI therapy, offering a new avenue for addressing the complex challenges of SCI management.
Collapse
Affiliation(s)
- Ang Li
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- National Center for Orthopaedics, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| | - Xin Miao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- National Center for Orthopaedics, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Department of Orthopaedics, The Third People's Hospital of Chengdu, Sichuan, China
| | - Zhengzhe Han
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- National Center for Orthopaedics, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| | - Junqing Lin
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- National Center for Orthopaedics, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| | - Jinghuan Huang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- National Center for Orthopaedics, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| | - Xianyou Zheng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- National Center for Orthopaedics, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
| |
Collapse
|
4
|
Jin SK, Baek KH. Unraveling the role of deubiquitinating enzymes on cisplatin resistance in several cancers. Biochim Biophys Acta Rev Cancer 2025; 1880:189297. [PMID: 40058507 DOI: 10.1016/j.bbcan.2025.189297] [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: 01/07/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
The use of platinum-based drugs in cancer treatment is one of the most common methods in chemotherapy. Especially, cisplatin induces cell death by interrupting DNA synthesis by binding to the DNA bases, thereby leading to the apoptosis via multiple pathways. However, the major hurdle in chemotherapy is drug resistance. To overcome drug resistance, the ubiquitin-proteasome system (UPS) has emerged as a potential therapeutic target. The UPS is a pivotal signaling pathway that regulates the majority of cellular proteins by attaching ubiquitin to substrates, leading to proteasomal degradation. Conversely, deubiquitinating enzymes (DUBs) remove tagged ubiquitin from the substrate and inhibit degradation, thereby maintaining proteostasis. Recently, studies have been conducted to identify the substrates of DUBs and investigated the cellular mechanisms, and now the development of therapeutics using DUB inhibitors is in clinical trials. However, the mechanism of the DUB response to cisplatin remains still unclear. In this review, we summarize the research reported on the function of DUBs responding to cisplatin.
Collapse
Affiliation(s)
- Sun-Kyu Jin
- Department of Biomedical Science, CHA University, Gyeonggi-Do 13488, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA University, Gyeonggi-Do 13488, Republic of Korea.
| |
Collapse
|
5
|
Yang Y, Du Y, Ma X, Yuan G, Li G, Zhang Q, Zhou S. Transcription factor addictions: exploring the potential Achilles' Heel of endometriosis. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2832-8. [PMID: 40163264 DOI: 10.1007/s11427-024-2832-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/15/2024] [Indexed: 04/02/2025]
Abstract
A considerable number of women of reproductive age suffer from endometriosis worldwide. There is a significant physical, mental, and financial burden on patients affected by this condition in terms of pelvic pain, either continuously or intermittently, dysmenorrhea, infertility, and a higher risk of certain types of cancer. Several treatments available in clinical settings for endometriosis management do not provide adequate efficacy and have undesirable side effects. Transcription factors (TFs) are crucial regulators of key biological processes involved in endometriosis. Here, we elaborated on the research progress regarding the crucial roles of TFs in endometriosis, emphasizing their implications for clinical outcomes and critical therapeutic contributions. By delving into their involvement in key processes, such as cell proliferation and apoptosis, we revealed the multifaceted role of key TFs in disease progression. We aimed to provide a systemic understanding of TFs regulation in endometriosis pathogenesis, establishing a foundation for innovative treatment approaches.
Collapse
Affiliation(s)
- Yang Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Yi Du
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Yuan
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Guobo Li
- Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Qian Zhang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China.
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China.
| |
Collapse
|
6
|
Zhao C. Exploring cell death pathways in oral cancer: mechanisms, therapeutic strategies, and future perspectives. Discov Oncol 2025; 16:395. [PMID: 40133563 PMCID: PMC11936869 DOI: 10.1007/s12672-025-02022-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/03/2025] [Indexed: 03/27/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) represents a significant global health challenge, characterized by aggressive progression and poor therapeutic response despite advances in treatment modalities. This review provides a comprehensive analysis of diverse cell death mechanisms in OSCC, encompassing traditional pathways (apoptosis, autophagy, and necrosis), newly characterized mechanisms (ferroptosis, pyroptosis, and necroptosis), and emerging pathways (cuproptosis, anoikis, parthanatos, and entosis). By examining the molecular basis of these pathways, particularly the crucial roles of p53 signaling and miRNA regulation, we highlight how their dysregulation contributes to treatment resistance and tumor progression. The review synthesizes recent evidence demonstrating the complex interplay between these ten distinct cell death mechanisms and their impact on the tumor microenvironment and immune response. We evaluate innovative therapeutic approaches that target these pathways, including novel small molecules, combination strategies, and immunomodulatory treatments that exploit specific cell death mechanisms to enhance therapeutic efficacy. Special attention is given to emerging personalized medicine strategies that consider individual tumor characteristics and cell death pathway profiles. By integrating current challenges with future research directions, this review provides a framework for developing more effective treatments that can leverage multiple cell death pathways to overcome therapy resistance and improve outcomes for oral cancer patients.
Collapse
Affiliation(s)
- Chenyi Zhao
- The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, No.1 Xincheng Blvd, Songshan Lake National High-tech Industrial Development Zone, 523808, Guangdong Province, China.
| |
Collapse
|
7
|
Xia C, Cai M, Lu Y, Wang B, Xu L, Wang K, Liu Z. Radioprotective Effects and Mechanisms of One-Year and Seven-Year White Tea Extracts Against 137Cs Radiation-Induced Cell Damage. Molecules 2025; 30:1448. [PMID: 40286032 PMCID: PMC11990172 DOI: 10.3390/molecules30071448] [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: 01/25/2025] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Ionizing radiation (IR) is widely present in the environment, with 137Cesium (Cs) radiation having particularly severe impacts during nuclear accidents. The objective of our study was to assess the radiation protection or repair effect of one year (WT-1Y) or seven years (WT-7Y) of storage on white teas, as well as to investigate the mechanism of radioprotection. HGC-27 cells exposed to 137Cs γ-rays (30 Gy) exhibited significant changes in cell structure, apoptosis, ROS, LDH, and their expression of p53 and Caspase-3. The results showed that WT-1Y and WT-7Y acted as antioxidants, showed reduced ROS and LDH levels, and had increased CAT and SOD activities as well as cell survival rate. The WT treatments significantly inhibited apoptosis in both the pre- and post-radiation groups, with WT-1 showing stronger effects in pretreatment by reducing LDH, p53, and Caspase-3 levels and enhancing ROS scavenging and enzyme activities. Post-treatment analysis revealed WT-7 had greater effects on cell viability and SOD activity. Overall, both WT-1 and WT-7 mitigated radiation damage, likely by inhibiting the p53/Caspase-3 apoptosis pathway. A Spearman analysis of the differential metabolites in WT-1Y and WT-7Y with cellular radioprotective indicators revealed that metabolites, such as EGC, procyanidin B4, and phenolic acids (abundant in WT-1Y), quercetin-3-glucosylrutinoside, and caffeine (enriched in WT-7Y) contributed to their distinct effects in the pre- and post-treatment of 137Cs γ-rays.
Collapse
Affiliation(s)
- Chen Xia
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Meisheng Cai
- Fuding Tea Industry Development Leading Group, Ningde 355200, China
| | - Yanting Lu
- Institute of Crops and Nuclear Technology Utilization, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Bingkui Wang
- Institute of Crops and Nuclear Technology Utilization, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Linglin Xu
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Kaixi Wang
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Zhonghua Liu
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
8
|
Hong CS, Wu NC, Lin YW, Lin YC, Shih JY, Niu KC, Lin MT, Chang CP, Chen ZC, Kan WC, Chang WT. Hyperbaric oxygen therapy attenuated limb ischemia in mice with high-fat diet by restoring Sirtuin 1 and mitochondrial function. Free Radic Biol Med 2025; 230:263-272. [PMID: 39956474 DOI: 10.1016/j.freeradbiomed.2025.01.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/18/2025] [Accepted: 01/31/2025] [Indexed: 02/18/2025]
Abstract
Hyperbaric oxygen therapy (HBO) shows promise as a treatment for peripheral artery disease (PAD), particularly when complicated by metabolic syndrome and diabetes. However, its precise effects on endothelial function remain unclear. This study explored the impact of HBO on angiogenesis and apoptosis in high-fat diet (HFD)-fed mice with limb ischemia, focusing on the role of sirtuin 1 (SIRT1). After 8 weeks on a chow or HFD, mice underwent unilateral femoral artery ligation and received HBO (3 ATA, 1 h/day for 5 days). HBO improved blood flow, enhanced vascular density, and reduced apoptosis in ischemic calf muscles of HFD-fed mice. In vitro, human umbilical vein endothelial cells (HUVECs) were subjected to high-glucose and oxygen-glucose deprivation (OGD) conditions, with or without HBO. HBO restored cell proliferation, migration, and tube formation under these conditions, reduced mitochondrial dysfunction, and decreased reactive oxygen species (ROS) production. However, these benefits were reversed by treatment with sirtinol, a SIRT1 inhibitor. HBO also increased SIRT1 expression and shifted mitochondrial dynamics toward fusion. Additionally, HBO upregulated angiogenesis-related proteins (VEGF, VEGFR, and SIRT1) while downregulating apoptosis-associated proteins (Bax, caspase-3, and p53). Collectively, these findings suggest that HBO enhances angiogenesis and reduces apoptosis in both in vivo and in vitro ischemia models, primarily through SIRT1 activation.
Collapse
Affiliation(s)
- Chon-Seng Hong
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan; Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Nan-Chun Wu
- Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy and Science, Tainan, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Yu-Wen Lin
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - You-Cheng Lin
- Department of Surgery, Section of Plastic and Reconstructive Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jhih-Yuan Shih
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan; Department of Cardiology, Chi Mei Medical Center, Tainan, 710, Taiwan; School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, 804, Taiwan
| | - Ko-Chi Niu
- Department of Hyperbaric Oxygen Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Mao-Tsun Lin
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Zhih-Cherng Chen
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan; Department of Cardiology, Chi Mei Medical Center, Tainan, 710, Taiwan; School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, 804, Taiwan
| | - Wei-Chih Kan
- Division of Nephrology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan; Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan, Taiwan; Department of Radiology, Chi-Mei Medical Center, Tainan, Taiwan.
| | - Wei-Ting Chang
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan; Department of Cardiology, Chi Mei Medical Center, Tainan, 710, Taiwan; School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, 804, Taiwan; School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan.
| |
Collapse
|
9
|
Ilyas S, Lee D. Exploring the Role of BCL2 Interactome in Cancer: A Protein/Residue Interaction Network Analysis. BIOLOGY 2025; 14:261. [PMID: 40136517 PMCID: PMC11940271 DOI: 10.3390/biology14030261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/27/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
BCL2 is a critical regulator of intrinsic and extrinsic pathways of apoptosis that have been implicated in cancer progression and therapeutic resistance. In this study, the protein-protein interactions (PPIs) of BCL2 with potential binding partners and their role in cancer was investigated. A comprehensive PPI network for BCL2 has been generated by using the Protein Interactions Network Analysis (PINA) platform to identify key interactors. To further investigate the network, Molecular Operating Environment (MOE), Search Tool for the Retrieval of Interacting Genes (STRING), Residue Interaction Network Generation (RING), and the gProfiler server were used. Docking and Molecular Dynamics (MD) simulations were performed by using HDOCK and Gromacs to analyze the binding dynamics and stability of protein complexes. The BCL2 interactome revealed that three key interactors (p53, RAF1, and MAPK1) are involved in cancer-related processes. Docking studies highlighted BCL2 residues such as ASP111, ASP140, ARG107, and ARG146 that were predominantly involved in multiple hydrogen bonds, ionic interactions, and van der Waals contacts, highlighting conserved binding sites that play critical roles in the stability and specificity of protein-protein interactions. MD simulations (200 ns) of the BCL2-p53 complex showed that the RMSD was increased, suggesting the suppression of BCL2's anti-apoptotic activity by p53. The RMSD for BCL2-RAF1 was also increased, showing protein domain structural rearrangements that enhance BCL2 anti-apoptotic activity. The BCL2-MAPK1 complex revealed structural, distinct flexibility patterns and dynamic hydrogen bonding interactions. These findings provide valuable insights into the molecular dynamics by which BCL2 modulates apoptosis and its potential as a promising therapeutic in cancer and apoptosis-related diseases.
Collapse
Affiliation(s)
- Sidra Ilyas
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdaero, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| | - Donghun Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdaero, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| |
Collapse
|
10
|
Du F, Zhang A, Qi X, Yin R, Jiang T, Li J. Novel Camptothecin Derivative 9c with Enhanced Antitumor Activity via NSA2-EGFR-P53 Signaling Pathway. Int J Mol Sci 2025; 26:1987. [PMID: 40076615 PMCID: PMC11900506 DOI: 10.3390/ijms26051987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/12/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Therapeutic challenges persist in the management of non-small cell lung cancer (NSCLC) in oncology. Camptothecins have demonstrated as crucial agents in tumor therapy; however, their efficacy is significantly hindered by adverse effects and drug resistance. Herein, we present a novel camptothecin derivative named 9c, which exhibits impressive anti-NSCLC potency surpassing the widely recognized camptothecin analog FL118 through a novel mechanism. Our findings demonstrated that 9c effectively inhibited tumor malignancy through cell cycle arrest and apoptosis induction with the transcriptional downregulation of anti-apoptotic genes including survivin, Mcl-1, Bcl-2, and XIAP. Mechanistically, 9c induced a wild-type p53 expression by destabilizing the NSA2-EGFR axis, thus delaying the cell cycle progression and ultimately triggering apoptosis. 9c significantly inhibited the growth of the NSCLC xenograft in vivo without observed side toxicity. Importantly, it complemented the therapeutic advantages of the novel drug AMG510 for addressing KRAS-mutant NSCLC. Collectively, these findings position 9c as a promising candidate with innovative approaches to combat NSCLC.
Collapse
Affiliation(s)
- Fu Du
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (F.D.); (A.Z.); (X.Q.); (R.Y.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National, Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Aotong Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (F.D.); (A.Z.); (X.Q.); (R.Y.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National, Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Xin Qi
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (F.D.); (A.Z.); (X.Q.); (R.Y.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National, Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Ruijuan Yin
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (F.D.); (A.Z.); (X.Q.); (R.Y.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National, Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (F.D.); (A.Z.); (X.Q.); (R.Y.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National, Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Jing Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (F.D.); (A.Z.); (X.Q.); (R.Y.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National, Laboratory for Marine Science and Technology, Qingdao 266003, China
| |
Collapse
|
11
|
Alaouna M, Molefi T, Khanyile R, Chauke-Malinga N, Chatziioannou A, Luvhengo TE, Raletsena M, Penny C, Hull R, Dlamini Z. The potential of the South African plant Tulbaghia Violacea Harv for the treatment of triple negative breast cancer. Sci Rep 2025; 15:5737. [PMID: 39962120 PMCID: PMC11832780 DOI: 10.1038/s41598-025-88417-2] [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: 07/03/2024] [Accepted: 01/28/2025] [Indexed: 02/20/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is difficult to treat and has a low five-year survival rate. In South Africa, a large percentage of the population still relies on traditional plant-based medicine. To establish the utility of both methanol and water-soluble extracts from the leaves of Tulbaghia violacea, cytotoxicity assays were carried out to establish the IC50 values against a TNBC cell line. Cell cycle and apoptosis assays were carried out using the extracts. To identify the molecular compounds, present in water-soluble leaf extracts, NMR spectroscopy was performed. Compounds of interest were then used in computational docking studies with the anti-apoptotic protein COX-2. The IC50 values for the water- and methanol-soluble extracts were determined to be 400 and 820 µg/mL, respectively. The water-soluble extract induced apoptosis in the TNBC cell line to a greater extent than in the normal cell line. RNAseq indicated that there was an increase in the transcription of pro-apoptotic genes in the TNBC cell line. The crude extract also caused these cells to stall in the S phase. Of the 61 compounds identified in this extract, five demonstrated a high binding affinity for COX-2. Based on these findings, the compounds within the extract show significant potential for further investigation as candidates for the development of cancer therapeutics, particularly for TNBC.
Collapse
Affiliation(s)
- Mohammed Alaouna
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Thulo Molefi
- Department of Medical Oncology, Steve Biko Academic Hospital, University of Pretoria, Pretoria, 0001, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa
| | - Richard Khanyile
- Department of Medical Oncology, Steve Biko Academic Hospital, University of Pretoria, Pretoria, 0001, South Africa
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa
| | - Nkhensani Chauke-Malinga
- Papillon Aesthetics, Suite 302b Netcare Linksfield Hospital, 24 12th Ave, Linksfield West, Johannesburg, 2192, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa
| | - Aristotelis Chatziioannou
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa
| | - Thifhelimbilu Emmanuel Luvhengo
- Department of Surgery, Charlotte Maxeke Johannesburg Academic Hospital, University of the Witwatersrand, Parktown, Johannesburg, 2193, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Maropeng Raletsena
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Chemistry, University of South Africa, Florida Campus, Johannesburg, South Africa
| | - Clement Penny
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Rodney Hull
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa.
| | - Zodwa Dlamini
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa.
| |
Collapse
|
12
|
Lin Z, Feng Y, Wang J, Men Z, Ma X. Microbiota governs host chenodeoxycholic acid glucuronidation to ameliorate bile acid disorder induced diarrhea. MICROBIOME 2025; 13:36. [PMID: 39905483 PMCID: PMC11792533 DOI: 10.1186/s40168-024-02011-8] [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: 03/08/2024] [Accepted: 12/17/2024] [Indexed: 02/06/2025]
Abstract
BACKGROUND Disorder in bile acid (BA) metabolism is known to be an important factor contributing to diarrhea. However, the pathogenesis of BA disorder-induced diarrhea remains unclear. METHODS The colonic BA pool and microbiota between health piglets and BA disorder-induced diarrheal piglets were compared. Fecal microbiota transplantation and various cell experiments further indicated that chenodeoxycholic acid (CDCA) metabolic disorder produced CDCA-3β-glucuronide, which is the main cause of BA disorder diarrhea. Non-targeted metabolomics uncovered the inhibition of the BA glucuronidation by Lactobacillus reuteri (L. reuteri) is through deriving indole-3-carbinol (I3C). In vitro, important gene involved in the reduction of BA disorder induced-diarrhea were screened by RNA transcriptomics sequencing, and activation pathway of FXR-SIRT1-LKB1 to alleviate BA disorder diarrhea and P53-mediated apoptosis were proposed in vitro by multifarious siRNA interference, CO-IP, immunofluorescence, and so on, which mechanism was also verified in a variety of mouse models. RESULTS Here, we reveal for the first time that core microbiota derived I3C represses gut epithelium glucuronidation, particularly 3β-glucuronic CDCA production, which reaction is mediated by host UDP glucuronosyltransferase family 1 member A4 (UGT1A4) and necessary of BA disorder induced diarrhea. Mechanistically, L. reuteri derived I3C activates aryl hydrocarbon receptor to decrease UGT1A4 transcription and CDCA-3β-glucuronide content, thereby upregulating FXR-SIRT1-LKB1 signal. LKB1 binds with P53 based on protein interaction, ultimately resists to apoptosis and diarrhea. Moreover, I3C assists CDCA to attain the ameliorative effects of FXR activation in BA disorder diarrhea, through reversion of abnormal metabolism pathway, improving the outcomes of CDCA supplement. CONCLUSION These findings uncover the crucial interplay between gut epithelial cells and microbes, highlighting UGT1A4-mediated conversion of CDCA-3β-glucuronide as a key target for ameliorating BA disorder-induced diarrhea. Video Abstract.
Collapse
Affiliation(s)
- Zishen Lin
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Yue Feng
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Jinping Wang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Zhaoyue Men
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China.
| |
Collapse
|
13
|
Chen X, Tan X, Peng Z, Wang X, Guo W, Li D, Yang Y, Zhou D, Chen L. Biomarkers and potential function analysis of triple-negative breast cancer screening based on bioinformatics. Cancer Biomark 2025; 42:18758592241308738. [PMID: 40179432 DOI: 10.1177/18758592241308738] [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] [Indexed: 04/05/2025]
Abstract
This study aims to identify and validate potential endogenous biomarkers for triple-negative breast cancer (TNBC). TNBC microarray data (GSE38959, GSE53752) were retrieved from the Gene Expression Omnibus (GEO) database, and principal component analysis (PCA) was performed to evaluate the reliability of the data. The microarray datasets were merged, and differentially expressed genes (DEGs) were identified using R software. Functional enrichment analysis of the DEGs was conducted using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. The most disease-relevant module was identified through Weighted Gene Co-expression Network Analysis (WGCNA), and genes within this module were intersected with the DEGs. The intersecting genes underwent Least Absolute Shrinkage and Selection Operator (LASSO) regression analysis to minimize errors and identify TNBC-specific genes. Sensitivity and survival analyses were performed on the identified specific genes. There were 10 TNBC-specific genes identified: RRM2, DEPDC1, FIGF, TACC3, E2F1, CDO1, DST, MCM4, CHEK1, and PLSCR4. RT-qPCR analysis showed significant upregulation of CDO1, MCM4, DEPDC1, RRM2, and E2F1 in MDA-MB-231, CAL-148, and MFM-223 compared to MCF-10A. Our findings provide new insights into TNBC pathogenesis and potential therapeutic strategies, with important clinical implications for further understanding TNBC mechanisms and developing innovative treatments.
Collapse
Affiliation(s)
- Xing Chen
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pharmacy, Chongqing Health Center for Women and Children, Chongqing, China
| | - Xiaodan Tan
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pharmacy, Chongqing Health Center for Women and Children, Chongqing, China
| | - Zhe Peng
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pharmacy, Chongqing Health Center for Women and Children, Chongqing, China
| | - Xiaoli Wang
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pharmacy, Chongqing Health Center for Women and Children, Chongqing, China
| | - Wenjia Guo
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pharmacy, Chongqing Health Center for Women and Children, Chongqing, China
| | - Dan Li
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pharmacy, Chongqing Health Center for Women and Children, Chongqing, China
| | - Yang Yang
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pharmacy, Chongqing Health Center for Women and Children, Chongqing, China
| | - Duanfang Zhou
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pharmacy, Chongqing Health Center for Women and Children, Chongqing, China
| | - Lin Chen
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pharmacy, Chongqing Health Center for Women and Children, Chongqing, China
| |
Collapse
|
14
|
Zhang Y, Qiu K, Ren J, Zhao Y, Cheng P. Roles of human papillomavirus in cancers: oncogenic mechanisms and clinical use. Signal Transduct Target Ther 2025; 10:44. [PMID: 39856040 PMCID: PMC11760352 DOI: 10.1038/s41392-024-02083-w] [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: 07/24/2024] [Revised: 10/19/2024] [Accepted: 11/24/2024] [Indexed: 01/27/2025] Open
Abstract
Human papillomaviruses, particularly high-risk human papillomaviruses, have been universally considered to be associated with the oncogenesis and progression of various cancers. The genome of human papillomaviruses is circular, double-stranded DNA that encodes early and late proteins. Each of the proteins is of crucial significance in infecting the epithelium of host cells persistently and supporting viral genome integrating into host cells. Notably, E6 and E7 proteins, classified as oncoproteins, trigger the incidence of cancers by fostering cell proliferation, hindering apoptosis, evading immune surveillance, promoting cell invasion, and disrupting the balance of cellular metabolism. Therefore, targeting human papillomaviruses and decoding molecular mechanisms by which human papillomaviruses drive carcinogenesis are of great necessity to better treat human papillomaviruses-related cancers. Human papillomaviruses have been applied clinically to different facets of human papillomavirus-related cancers, including prevention, screening, diagnosis, treatment, and prognosis. Several types of prophylactic vaccines have been publicly utilized worldwide and have greatly decreased the occurrence of human papillomavirus-related cancers, which have benefited numerous people. Although various therapeutic vaccines have been developed and tested clinically, none of them have been officially approved to date. Enhancing the efficacy of vaccines and searching for innovative technologies targeting human papillomaviruses remain critical challenges that warrant continuous research and attention in the future.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ke Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jianjun Ren
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Yu Zhao
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Ping Cheng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
15
|
Zhou H, Yan S. Mechanisms of p53 core tetramer stability mediated by multi-interface interactions: A molecular dynamics study. Arch Biochem Biophys 2025; 763:110210. [PMID: 39603375 DOI: 10.1016/j.abb.2024.110210] [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: 09/21/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 11/29/2024]
Abstract
p53 is a tumor suppressor protein for impeding cancer development and maintaining genetic integrity. The formation of the p53 core tetramer is regulated by multiple cooperative interaction interfaces. To investigate the internal mechanisms of tetramer stability, we performed all-atom molecular dynamics simulations. Our findings indicate that the symmetric interface maintains highly conserved interactions, while the dimer-dimer interface displays notable flexibility. Additionally, we identified a novel salt bridge at the dimer-dimer interface that significantly contributes to the interaction energy. Moreover, the affinity of p53 for DNA is more than twice that of protein-protein interactions, driven primarily by five key residues that form multiple hydrogen bonds. Through independent simulations of the two dimeric models, we provide a theoretical explanation for why only the symmetric dimeric structure has been observed experimentally. The study identifies key regions and residues that contribute to stability at the inter-molecular interaction interfaces within the p53 tetramer, and highlight the important roles of each contact surface in the formation and stability of the tetramer.
Collapse
Affiliation(s)
- Han Zhou
- School of Physics and Astronomy, Beijing Normal University, Beijing 100875, China
| | - Shiwei Yan
- School of Physics and Astronomy, Beijing Normal University, Beijing 100875, China; Faculty of Arts and Sciences, Beijing Normal University at Zhuhai, Zhuhai 519087, China.
| |
Collapse
|
16
|
Das A, Mitra A, Ghosh S, Sarkar S, Pal PK, Bandyopadhyay D, Chattopadhyay S. Arsenic-induced transition of thymic inflammation-to-fibrosis involves Stat3-Twist1 interaction: Melatonin to the rescue. Biofactors 2025; 51:e2110. [PMID: 39096306 DOI: 10.1002/biof.2110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 07/19/2024] [Indexed: 08/05/2024]
Abstract
Groundwater arsenic is a notorious toxicant and exposure to environmentally relevant concentrations persists as a healthcare burden across the world. Arsenic has been reported to jeopardize the normal functioning of the immune system, but there are still gaps in the understanding of thymic T cell biology. Immunotoxic influence of arsenic in thymic integrity demands a potent restorative molecule. The objectives of this study were to examine key signaling cross-talks associated with arsenic-induced immune alterations in the thymus and propose melatonin as a potential candidate against immunological complications arising from arsenic exposure. Swiss albino mice were exposed to sodium arsenite (0.05 mg/L; in drinking water) and melatonin (IP:10 mg/kg BW) for 28 days. Melatonin successfully protected thymus from arsenic-mediated tissue degeneration and maintained immune homeostasis including T cell maturation and proliferation by mitigating oxidative stress through Nrf2 upregulation. Additionally, melatonin exerted ameliorative effect against arsenic-induced apoptosis and inflammation by inhibiting p53-mediated mitochondrial cell death pathway and NF-κB-p65/STAT3-mediated proinflammatory pathway, respectively. For the first time, we showed that arsenic-induced profibrotic changes were inhibited by melatonin through targeting of inflammation-associated EMT. Our findings clearly demonstrate that melatonin can be a viable and promising candidate in combating arsenic-induced immune toxicity with no collateral damage, making it an important research target.
Collapse
Affiliation(s)
- Ankur Das
- Department of Physiology, University of Calcutta, Kolkata, India
| | - Ankan Mitra
- Department of Physiology, University of Calcutta, Kolkata, India
| | - Sourav Ghosh
- Department of Physiology, University of Calcutta, Kolkata, India
| | - Swaimanti Sarkar
- Department of Physiology, University of Calcutta, Kolkata, India
| | - Palash Kumar Pal
- Department of Physiology, University of Calcutta, Kolkata, India
| | | | - Sreya Chattopadhyay
- Department of Physiology, University of Calcutta, Kolkata, India
- Centre for Research in Nanoscience and Nanotechnology (CRNN), University of Calcutta, Kolkata, India
| |
Collapse
|
17
|
Chen L, Chen D, Pan Y, Mo Y, Lai B, Chen H, Zhang DW, Xia XD. Inhibition of mitochondrial OMA1 ameliorates osteosarcoma tumorigenesis. Cell Death Dis 2024; 15:786. [PMID: 39487118 PMCID: PMC11530700 DOI: 10.1038/s41419-024-07127-1] [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: 04/05/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 11/04/2024]
Abstract
OMA1 is an ATP-independent zinc metalloprotease essential for maintaining mitochondrial homeostasis and plays a vital role in tumorigenesis. Depending on the type of cancer, a decrease in OMA1 expression has been linked to a varying prognosis for patients. The role of OMA1 in human osteosarcoma (OS), one of the most prevalent malignant bone tumors, remains elusive. Here, we observed elevated OMA1 expression in OS tumor tissues from four patients with advanced OS. Knockout of OMA1 in OS cells significantly reduces OS tumor weight and size, and lung metastatic nodules in BALB/c nude mice. Immunohistochemistry analysis showed a significant decrease in Ki67 and an increase in Cleaved-caspase 3 in OMA1 knockout tumor samples. Mechanistically, we found that OMA1 deficiency increases the levels of PINK1 and Parkin and consequently induces excessive mitophagy, leading to increased apoptosis and reduced cell proliferation and invasion in OS cells. Specifically, OMA1 deficiency reduces the amount of cytosolic p53 and p53-associated cytosolic Parkin but increases mitochondrial p53, which may lead to enhanced apoptosis. Regarding the effect on cell proliferation and invasion, loss of OMA1 reduces mitochondrial ROS levels and increases cytosolic glycogen synthase kinase 3β (GSK3β) levels, thereby increasing interaction between GSK3β and β-catenin and then reducing cytosolic and nuclear β-catenin. This contributes to reduced cell proliferation and migration in OMA1-deficient cells. Moreover, we found that ciclopirox (CPX), an antifungal drug, induces OMA1 self-cleavage and L-OMA1 degradation in cultured OS cells. CPX also reduces tumor development of control OS cells but not OMA1-deficient OS cells in mice. These findings strongly support the important role of OMA1 in OS tumorigenesis and suggest that OMA1 may be a valuable prognostic marker and a promising therapeutic target for OS.
Collapse
Affiliation(s)
- Lingyan Chen
- Department of Orthopedics, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China.
| | - Dejian Chen
- Department of Orthopedics, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Yiming Pan
- Department of Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yimei Mo
- Department of Orthopedics, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Biyu Lai
- Department of Radiology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Huiguang Chen
- Department of Orthopedics, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Da-Wei Zhang
- Department of Pediatrics, Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| | - Xiao-Dan Xia
- Department of Orthopedics, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China.
| |
Collapse
|
18
|
Wang H, Liu C, Jin K, Li X, Zheng J, Wang D. Research advances in signaling pathways related to the malignant progression of HSIL to invasive cervical cancer: A review. Biomed Pharmacother 2024; 180:117483. [PMID: 39353319 DOI: 10.1016/j.biopha.2024.117483] [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: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
The progression of high-grade squamous intraepithelial lesion (HSIL) to invasive cervical cancer (ICC) is a complex process involving persistent human papillomavirus (HPV) infection and changes in signal transduction regulation, energy and material metabolism, cell proliferation, autoimmune, and other biological process in vaginal microenvironment and immune microenviroment. Signaling pathways are a series of interacting molecules in cells that regulate various physiological functions of cells, such as growth, differentiation, metabolism, and death. In the progression of HSIL to ICC, abnormal activation or inhibition in signaling pathways plays an essensial role. This review presented some signaling pathways related to the malignant progression of HSIL to ICC, including p53, Rb, PI3K/AKT/mTOR, Wnt/β-catenin, Notch, NF-κB, MAPK, TGF-β, JAK-STAT, Hippo, and Hedgehog. The molecular mechanisms involved in the biological process of pathway regulation were also analyzed, in order to illustrate the molecular pathway of HSIL progression to ICC and provide references for the development of more effective prevention and treatment methods.
Collapse
Affiliation(s)
- Huifang Wang
- Department of Obstetrics and Gynecology, Quanzhou Medical College, Quanzhou, Fujian 362010, China
| | - Chang Liu
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; Key Clinical Specialty of Liaoning Province, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Keer Jin
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; Key Clinical Specialty of Liaoning Province, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Xiang Li
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Jiaxin Zheng
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Danbo Wang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; Key Clinical Specialty of Liaoning Province, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China.
| |
Collapse
|
19
|
Zhai P, Ouyang XH, Yang ML, Lin L, Li JY, Li YM, Cheng X, Zhu R, Hu DS. Luteolin protects against myocardial ischemia/reperfusion injury by reducing oxidative stress and apoptosis through the p53 pathway. JOURNAL OF INTEGRATIVE MEDICINE 2024; 22:652-664. [PMID: 39343710 DOI: 10.1016/j.joim.2024.09.001] [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: 09/01/2023] [Accepted: 08/13/2024] [Indexed: 10/01/2024]
Abstract
OBJECTIVE Myocardial ischemia/reperfusion injury (MIRI) is an obstacle to the success of cardiac reperfusion therapy. This study explores whether luteolin can mitigate MIRI by regulating the p53 signaling pathway. METHODS Model mice were subjected to a temporary surgical ligation of the left anterior descending coronary artery, and administered luteolin. The myocardial infarct size, myocardial enzyme levels, and cardiac function were measured. Latent targets and signaling pathways were screened using network pharmacology and molecular docking. Then, proteins related to the p53 signaling pathway, apoptosis and oxidative stress were measured. Hypoxia/reoxygenation (HR)-incubated HL1 cells were used to validate the effects of luteolin in vitro. In addition, a p53 agonist and an inhibitor were used to investigate the mechanism. RESULTS Luteolin reduced the myocardial infarcted size and myocardial enzymes, and restored cardiac function in MIRI mice. Network pharmacology identified p53 as a hub target. The bioinformatic analyses showed that luteolin had anti-apoptotic and anti-oxidative properties. Additionally, luteolin halted the activation of p53, and prevented both apoptosis and oxidative stress in myocardial tissue in vivo. Furthermore, luteolin inhibited cell apoptosis, JC-1 monomer formation, and reactive oxygen species elevation in HR-incubated HL1 cells in vitro. Finally, the p53 agonist NSC319726 downregulated the protective attributes of luteolin in the MIRI mouse model, and both luteolin and the p53 inhibitor pifithrin-α demonstrated a similar therapeutic effect in the MIRI mice. CONCLUSION Luteolin effectively treats MIRI and may ameliorate myocardial damage by regulating apoptosis and oxidative stress through its targeting of the p53 signaling pathway. Please cite this article as: Zhai P, Ouyang XH, Yang ML, Lin L, Li JY, Li YM, Cheng X, Zhu R, Hu DS. Luteolin protects against myocardial ischemia/reperfusion injury by reducing oxidative stress and apoptosis through the p53 pathway. J Integr Med. 2024; 22(6): 652-664.
Collapse
Affiliation(s)
- Pan Zhai
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Xiao-Hu Ouyang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Meng-Ling Yang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Lan Lin
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Jun-Yi Li
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Yi-Ming Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Rui Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - De-Sheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China; China-Russia Medical Research Center for Stress Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| |
Collapse
|
20
|
Zhao J, Xing W, Ji C, Hu H, Zhang Y, Wang Z, Liu J. Nucleophosmin 1 overexpression enhances neuroprotection by attenuating cellular stress in traumatic brain injury. Exp Neurol 2024; 383:115019. [PMID: 39428041 DOI: 10.1016/j.expneurol.2024.115019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/08/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Traumatic Brain Injury (TBI) is a multifaceted injury that can cause a wide range of symptoms and impairments, leading to significant effects on brain function. Nucleophosmin 1 (NPM1), a versatile phosphoprotein located in the nucleolus, is being recognized as a possible controller of cellular stress reactions and could be important in reducing neuro dysfunction caused by TBI. However the critical roles of NPM1 in cellular stress in TBI remains unclear. METHODS We employed a control cortical impact mouse model and a scratch-induced primary neuronal culture model. Hematoxylin and eosin staining was used to evaluate tissue damage and cellular changes, with NPM1 expression in the cortical area assessed through immunofluorescence staining and Western blot analysis. Neuronal morphology was assessed using Nissl staining. Behavioral assessments were performed to evaluate the impact of NPM1 overexpression on neurobehavioral results in TBI mice. Mitochondrial function was assessed using an Extracellular Flux Analyzer. RESULTS Following TBI, an increase in NPM1 expression was observed, with a peak at 72 h post-injury. Increased levels of NPM1 resulted in decreased neuronal cell death, as shown by Nissl staining, and lower levels of Caspase 8, APE1, H2AX, and 8-OHDG expression, indicating a reduction in DNA damage. NPM1 overexpression also resulted in improved neurobehavioral outcomes, characterized by decreased neurological deficits and enhanced motor function post-TBI. Additionally, in vitro, scratch-induction experiments revealed that NPM1 overexpression mitigated mitochondrial damage, as evidenced by the downregulation of P53, BCL2, and Cyto C expression levels and improvements in mitochondrial respiratory function. CONCLUSION These findings suggest NPM1 as a promising target for developing interventions to alleviate TBI-related cellular stress and promote neuronal survival.
Collapse
Affiliation(s)
- Jiashuo Zhao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China; Department of Neurosurgery, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu Province, 215000, China
| | - Weixin Xing
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China; Department of Neurosurgery, The 928th Hospital of People's Liberation Army Joint Logistic Support Force, Haikou, Hainan Province, 570000, China
| | - Chengyuan Ji
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China
| | - Hongwei Hu
- Department of Neurosurgery, Changzhou Jintan First People's Hospital Affiliated to Jiangsu University, 500 Jintan Avenue, Jintan 210036, China
| | - Yuanqing Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China
| | - Zongqi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China.
| | - Jiangang Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China.
| |
Collapse
|
21
|
Cang T, Huang N, Nie D, Chen L, Shao K, Wu C, Chen C, Wang Y. Mixture effect of parental exposure to triazophos and fenvalerate on the early development of zebrafish offspring. CHEMOSPHERE 2024; 365:143415. [PMID: 39332582 DOI: 10.1016/j.chemosphere.2024.143415] [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: 08/02/2024] [Revised: 09/15/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Triazophos (TRI) and fenvalerate (FEN) have been extensively used in the world and frequently coexist in the water environments, might pose health risk to aquatic species. However, investigations of their mixture toxic effects on offspring after parental exposure have been neglected, especially for aquatic vertebrates such fish. To address this knowledge gap, parental zebrafish (F0 generation) were exposed to TRI, FEN and their mixture for 60 days, as well as the embryos (F1 generation) were hatched without or with continued corresponding exposures at the same concentrations until 7 days post fertilization. The results exhibited that exposure to TRI and FEN altered the expression levels of biomarkers associated with several biological processes, such as apoptosis and inflammatory response. Compared to individual exposure in the F1 generation, the co-exposure to TRI and FEN resulted in increased the expression of T4 and cc-chem mRNA and decreased the expression of ROS, trα, il-8, and gpx mRNA when the F0 generation was similarly exposed. These results revealed that the co-exposure to TRI and FEN has detrimental effects on fish progeny following parental exposure, even if the progeny are not directly exposed to the pesticides, and such negative effects may be intensified if the offspring continue to be exposed. This study enhances the understanding of the harmful impacts of parental exposure to the pesticide mixture on descendants and holds implications for the ecological risk assessment of pesticide mixtures in aquatic vertebrates. Further mechanistic studies are necessary to gain a deeper insight into the mixture effects of pesticides and other kinds of pollutants on subsequent offspring following parental exposure.
Collapse
Affiliation(s)
- Tao Cang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China
| | - Nan Huang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Dongxing Nie
- Institute for the Control of Agrochemicals, Ministry of Agriculture and Rural Affairs, Beijing, 100125, China
| | - Liping Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China
| | - Kan Shao
- Department of Environmental and Occupational Health, School of Public Health, Indiana University, Bloomington, 47405, USA
| | - Changxing Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China
| | - Chen Chen
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Yanhua Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China.
| |
Collapse
|
22
|
Brogyanyi T, Kejík Z, Veselá K, Dytrych P, Hoskovec D, Masařik M, Babula P, Kaplánek R, Přibyl T, Zelenka J, Ruml T, Vokurka M, Martásek P, Jakubek M. Iron chelators as mitophagy agents: Potential and limitations. Biomed Pharmacother 2024; 179:117407. [PMID: 39265234 DOI: 10.1016/j.biopha.2024.117407] [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: 06/14/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
Mitochondrial autophagy (mitophagy) is very important process for the maintenance of cellular homeostasis, functionality and survival. Its dysregulation is associated with high risk and progression numerous serious diseases (e.g., oncological, neurodegenerative and cardiovascular ones). Therefore, targeting mitophagy mechanisms is very hot topic in the biological and medicinal research. The interrelationships between the regulation of mitophagy and iron homeostasis are now becoming apparent. In short, mitochondria are central point for the regulation of iron homeostasis, but change in intracellular cheatable iron level can induce/repress mitophagy. In this review, relationships between iron homeostasis and mitophagy are thoroughly discussed and described. Also, therapeutic applicability of mitophagy chelators in the context of individual diseases is comprehensively and critically evaluated.
Collapse
Affiliation(s)
- Tereza Brogyanyi
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Petr Dytrych
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - David Hoskovec
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - Michal Masařik
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Petr Babula
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Tomáš Přibyl
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Jaroslav Zelenka
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Martin Vokurka
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic.
| |
Collapse
|
23
|
Toshkova-Yotova T, Sulikovska I, Djeliova V, Petrova Z, Ognyanov M, Denev P, Toshkova R, Georgieva A. Exopolysaccharides from the Green Microalga Strain Coelastrella sp. BGV-Isolation, Characterization, and Assessment of Anticancer Potential. Curr Issues Mol Biol 2024; 46:10312-10334. [PMID: 39329966 PMCID: PMC11431334 DOI: 10.3390/cimb46090614] [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: 07/31/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024] Open
Abstract
Algal metabolites have been extensively studied as potential anticancer therapeutics. Among them, polysaccharides have attracted much attention because of their beneficial biological effects and safety. In the present research, the chemical characteristics, antitumor, and proapoptotic activities of extracellular polysaccharides (EPS) isolated from a new Bulgarian strain of the green microalga Coelastrella sp. BGV were investigated. A fast and convenient method of precipitation with cold ethanol was used to isolate EPS from the culture medium. The chemical characteristics of the isolated EPS were examined by colorimetric and spectrophotometric analyses, HPSEC-RID and HPLC-UV chromatography, and FT-IR spectroscopy. The results showed that the isolated EPS sample consists of three carbohydrate fractions with different molecular weights (11.5 × 104 Da, 30.7 × 104 Da, and 72.4 × 104 Da, respectively) and contains 7.14 (w/w%) protein. HPLC-UV analysis revealed the presence of galactose and fucose. The total uronic acid content in the sample was 4.5 (w/w%). The IR-FT spectrum of EPS revealed the presence of various functional groups typical of a polysaccharide (or proteoglycan) composed primarily of neutral sugars. The anticancer potential of the obtained EPS was assessed using cell lines with cancerous and non-cancerous origins as in vitro experimental models. The results of the performed MTT assay showed that EPS reduced the viability of the cervical and mammary carcinoma cell lines HeLa and MCF-7, while the control non-cancer cell lines BALB/3T3 and HaCaT were less affected. The HeLa cell line showed the highest sensitivity to the effects of EPS and was therefore used for further studies of its anticancer potential. The ability of EPS to inhibit cancer cell migration was demonstrated by wound-healing (scratch) assay. The cell cycle FACS analysis indicated that the EPS treatment induced significant increases in the sub G1 cell population and decreases of the percentages of cells in the G1, S, and G2-M phases, compared to the control. The fluorescent microscopy studies performed using three different staining methods in combination with Annexin V-FITC flow cytometric analysis clearly demonstrate the ability of EPS to induce cancer cell death via the apoptosis pathway. Moreover, an altered pattern and intensity of the immunocytochemical staining for the apoptosis- and proliferation-related proteins p53, bcl2, and Ki67 was detected in EPS-treated HeLa cancer cells as compared to the untreated controls. The obtained results characterize the new local strain of green microalgae Coelastrella sp. BGV as a producer of EPS with selective antitumor activity and provide an opportunity for further studies of its pharmacological and biotechnological potential.
Collapse
Affiliation(s)
- Tanya Toshkova-Yotova
- Department of Plant Ecophysiology, Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., 21, 1113 Sofia, Bulgaria
| | - Inna Sulikovska
- Department of Pathology, Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., 25, 1113 Sofia, Bulgaria
| | - Vera Djeliova
- Department of Molecular Biology of Cell Cycle, Institute of Molecular Biology "Acad. R. Tsanev", Bulgarian Academy of Sciences, Acad. G. Bonchev Str., 21, 1113 Sofia, Bulgaria
| | - Zdravka Petrova
- Department of Pathology, Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., 25, 1113 Sofia, Bulgaria
| | - Manol Ognyanov
- Laboratory of Biologically Active Substances, Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000 Plovdiv, Bulgaria
| | - Petko Denev
- Laboratory of Biologically Active Substances, Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000 Plovdiv, Bulgaria
| | - Reneta Toshkova
- Department of Pathology, Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., 25, 1113 Sofia, Bulgaria
| | - Ani Georgieva
- Department of Pathology, Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., 25, 1113 Sofia, Bulgaria
| |
Collapse
|
24
|
Lei YR, He JY, Fu XM, Huang CF, Lin YX, Dai LL, Chen ZA, Zhang ZP, Liu FM, Qin QW, Sun HY. Epinephelus coioides Sec3 promotes Singapore grouper iridovirus infection by negatively regulates immune response. FISH & SHELLFISH IMMUNOLOGY 2024; 152:109784. [PMID: 39067495 DOI: 10.1016/j.fsi.2024.109784] [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: 05/04/2024] [Revised: 06/19/2024] [Accepted: 07/19/2024] [Indexed: 07/30/2024]
Abstract
Exocyst, a protein complex, plays a crucial role in various cellular functions, including cell polarization, migration, invasion, cytokinesis, and autophagy. Sec3, known as Exoc1, is a key subunit of the Exocyst complex and can be involved in cell survival and apoptosis. In this study, two subtypes of Sec3 were isolated from Epinephelus coioides, an important marine fish in China. The role of E. coioides Sec3 was explored during Singapore grouper iridovirus (SGIV) infection, an important pathogen of marine fish which could induce 90 % mortality. E. coioides Sec3 sequences showed a high similarity with that from other species, indicating the presence of a conserved Sec3 superfamily domain. E. coioides Sec3 mRNA could be detected in all examined tissues, albeit at varying expression levels. SGIV infection could upregulate E. coioides Sec3 mRNA. Upregulated Sec3 significantly promoted SGIV-induced CPE, and the expressions of viral key genes. E. coioides Sec3 could inhibit the activation of NF-κB and AP-1, as well as SGIV-induced cell apoptosis. The results illustrated that E. coioides Sec3 promotes SGIV infection by regulating the innate immune response.
Collapse
Affiliation(s)
- Yu-Rong Lei
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Jia-Yang He
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Xue-Mei Fu
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Cui-Fen Huang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Yun-Xiang Lin
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Li-Ling Dai
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Zi-An Chen
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Ze-Peng Zhang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Fu-Min Liu
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Qi-Wei Qin
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China.
| | - Hong-Yan Sun
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China.
| |
Collapse
|
25
|
Xue Y, Cheng X, Ma ZQ, Wang HP, Zhou C, Li J, Zhang DL, Hu LL, Cui YF, Huang J, Luo T, Zheng LP. Polystyrene nanoplastics induce apoptosis, autophagy, and steroidogenesis disruption in granulosa cells to reduce oocyte quality and fertility by inhibiting the PI3K/AKT pathway in female mice. J Nanobiotechnology 2024; 22:460. [PMID: 39090717 PMCID: PMC11293132 DOI: 10.1186/s12951-024-02735-7] [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: 05/11/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Nanoplastics (NPs) are emerging pollutants that pose risks to living organisms. Recent findings have unveiled the reproductive harm caused by polystyrene nanoparticles (PS-NPs) in female animals, yet the intricate mechanism remains incompletely understood. Under this research, we investigated whether sustained exposure to PS-NPs at certain concentrations in vivo can enter oocytes through the zona pellucida or through other routes that affect female reproduction. RESULTS We show that PS-NPs disrupted ovarian functions and decreased oocyte quality, which may be a contributing factor to lower female fertility in mice. RNA sequencing of mouse ovaries illustrated that the PI3K-AKT signaling pathway emerged as the predominant environmental information processing pathway responding to PS-NPs. Western blotting results of ovaries in vivo and cells in vitro showed that PS-NPs deactivated PI3K-AKT signaling pathway by down-regulating the expression of PI3K and reducing AKT phosphorylation at the protein level, PI3K-AKT signaling pathway which was accompanied by the activation of autophagy and apoptosis and the disruption of steroidogenesis in granulosa cells. Since PS-NPs penetrate granulosa cells but not oocytes, we examined whether PS-NPs indirectly affect oocyte quality through granulosa cells using a granulosa cell-oocyte coculture system. Preincubation of granulosa cells with PS-NPs causes granulosa cell dysfunction, resulting in a decrease in the quality of the cocultured oocytes that can be reversed by the addition of 17β-estradiol. CONCLUSIONS This study provides findings on how PS-NPs impact ovarian function and include transcriptome sequencing analysis of ovarian tissue. The study demonstrates that PS-NPs impair oocyte quality by altering the functioning of ovarian granulosa cells. Therefore, it is necessary to focus on the research on the effects of PS-NPs on female reproduction and the related methods that may mitigate their toxicity.
Collapse
Affiliation(s)
- Yue Xue
- School of Basic Medical Sciences and School of Public and Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiu Cheng
- School of Basic Medical Sciences and School of Public and Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, 330006, China
| | - Zhang-Qiang Ma
- School of Basic Medical Sciences and School of Public and Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, 330006, China
| | - Hou-Peng Wang
- School of Basic Medical Sciences and School of Public and Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, 330006, China
| | - Chong Zhou
- School of Basic Medical Sciences and School of Public and Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Jia Li
- School of Basic Medical Sciences and School of Public and Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Da-Lei Zhang
- School of Basic Medical Sciences and School of Public and Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, 330006, China
| | - Liao-Liao Hu
- The 2nd affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yan-Fan Cui
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University; Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Jian Huang
- School of Basic Medical Sciences and School of Public and Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Tao Luo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University; Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, 330006, China.
| | - Li-Ping Zheng
- School of Basic Medical Sciences and School of Public and Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
26
|
Fang T, Wang J, Sun S, Deng X, Xue M, Han F, Sun B, Chen L. JinLiDa granules alleviates cardiac hypertrophy and inflammation in diabetic cardiomyopathy by regulating TP53. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155659. [PMID: 38759318 DOI: 10.1016/j.phymed.2024.155659] [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: 11/26/2023] [Revised: 03/14/2024] [Accepted: 04/19/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND JinLiDa granules (JLD) is a traditional Chinese medicine (TCM) used to treat type 2 diabetes mellitus with Qi and Yin deficiency. Clinical evidence has shown that JLD can alleviate diabetic cardiomyopathy, but the exact mechanism is not yet clear. PURPOSE The purpose of this study was to examine the potential role and mechanism of JLD in the treatment of diabetic cardiomyopathy through network pharmacological analysis and basic experiments. METHODS The targets of JLD associated with diabetic cardiomyopathy were examined by network pharmacology. Protein interaction analysis was performed on the targets, and the associated pathways were searched by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Diabetic mice were treated with low or high doses of JLD by gavage, and AC16 and H9C2 cardiomyocytes exposed to high-glucose conditions were treated with JLD. The analysis results were verified by various experimental techniques to examine molecular mechanisms. RESULTS Network pharmacological analysis revealed that JLD acted on the tumor suppressor p53 (TP53) during inflammation and fibrosis associated with diabetic cardiomyopathy. The results of basic experiments showed that after JLD treatment, ventricular wall thickening in diabetic mouse hearts was attenuated, cardiac hypertrophy and myocardial inflammation were alleviated, and the expression of cardiac hypertrophy- and inflammation-related factors in cardiomyocytes exposed to a high-glucose environment was decreased. Cardiomyocyte morphology also improved after JLD treatment. TP53 expression and the tumor necrosis factor (TNF) and transforming growth factor beta-1 (TGFβ1) signaling pathways were significantly altered, and inhibiting TP53 expression effectively alleviated the activation of the TNF and TGFβ1 signaling pathways under high glucose conditions. Overexpression of TP53 activated these signaling pathways. CONCLUSIONS JLD acted on TP53 to regulate the TNF and TGFβ1 signaling pathways, effectively alleviating cardiomyocyte hypertrophy and inflammation in high glucose and diabetic conditions. Our study provides a solid foundation for the future treatment of diabetic cardiomyopathy with JLD.
Collapse
Affiliation(s)
- Ting Fang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Jingyi Wang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Shengnan Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Xiaoqing Deng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Mei Xue
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| | - Fei Han
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Bei Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China.
| | - Liming Chen
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China.
| |
Collapse
|
27
|
Xia T, Xia Z, Tang P, Fan J, Peng X. Light-Driven Mitochondrion-to-Nucleus DNA Cascade Fluorescence Imaging and Enhanced Cancer Cell Photoablation. J Am Chem Soc 2024; 146:12941-12949. [PMID: 38685727 DOI: 10.1021/jacs.3c13095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Nucleic acids are mainly found in the mitochondria and nuclei of cells. Detecting nucleic acids in the mitochondrion and nucleus in cascade mode is crucial for understanding diverse biological processes. This study introduces a novel nucleic acid-based fluorescent styrene dye (SPP) that exhibits light-driven cascade migration from the mitochondrion to the nucleus. By introducing N-arylpyridine on one side of the styrene dye skeleton and a bis(2-ethylsulfanyl-ethy)-amino unit on the other side, we found that SPP exhibits excellent DNA specificity (16-fold, FDNA/Ffree) and a stronger binding force to nuclear DNA (-5.09 kcal/mol) than to mitochondrial DNA (-2.59 kcal/mol). SPP initially accumulates in the mitochondrion and then migrates to the nucleus within 10 s under light irradiation. By tracking the damage to nucleic acids in apoptotic cells, SPP allows the successful visualization of the differences between apoptosis and ferroptosis. Finally, a triphenylamine segment with photodynamic effects was incorporated into SPP to form a photosensitizer (MTPA-SPP), which targets the mitochondria for photosensitization and then migrates to the nucleus under light irradiation for enhanced photodynamic cancer cell treatment. This innovative nucleic acid-based fluorescent molecule with light-triggered mitochondrion-to-nucleus migration ability provides a feasible approach for the in situ identification of nucleic acids, monitoring of subcellular physiological events, and efficient photodynamic therapy.
Collapse
Affiliation(s)
- Tianping Xia
- State Key Laboratory of Fine Chemicals, Frontier Science Center for Smart Materials, Dalian University of Technology, Dalian 116024, China
| | - Zhuoran Xia
- State Key Laboratory of Fine Chemicals, Frontier Science Center for Smart Materials, Dalian University of Technology, Dalian 116024, China
| | - Peichen Tang
- State Key Laboratory of Fine Chemicals, Frontier Science Center for Smart Materials, Dalian University of Technology, Dalian 116024, China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Frontier Science Center for Smart Materials, Dalian University of Technology, Dalian 116024, China
- Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontier Science Center for Smart Materials, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
28
|
Haase A, Alefeld E, Yalinci F, Meenen DV, Busch MA, Dünker N. Gastric Inhibitory Polypeptide Receptor (GIPR) Overexpression Reduces the Tumorigenic Potential of Retinoblastoma Cells. Cancers (Basel) 2024; 16:1656. [PMID: 38730608 PMCID: PMC11083251 DOI: 10.3390/cancers16091656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Retinoblastoma (RB) is the most common malignant intraocular tumor in early childhood. Gene expression profiling revealed that the gastric inhibitory polypeptide receptor (GIPR) is upregulated following trefoil factor family peptide 1 (TFF1) overexpression in RB cells. In the study presented, we found this G protein-coupled transmembrane receptor to be co-expressed with TFF1, a new diagnostic and prognostic RB biomarker for advanced subtype 2 RBs. Functional analyses in two RB cell lines revealed a significant reduction in cell viability and growth and a concomitant increase in apoptosis following stable, lentiviral GIPR overexpression, matching the effects seen after TFF1 overexpression. In chicken chorioallantoic membrane (CAM) assays, GIPR-overexpressing RB cells developed significantly smaller CAM tumors. The effect of GIPR overexpression in RB cells was reversed by the GIPR inhibitor MK0893. The administration of recombinant TFF1 did not augment GIPR overexpression effects, suggesting that GIPR does not serve as a TFF1 receptor. Investigations of potential GIPR up- and downstream mediators suggest the involvement of miR-542-5p and p53 in GIPR signaling. Our results indicate a tumor suppressor role of GIPR in RB, suggesting its pathway as a new potential target for future retinoblastoma therapy.
Collapse
|
29
|
Zhang B, Zhang M, Tian J, Zhang X, Zhang D, Li J, Yang L. Advances in the regulation of radiation-induced apoptosis by polysaccharides: A review. Int J Biol Macromol 2024; 263:130173. [PMID: 38360238 DOI: 10.1016/j.ijbiomac.2024.130173] [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: 12/02/2023] [Revised: 02/03/2024] [Accepted: 02/12/2024] [Indexed: 02/17/2024]
Abstract
Polysaccharides are biomolecules composed of monosaccharides that are widely found in animals, plants and microorganisms and are of interest for their various health benefits. Cumulative studies have shown that the modulation of radiation-induced apoptosis by polysaccharides can be effective in preventing and treating a wide range of radiation injuries with safety and few side effects. Therefore, this paper summarizes the monosaccharide compositions, molecular weights, and structure-activity relationships of natural polysaccharides that regulate radiation-induced apoptosis, and also reviews the molecular mechanisms by which these polysaccharides modulate radiation-induced apoptosis, primarily focusing on promoting cancer cell apoptosis to enhance radiotherapy efficacy, reducing radiation damage to normal tissues, and inhibiting apoptosis in normal cells. Additionally, the role of gut microbiota in mediating the interaction between polysaccharides and radiation is discussed, providing innovative ideas for various radiation injuries, including hematopoiesis, immunity, and organ damage. This review will contribute to a better understanding of the value of natural polysaccharides in the field of radiation and provide guidance for the development of natural radioprotective agents and radiosensitizers.
Collapse
Affiliation(s)
- Beibei Zhang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China; Department of Nutrition, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China
| | - Mingyu Zhang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China; Department of Nutrition, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China
| | - Jinlong Tian
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| | - Xi Zhang
- Department of Nutrition, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China
| | - Dan Zhang
- Department of Nutrition, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China
| | - Jiabao Li
- Department of Nutrition, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China
| | - Lei Yang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China; Department of Nutrition, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China.
| |
Collapse
|
30
|
Wang Y, He Q, Rong K, Zhu M, Zhao X, Zheng P, Mi Y. Vitamin D3 promotes gastric cancer cell autophagy by mediating p53/AMPK/mTOR signaling. Front Pharmacol 2024; 14:1338260. [PMID: 38259281 PMCID: PMC10800859 DOI: 10.3389/fphar.2023.1338260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Objective: Vitamin D3 has the general properties of a lipid-soluble vitamin, but is also an active steroid hormone that can regulate the proliferation, apoptosis and differentiation of many tumor cells, and exerts anticancer activity against numerous malignancies. However, the mechanism underlying the effects of vitamin D3 on tumors is not fully understood. Here, we used network pharmacology and in vitro experimental approaches to explore the mechanism of vitamin D3 activity in the context of gastric cancer. Methods: The Targetnet, SuperPred, SwissTargetPrediction, and PharmMapper databases were screened for potential drug-related targets, while we used data from the PharmGKB, Drugbank, OMIM, DisGeNET, CTD, and GeneCards databases to identify potential targets associated with gastric cancer. Disease-drug crossover genes were obtained by constructing Venn diagrams. Gene ontology and Kyoto Encyclopedia of Genomes (KEGG) enrichment analyses of crossover genes were conducted and STRING was used to generate protein interaction networks and identify core targets. CCK-8 experiments were performed and apoptosis detected to assess the effect of vitamin D3 on gastric cancer cells. Western blotting was applied to detect p53/AMPK/mTOR signaling, as well as autophagy-, cell cycle-, and apoptosis-related proteins. Results: A total of 485 targets of vitamin D3 activity were obtained and 1200 gastric cancer disease-related targets discovered. Further, 60 potential targets for vitamin D3 in gastric cancer treatment were identified. KEGG analysis indicated that potential targets were mainly involved in the cell cycle, HIF-1 signaling, and the AMPK pathway, among other pathways. These findings were validated using cellular experiments, which demonstrated that the viability of AGS and SGC-7901 cells was impeded by vitamin D3. Further, vitamin D3 promoted apoptosis and inhibited the cell cycle in those cell lines, as well as activating the p53/AMPK/mTOR pathway, which promotes autophagy and inhibits tumor development. Conclusion: Our network pharmacological analyses provide preliminarily data supporting a role for vitamin D3 in promoting autophagy and apoptosis in gastric cancer cells, and in activating the p53/AMPK/mTOR pathway, which inhibits gastric cancer cell proliferation. Our findings demonstrate the molecular mechanism underlying the effect of vitamin D3 in cure of gastric cancer.
Collapse
Affiliation(s)
- Yanan Wang
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Qingmin He
- Department of Gastroenterology, Ankang Central Hospital, Ankang, Shaanxi, China
| | - Kang Rong
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingyang Zhu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoxiao Zhao
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pengyuan Zheng
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Mi
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
31
|
Zhou Y, Nakajima R, Shirasawa M, Fikriyanti M, Zhao L, Iwanaga R, Bradford AP, Kurayoshi K, Araki K, Ohtani K. Expanding Roles of the E2F-RB-p53 Pathway in Tumor Suppression. BIOLOGY 2023; 12:1511. [PMID: 38132337 PMCID: PMC10740672 DOI: 10.3390/biology12121511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
The transcription factor E2F links the RB pathway to the p53 pathway upon loss of function of pRB, thereby playing a pivotal role in the suppression of tumorigenesis. E2F fulfills a major role in cell proliferation by controlling a variety of growth-associated genes. The activity of E2F is controlled by the tumor suppressor pRB, which binds to E2F and actively suppresses target gene expression, thereby restraining cell proliferation. Signaling pathways originating from growth stimulative and growth suppressive signals converge on pRB (the RB pathway) to regulate E2F activity. In most cancers, the function of pRB is compromised by oncogenic mutations, and E2F activity is enhanced, thereby facilitating cell proliferation to promote tumorigenesis. Upon such events, E2F activates the Arf tumor suppressor gene, leading to activation of the tumor suppressor p53 to protect cells from tumorigenesis. ARF inactivates MDM2, which facilitates degradation of p53 through proteasome by ubiquitination (the p53 pathway). P53 suppresses tumorigenesis by inducing cellular senescence or apoptosis. Hence, in almost all cancers, the p53 pathway is also disabled. Here we will introduce the canonical functions of the RB-E2F-p53 pathway first and then the non-classical functions of each component, which may be relevant to cancer biology.
Collapse
Affiliation(s)
- Yaxuan Zhou
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Rinka Nakajima
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Mashiro Shirasawa
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Mariana Fikriyanti
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Lin Zhao
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Ritsuko Iwanaga
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA; (R.I.); (A.P.B.)
| | - Andrew P. Bradford
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA; (R.I.); (A.P.B.)
| | - Kenta Kurayoshi
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| | - Keigo Araki
- Department of Morphological Biology, Ohu University School of Dentistry, 31-1 Misumido Tomitamachi, Koriyama, Fukushima 963-8611, Japan;
| | - Kiyoshi Ohtani
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| |
Collapse
|
32
|
Fang Y, Bai Z, Cao J, Zhang G, Li X, Li S, Yan Y, Gao P, Kong X, Zhang Z. Low-intensity ultrasound combined with arsenic trioxide induced apoptosis of glioma via EGFR/AKT/mTOR. Life Sci 2023; 332:122103. [PMID: 37730111 DOI: 10.1016/j.lfs.2023.122103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 09/22/2023]
Abstract
AIMS This study aimed to explore whether low-intensity ultrasound (LIUS) combined with low-concentration arsenic trioxide (ATO) could inhibit the proliferation of glioma and, if so, to clarify the potential mechanism. MAIN METHODS The effects of ATO and LIUS alone or in combination on glioma were examined by CCK8, EdU, and flow cytometry assays. Western blot analysis was used to detect changes in expression of apoptosis-related proteins and their effects on the EGFR/AKT/mTOR pathway. The effects of ATO and LIUS were verified in vivo in orthotopic xenograft models, and tumor size, arsenic content in brain tissue, survival, and immunohistochemical changes were observed. KEY FINDINGS LIUS enhanced the inhibitory effect of ATO on the proliferation of glioma, and EGF reversed the proliferation inhibition and protein changes induced by ATO and LIUS. The anti-glioma effect of ATO combined with LIUS was related to downstream AKT/mTOR pathway changes caused by inhibition of EGFR activation, which enhanced apoptosis of U87MG and U373 cells. In vivo experiments showed significant increases in arsenic content in brain tissue, as well as decreased tumor sizes and longer survival times in the combined treatment group compared with other groups. The trends of immunohistochemical protein changes were consistent with the in vitro results. SIGNIFICANCE This study showed that LIUS enables ATO to exert anti-glioma effects at a safe dose by inhibiting the activation of EGFR and the downstream AKT/mTOR pathway to regulate apoptosis. LIUS in combination with ATO is a promising novel method for treating glioma and could improve patient prognosis.
Collapse
Affiliation(s)
- Yi Fang
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Zhiqun Bai
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Jibin Cao
- Department of Radiology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Gaosen Zhang
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Xiang Li
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Shufeng Li
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Yudie Yan
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Peirong Gao
- Department of Ultrasound, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiangkai Kong
- Department of Ultrasound, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Zhen Zhang
- Department of Ultrasound, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
33
|
Wang H, Yu W, Wang Y, Wu R, Dai Y, Deng Y, Wang S, Yuan J, Tan R. p53 contributes to cardiovascular diseases via mitochondria dysfunction: A new paradigm. Free Radic Biol Med 2023; 208:846-858. [PMID: 37776918 DOI: 10.1016/j.freeradbiomed.2023.09.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 10/02/2023]
Abstract
Cardiovascular diseases (CVDs) are leading causes of global mortality; however, their underlying mechanisms remain unclear. The tumor suppressor factor p53 has been extensively studied for its role in cancer and is also known to play an important role in regulating CVDs. Abnormal p53 expression levels and modifications contribute to the occurrence and development of CVDs. Additionally, mounting evidence underscores the critical involvement of mitochondrial dysfunction in CVDs. Notably, studies indicate that p53 abnormalities directly correlate with mitochondrial dysfunction and may even interact with each other. Encouragingly, small molecule inhibitors targeting p53 have exhibited remarkable effects in animal models of CVDs. Moreover, therapeutic strategies aimed at mitochondrial-related molecules and mitochondrial replacement therapy have demonstrated their advantageous potential. Therefore, targeting p53 or mitochondria holds immense promise as a pioneering therapeutic approach for combating CVDs. In this comprehensive review, we delve into the mechanisms how p53 influences mitochondrial dysfunction, including energy metabolism, mitochondrial oxidative stress, mitochondria-induced apoptosis, mitochondrial autophagy, and mitochondrial dynamics, in various CVDs. Furthermore, we summarize and discuss the potential significance of targeting p53 or mitochondria in the treatment of CVDs.
Collapse
Affiliation(s)
- Hao Wang
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, China
| | - Wei Yu
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yibo Wang
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, China
| | - Ruihao Wu
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yifei Dai
- School of Stomatology, Xuzhou Medical University, Xuzhou, 221004, China
| | - Ye Deng
- School of Stomatology, Xuzhou Medical University, Xuzhou, 221004, China
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China.
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining, 272000, China.
| | - Rubin Tan
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou, 221004, China.
| |
Collapse
|
34
|
Rius-Pérez S. p53 at the crossroad between mitochondrial reactive oxygen species and necroptosis. Free Radic Biol Med 2023; 207:183-193. [PMID: 37481144 DOI: 10.1016/j.freeradbiomed.2023.07.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/10/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
p53 is a redox-sensitive transcription factor that can regulate multiple cell death programs through different signaling pathways. In this review, we assess the role of p53 in the regulation of necroptosis, a programmed form of lytic cell death highly involved in the pathophysiology of multiple diseases. In particular, we focus on the role of mitochondrial reactive oxygen species (mtROS) as essential contributors to modulate necroptosis execution through p53. The enhanced generation of mtROS during necroptosis is critical for the correct interaction between receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and 3 (RIPK3), two key components of the functional necrosome. p53 controls the occurrence of necroptosis by modulating the levels of mitochondrial H2O2 via peroxiredoxin 3 and sulfiredoxin. Furthermore, in response to increased levels of H2O2, p53 upregulates the long non-coding RNA necrosis-related factor, favoring the translation of RIPK1 and RIPK3. In parallel, a fraction of cytosolic p53 migrates into mitochondria, a process notably involved in necroptosis execution via its interaction with the mitochondrial permeability transition pore. In conclusion, p53 is located at the intersection between mtROS and the necroptosis machinery, making it a key protein to orchestrate redox signaling during necroptosis.
Collapse
Affiliation(s)
- Sergio Rius-Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjasot, 46100, Valencia, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain.
| |
Collapse
|
35
|
Wu R, Zhao J, Wei P, Tang M, Ma Z, Zhao Y, Du L, Wan L. Piper nigrum Extract Inhibits the Growth of Human Colorectal Cancer HT-29 Cells by Inducing p53-Mediated Apoptosis. Pharmaceuticals (Basel) 2023; 16:1325. [PMID: 37765133 PMCID: PMC10537627 DOI: 10.3390/ph16091325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Colorectal cancer (CRC) is a prevalent malignancy of the digestive tract with the second highest mortality rate globally. Piper nigrum is a widely used traditional medicinal plant, exhibiting antitumor activity against various tumor cells. At present, research on the effect of Piper nigrum on CRC is limited to in vitro cytotoxicity, lacking comprehensive mechanism investigations. This study aimed to explore the inhibitory effect and mechanism of Piper nigrum extract (PNE) on HT-29 cells. Firstly, we identified the chemical components of PNE. Then, MTT assay, colony formation assay, JC-1 staining, and flow cytometry were used to analyze the effect of PNE on HT-29 cells in vitro. A xenograft model, histopathological examination, immunohistochemistry, and western blot were used to evaluate the tumor growth inhibitory activity and mechanism of PNE in vivo. The results indicated that PNE could inhibit cell proliferation and colony formation, reduce mitochondrial membrane potential, induce cell apoptosis in vitro, and inhibit tumor growth in vivo. Furthermore, PNE could regulate p53 and its downstream proteins, and subsequently activate the caspase-3 pathway. In summary, PNE probably induced apoptosis of HT-29 cells through the mitochondrial pathway mediated by p53. All these results suggested that PNE might be a potential natural-origin anti-CRC drug candidate.
Collapse
Affiliation(s)
- Rui Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (R.W.); (J.Z.); (P.W.); (Y.Z.); (L.D.)
| | - Jiajia Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (R.W.); (J.Z.); (P.W.); (Y.Z.); (L.D.)
| | - Panhong Wei
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (R.W.); (J.Z.); (P.W.); (Y.Z.); (L.D.)
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China; (M.T.); (Z.M.)
| | - Ziyan Ma
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China; (M.T.); (Z.M.)
| | - Yunyan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (R.W.); (J.Z.); (P.W.); (Y.Z.); (L.D.)
| | - Leilei Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (R.W.); (J.Z.); (P.W.); (Y.Z.); (L.D.)
| | - Li Wan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (R.W.); (J.Z.); (P.W.); (Y.Z.); (L.D.)
| |
Collapse
|
36
|
Harakeh S, Al-Raddadi R, Alamri T, Al-Jaouni S, Qari M, Qari Y, Kumar A, Tashkandi HM, Moulay M, Aldahlawi AM, Slama P, Haque S. Apoptosis induction in human hepatoma cell line HepG2 cells by trans- Anethole via activation of mitochondria-mediated apoptotic pathways. Biomed Pharmacother 2023; 165:115236. [PMID: 37531780 DOI: 10.1016/j.biopha.2023.115236] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023] Open
Abstract
trans-Anethole a valuable compound derived from star anise widely used by ethnic tribals to manage numerous human diseases. In this study antiproliferative activities of trans-Anethole towards human liver cancer (HepG2), cervical cancer (HeLa) and breast cancer (MCF-7) cells were explored. trans-Anethole showed free radical scavenging potential as assessed by DNA nicking assay. trans-Anethole exhibited strong antiproliferative potential towards HepG2 cells compared to other cell lines. trans-Anethole strongly induced apoptosis in HepG2 cells by significantly upregulating the protein expressions of p53, Caspase-3 and Caspase-9 were assessed by western blotting analysis which highlighted apoptosis-inducing capacity of trans-Anethole against HepG2 cells. Rt-qPCR analysis revealed that trans- Anethole upregulated p53, caspase - 3 and - 9 in comparison to untreated HepG2 cancer cells. Moreover, trans-Anethole provoked the generation of ROS and disruption of MMP. Our research suggests that trans-Anethole may have a significant anticancer therapeutic potential for treating liver cancer.
Collapse
Affiliation(s)
- Steve Harakeh
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Yousef Abdul Latif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rajaa Al-Raddadi
- Department of Community Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Turki Alamri
- Family and Community Medicine Department, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Soad Al-Jaouni
- Yousef Abdul Latif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Department of Hematology/ Pediatric Oncology, King Abdulaziz University Hospital, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammed Qari
- Yousef Abdul Latif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Department of Hematology/ Pediatric Oncology, King Abdulaziz University Hospital, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Yousef Qari
- Department of Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ajay Kumar
- University Centre for Research and Development, Biotechnology Engineering & Food Technology, Chandigarh University, Mohali 140413, Punjab, India
| | - Hanaa M Tashkandi
- Department of General Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammed Moulay
- Embryonic Stem Cell Research Unit, King Fahd Medical Research Center, and Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Alia M Aldahlawi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Petr Slama
- Laboratory of Animal Immunology and Biotechnology, Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, 61300 Brno, Czech Republic
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut 11022801, Lebanon; Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 13306, United Arab Emirates.
| |
Collapse
|
37
|
Deng Y, Jin H, Ning J, Cui D, Zhang M, Yang H. Elevated galectin-3 levels detected in women with hyperglycemia during early and mid-pregnancy antagonizes high glucose - induced trophoblast cells apoptosis via galectin-3/foxc1 pathway. Mol Med 2023; 29:115. [PMID: 37626284 PMCID: PMC10463409 DOI: 10.1186/s10020-023-00707-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
OBJECTIVE This study was to evaluate plasma galectin-3 levels from early pregnancy to delivery and explore the effects of galectin-3 on the function of trophoblast cells under high glucose exposure. METHODS The plasma galectin-3 levels were quantified by enzyme-linked immunosorbent assay (ELISA) in the China National Birth Cohort (CNBC) at Peking University First Hospital, and the underlying signaling pathway was identified by protein-protein interaction (PPI) analysis, gene set enrichment analysis (GSEA), quantitative PCR (qPCR), western blotting, small interfering RNA (siRNA) transfections, and flow cytometry. RESULTS Significantly higher galectin-3 levels were found in patients with gestational diabetes mellitus (GDM group; n = 77) during the first and second trimesters than that in healthy pregnant women (HP group; n = 113) (P < 0.05). No significant differences in plasma galectin-3 levels were detected between GDM and HP groups in maternal third-trimester blood and cord blood. PPI analysis suggested potential interactions between galectin-3 and foxc1. The findings of GSEA showed that galectin-3 was involved in the cytochrome P450-related and complement-related pathways, and foxc1 was associated with type I diabetes mellitus. Additionally, high glucose (25 mM) significantly increased the expression levels of galectin-3 and foxc1 and induced apoptosis in HTR-8/SVneo cells. Further in vitro experiments showed that galectin-3/foxc1 pathway could protect HTR-8/SVneo cells against high glucose - induced apoptosis. CONCLUSION Future studies were required to validate whether plasma galectin-3 might become a potential biomarker for hyperglycemia during pregnancy. Elevated galectin-3 levels might be a vital protective mechanism among those exposed to hyperglycemia during pregnancy.
Collapse
Affiliation(s)
- Yu Deng
- Department of Obstetrics and Gynecology, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, 100034, China
| | - Hongyan Jin
- Department of Obstetrics and Gynecology, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
| | - Jie Ning
- Department of Obstetrics and Gynecology, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, 100034, China
| | - Dong Cui
- Department of Obstetrics and Gynecology, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, 100034, China
| | - Muqiu Zhang
- Department of Obstetrics and Gynecology, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, 100034, China
| | - Huixia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, No. 8 Xishiku Street, Beijing, 100034, China.
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, 100034, China.
| |
Collapse
|
38
|
Guo L, Xia Y, Li H, Wang Z, Xu H, Dai X, Zhang Y, Zhang H, Fan W, Wei F, Li Q, Zhang L, Cao L, Zhang S, Hu W, Gu H. FIT links c-Myc and P53 acetylation by recruiting RBBP7 during colorectal carcinogenesis. Cancer Gene Ther 2023; 30:1124-1133. [PMID: 37225855 DOI: 10.1038/s41417-023-00624-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/07/2023] [Accepted: 05/04/2023] [Indexed: 05/26/2023]
Abstract
Colorectal cancer (CRC) poses one of the most serious threats to human health worldwide, and abnormally expressed c-Myc and p53 are deemed the pivotal driving forces of CRC progression. In this study, we discovered that the lncRNA FIT, which was downregulated in CRC clinical samples, was transcriptionally suppressed by c-Myc in vitro and promoted CRC cell apoptosis by inducing FAS expression. FAS is a p53 target gene, and we found that FIT formed a trimer with RBBP7 and p53 that facilitated p53 acetylation and p53-mediated FAS gene transcription. Moreover, FIT was capable of retarding CRC growth in a mouse xenograft model, and FIT expression was positively correlated with FAS expression in clinical samples. Thus, our study elucidates the role of the lncRNA FIT in human colorectal cancer growth and provides a potential target for anti-CRC drugs.
Collapse
Affiliation(s)
- Lili Guo
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yang Xia
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Hao Li
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Zifei Wang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Hui Xu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xiangyu Dai
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yaqin Zhang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Hao Zhang
- Department of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenhu Fan
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Feng Wei
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Qun Li
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Ling Zhang
- Department of Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Limian Cao
- Department of Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shangxin Zhang
- Department of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Wanglai Hu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou University, Zhengzhou, China.
| | - Hao Gu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
39
|
Wei H, Wang H, Wang G, Qu L, Jiang L, Dai S, Chen X, Zhang Y, Chen Z, Li Y, Guo M, Chen Y. Structures of p53/BCL-2 complex suggest a mechanism for p53 to antagonize BCL-2 activity. Nat Commun 2023; 14:4300. [PMID: 37463921 DOI: 10.1038/s41467-023-40087-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 07/11/2023] [Indexed: 07/20/2023] Open
Abstract
Mitochondrial apoptosis is strictly controlled by BCL-2 family proteins through a subtle network of protein interactions. The tumor suppressor protein p53 triggers transcription-independent apoptosis through direct interactions with BCL-2 family proteins, but the molecular mechanism is not well understood. In this study, we present three crystal structures of p53-DBD in complex with the anti-apoptotic protein BCL-2 at resolutions of 2.3-2.7 Å. The structures show that two loops of p53-DBD penetrate directly into the BH3-binding pocket of BCL-2. Structure-based mutations at the interface impair the p53/BCL-2 interaction. Specifically, the binding sites for p53 and the pro-apoptotic protein Bax in the BCL-2 pocket are mostly identical. In addition, formation of the p53/BCL-2 complex is negatively correlated with the formation of BCL-2 complexes with pro-apoptotic BCL-2 family members. Defects in the p53/BCL-2 interaction attenuate p53-mediated cell apoptosis. Overall, our study provides a structural basis for the interaction between p53 and BCL-2, and suggests a molecular mechanism by which p53 regulates transcription-independent apoptosis by antagonizing the interaction of BCL-2 with pro-apoptotic BCL-2 family members.
Collapse
Affiliation(s)
- Hudie Wei
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratroy for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Haolan Wang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratroy for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Genxin Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Lingzhi Qu
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratroy for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Longying Jiang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratroy for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuyan Dai
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratroy for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xiaojuan Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratroy for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Ye Zhang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratroy for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhuchu Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratroy for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Youjun Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Ming Guo
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratroy for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratroy for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
40
|
Choe JH, Kawase T, Xu A, Guzman A, Obradovic AZ, Low-Calle AM, Alaghebandan B, Raghavan A, Long K, Hwang PM, Schiffman JD, Zhu Y, Zhao R, Lee DF, Katz C, Prives C. Li-Fraumeni Syndrome-Associated Dimer-Forming Mutant p53 Promotes Transactivation-Independent Mitochondrial Cell Death. Cancer Discov 2023; 13:1250-1273. [PMID: 37067901 PMCID: PMC10287063 DOI: 10.1158/2159-8290.cd-22-0882] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/11/2023] [Accepted: 03/02/2023] [Indexed: 04/18/2023]
Abstract
Cancer-relevant mutations in the oligomerization domain (OD) of the p53 tumor suppressor protein, unlike those in the DNA binding domain, have not been well elucidated. Here, we characterized the germline OD mutant p53(A347D), which occurs in cancer-prone Li-Fraumeni syndrome (LFS) patients. Unlike wild-type p53, mutant p53(A347D) cannot form tetramers and exists as a hyperstable dimeric protein. Further, p53(A347D) cannot bind or transactivate the majority of canonical p53 target genes. Isogenic cell lines harboring either p53(A347D) or no p53 yield comparable tumorigenic properties, yet p53(A347D) displays remarkable neomorphic activities. Cells bearing p53(A347D) possess a distinct transcriptional profile and undergo metabolic reprogramming. Further, p53(A347D) induces striking mitochondrial network aberration and associates with mitochondria to drive apoptotic cell death upon topoisomerase II inhibition in the absence of transcription. Thus, dimer-forming p53 demonstrates both loss-of-function (LOF) and gain-of-function (GOF) properties compared with the wild-type form of the protein. SIGNIFICANCE A mutant p53 (A347D), which can only form dimers, is associated with increased cancer susceptibility in LFS individuals. We found that this mutant wields a double-edged sword, driving tumorigenesis through LOF while gaining enhanced apoptogenic activity as a new GOF, thereby yielding a potential vulnerability to select therapeutic approaches. See related commentary by Stieg et al., p. 1046. See related article by Gencel-Augusto et al., p. 1230. This article is highlighted in the In This Issue feature, p. 1027.
Collapse
Affiliation(s)
- Joshua H. Choe
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Tatsuya Kawase
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
- Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - An Xu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Asja Guzman
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Aleksandar Z. Obradovic
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York 10032, USA
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Ana Maria Low-Calle
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Bita Alaghebandan
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Ananya Raghavan
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Kaitlin Long
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Paul M. Hwang
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Joshua D. Schiffman
- Department of Pediatrics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
- Peel Therapeutics, Inc., Salt Lake City, UT 84112, USA
| | - Yan Zhu
- Department of Biological Sciences, St. John’s University, New York, NY 11439, USA
| | - Ruiying Zhao
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Dung-Fang Lee
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Chen Katz
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| |
Collapse
|
41
|
Cai YJ, He JY, Yang XY, Huang W, Fu XM, Guo SQ, Yang JJ, Dong JD, Zeng HT, Wu YJ, Qin Z, Qin QW, Sun HY. Molecular characterization, expression and function analysis of Epinephelus coioides PKC-ɑ response to Singapore grouper iridovirus (SGIV) infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 142:104646. [PMID: 36702214 DOI: 10.1016/j.dci.2023.104646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/28/2022] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
Protein kinase C (PKC) constitutes the main signal transduction pathway, and participates in the signal pathway of cell proliferation and movement in mammals. In this study, PKC-ɑ was obtained from Epinephelus coioides, an important marine fish cultivated in the coastal areas of southern China and Southeast Asia. The full length cDNA of PKC-ɑ was 3362 bp in length containing a 23 bp 5'UTR, a 1719 bp 3'UTR, and a 1620 bp open reading frame encoding 539 amino acids. It contains three conservative domains including protein kinase C conserved region 2 (C2), Serine/Threonine protein kinases, catalytic domain (S_TKc) and ser/thr-type protein kinases (S_TK_X). Its mRNA can be detected in all 11 tissues examined of E. coioides, and the expression was significantly upregulated response to Singapore grouper iridovirus (SGIV) infection, one of the important pathogens of marine fish. Upregulated E. coioides PKC-ɑ significantly inhibited the activation of nuclear factor kappa-B (NF-κB) and activator protein-1 (AP-1), and SGIV-induced cell apoptosis. The results indicated that the PKC-ɑ may play an important role in pathogenic stimulation.
Collapse
Affiliation(s)
- Yi-Jie Cai
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Jia-Yang He
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Xin-Yue Yang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Wei Huang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Xue-Mei Fu
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Shi-Qing Guo
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Jie-Jia Yang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Jun-De Dong
- Guangdong Provincial Key Laboratory of Applied Marine Biology, 510301, PR China
| | - Hai-Tian Zeng
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Yan-Jun Wu
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Zhou Qin
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Qi-Wei Qin
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China; Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, 519000, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266000, PR China.
| | - Hong-Yan Sun
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China.
| |
Collapse
|
42
|
Zhou Y, Fu R, Yang M, Liu W, Tong Z. Lycopene suppresses gastric cancer cell growth without affecting normal gastric epithelial cells. J Nutr Biochem 2023; 116:109313. [PMID: 36871837 DOI: 10.1016/j.jnutbio.2023.109313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/07/2023]
Abstract
Gastric cancer is one of the leading causes of cancer-related death worldwide. Lycopene, a natural carotenoid, has potent antioxidant activity and anti-cancer effects against several types of cancers. However, the mechanism for the anti-gastric cancer effects of lycopene remains to be fully clarified. Normal gastric epithelial cell line GES-1 and gastric cancer cell line AGS, SGC-7901, Hs746T cells were treated with different concentrations of lycopene and the effects of lycopene were compared. Lycopene specifically suppressed cell growth monitored by Real-Time Cell Analyzer, induced cell cycle arrest and cell apoptosis detected by flow cytometry, and lowered mitochondrial membrane potentials assessed by JC-1 staining of AGS and SGC-7901 cells, while did not affect those of GES-1 cells. Lycopene did not affect the cell growth of Hs746T cells harboring TP53 mutation. Further bioinformatics analysis predicted 57 genes with up-regulated expression levels in gastric cancer and decreased function in cells after lycopene treatment. Quantitative PCR and Western Blot were used to check the critical factors in the cell cycle and apoptosis signaling pathway. Lycopene decreased the high expression levels of CCNE1 and increased the levels of TP53 in AGS and SGC-7901 cells without affecting those in GES-1 cells. In summary, lycopene could effectively suppress gastric cancer cells with CCNE1-amplification, which could be a promising target therapy reagent for gastric cancer.
Collapse
Affiliation(s)
- Ying Zhou
- TaiKang Medical School (School of Basic Medicine Sciences), Wuhan University, Wuhan, China
| | - Rishun Fu
- TaiKang Medical School (School of Basic Medicine Sciences), Wuhan University, Wuhan, China
| | - Mei Yang
- TaiKang Medical School (School of Basic Medicine Sciences), Wuhan University, Wuhan, China
| | - Weihuang Liu
- TaiKang Medical School (School of Basic Medicine Sciences), Wuhan University, Wuhan, China
| | - Zan Tong
- TaiKang Medical School (School of Basic Medicine Sciences), Wuhan University, Wuhan, China.
| |
Collapse
|
43
|
Wang H, Guo M, Wei H, Chen Y. Targeting p53 pathways: mechanisms, structures, and advances in therapy. Signal Transduct Target Ther 2023; 8:92. [PMID: 36859359 PMCID: PMC9977964 DOI: 10.1038/s41392-023-01347-1] [Citation(s) in RCA: 323] [Impact Index Per Article: 161.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/19/2022] [Accepted: 02/07/2023] [Indexed: 03/03/2023] Open
Abstract
The TP53 tumor suppressor is the most frequently altered gene in human cancers, and has been a major focus of oncology research. The p53 protein is a transcription factor that can activate the expression of multiple target genes and plays critical roles in regulating cell cycle, apoptosis, and genomic stability, and is widely regarded as the "guardian of the genome". Accumulating evidence has shown that p53 also regulates cell metabolism, ferroptosis, tumor microenvironment, autophagy and so on, all of which contribute to tumor suppression. Mutations in TP53 not only impair its tumor suppressor function, but also confer oncogenic properties to p53 mutants. Since p53 is mutated and inactivated in most malignant tumors, it has been a very attractive target for developing new anti-cancer drugs. However, until recently, p53 was considered an "undruggable" target and little progress has been made with p53-targeted therapies. Here, we provide a systematic review of the diverse molecular mechanisms of the p53 signaling pathway and how TP53 mutations impact tumor progression. We also discuss key structural features of the p53 protein and its inactivation by oncogenic mutations. In addition, we review the efforts that have been made in p53-targeted therapies, and discuss the challenges that have been encountered in clinical development.
Collapse
Affiliation(s)
- Haolan Wang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Ming Guo
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hudie Wei
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
44
|
Yang Y, Guo FF, Chen CF, Li YL, Liang H, Chen ZF. Antitumor activity of synthetic three copper(II) complexes with terpyridine ligands. J Inorg Biochem 2023; 240:112093. [PMID: 36525715 DOI: 10.1016/j.jinorgbio.2022.112093] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 12/03/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
Three new synthetic terpyridine copper(II) complexes were characterized. The copper(II) complexes induced apoptosis of three cancer cell lines and arrested T-24 cell cycle in G1 phase. The complexes were accumulated in mitochondria of T-24 cells and caused significant reduction of the mitochondrial membrane potential. The complexes increased both intracellular ROS and Ca2+ levels and activated the caspase-3/9 expression. The apoptosis was further confirmed by Western Blotting analysis. Bcl-2 was down-regulated and Bax was upregulated after treatment with complexes 1-3. The in vivo studies showed that complexes 1-3 obviously inhibited the growth of tumor without significant toxicity to other organs.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China; Department of Chemistry and Pharmacy, Guilin Normal College, Guilin 541199, China
| | - Fei-Fei Guo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Cai-Feng Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Yu-Lan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Zhen-Feng Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| |
Collapse
|
45
|
Fan CW, Li MS, Song XX, Luo L, Jiang JC, Luo JZ, Wang HS. Discovery of novel 2-oximino-2-indolylacetamide derivatives as potent anticancer agents capable of inducing cell autophagy and ferroptosis. Bioorg Med Chem 2023; 80:117176. [PMID: 36709571 DOI: 10.1016/j.bmc.2023.117176] [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: 12/01/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 01/21/2023]
Abstract
A series of 2-oximino-2-indolylacetamide derivatives were designed, synthesized and evaluated for their antitumour effects. Among them, 4d exhibited the most potent antiproliferative effect in vitro on the tested human cancer cells. Additionally, 4d significantly induced cell apoptosis, caused mitochondrial dysfunction, promoted Bax, cleaved-PARP and p53 expression and inhibited Bcl-2 expression in 5-8F cells. Moreover, 4d remarkably promoted autophagosome formation, leading to cell apoptosis. Further investigation indicated that 4d could trigger cell death through cell ferroptosis, including increased ROS generation and lipid peroxidation and decreased glutathione peroxidase 4 (GPx4) expression and glutathione (GSH) levels. More importantly, 4d induced 5-8F cell death by activating ROS/MAPK and inhibiting the AKT/mTOR and STAT3 signalling pathways. Interestingly, 4d significantly suppressed tumour growth in a 5-8F cell xenograft model without obvious toxicity to mice. Overall, these results demonstrate that 4d may be a potential compound for cancer therapy.
Collapse
Affiliation(s)
- Cai-Wen Fan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China; Scientific Experiment Center, Guilin Medical University, Guilin 541199, China
| | - Mei-Shan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Xi-Xi Song
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Li Luo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Jing-Chen Jiang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Jia-Zi Luo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Heng-Shan Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| |
Collapse
|
46
|
Hu J, Jiang W, Zuo J, Shi D, Chen X, Yang X, Zhang W, Ma L, Liu Z, Xing Q. Structural basis of bacterial effector protein azurin targeting tumor suppressor p53 and inhibiting its ubiquitination. Commun Biol 2023; 6:59. [PMID: 36650277 PMCID: PMC9845241 DOI: 10.1038/s42003-023-04458-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
Tumor suppressor p53 prevents tumorigenesis by promoting cell cycle arrest and apoptosis through transcriptional regulation. Dysfunction of p53 occurs frequently in human cancers. Thus, p53 becomes one of the most promising targets for anticancer treatment. A bacterial effector protein azurin triggers tumor suppression by stabilizing p53 and elevating its basal level. However, the structural and mechanistic basis of azurin-mediated tumor suppression remains elusive. Here we report the atomic details of azurin-mediated p53 stabilization by combining X-ray crystallography with nuclear magnetic resonance. Structural and mutagenic analysis reveals that the p28 region of azurin, which corresponds to a therapeutic peptide, significantly contributes to p53 binding. This binding stabilizes p53 by disrupting COP1-mediated p53 ubiquitination and degradation. Using the structure-based design, we obtain several affinity-enhancing mutants that enable amplifying the effect of azurin-induced apoptosis. Our findings highlight how the structure of the azurin-p53 complex can be leveraged to design azurin derivatives for cancer therapy.
Collapse
Affiliation(s)
- Jianjian Hu
- grid.35155.370000 0004 1790 4137National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070 China
| | - Wenxue Jiang
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| | - Jiaqi Zuo
- grid.35155.370000 0004 1790 4137National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070 China
| | - Dujuan Shi
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| | - Xiaoqi Chen
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| | - Xiao Yang
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| | - Wenhui Zhang
- grid.35155.370000 0004 1790 4137National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070 China
| | - Lixin Ma
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| | - Zhu Liu
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| | - Qiong Xing
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| |
Collapse
|
47
|
Wallis B, Bowman KR, Lu P, Lim CS. The Challenges and Prospects of p53-Based Therapies in Ovarian Cancer. Biomolecules 2023; 13:159. [PMID: 36671544 PMCID: PMC9855757 DOI: 10.3390/biom13010159] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/07/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
It has been well established that mutations in the tumor suppressor gene, p53, occur readily in a vast majority of cancer tumors, including ovarian cancer. Typically diagnosed in stages three or four, ovarian cancer is the fifth leading cause of death in women, despite accounting for only 2.5% of all female malignancies. The overall 5-year survival rate for ovarian cancer is around 47%; however, this drops to an abysmal 29% for the most common type of ovarian cancer, high-grade serous ovarian carcinoma (HGSOC). HGSOC has upwards of 96% of cases expressing mutations in p53. Therefore, wild-type (WT) p53 and p53-based therapies have been explored as treatment options via a plethora of drug delivery vehicles including nanoparticles, viruses, polymers, and liposomes. However, previous p53 therapeutics have faced many challenges, which have resulted in their limited translational success to date. This review highlights a selection of these historical p53-targeted therapeutics for ovarian cancer, why they failed, and what the future could hold for a new generation of this class of therapies.
Collapse
Affiliation(s)
| | | | | | - Carol S. Lim
- Department of Molecular Pharmaceutics, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
48
|
Wang D, Wang Q, Zuo Z, Dong Z, He J, Ye X, Tang H, Zou J. Koumine induces apoptosis in Cyprinus carpio liver cells by regulating JAK-STAT and p53 signaling pathways. FISH & SHELLFISH IMMUNOLOGY 2023; 132:108475. [PMID: 36496140 DOI: 10.1016/j.fsi.2022.108475] [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: 05/14/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 06/17/2023]
Abstract
Koumine is an alkaloid with significant anti-anxiety, anticancer cell proliferation, and analgesic activities, and our previous studies have shown that koumine can be used as an immunostimulant in aquaculture, but the molecular mechanism of its effect remains unclear. We fed a basal diet with 0, 0.2, 2, and 20 mg/kg koumine to C. carpio for 10 weeks, and comprehensive studies of the histological and biochemical parameters and transcriptomes of the four groups were performed. Histological results indicated that the number of apoptotic cells in the liver increased with increasing koumine concentration. Compared with those of the control group, the malondialdehyde, superoxide dismutase, catalase, acid phosphatase, alkaline phosphatase, and lactate dehydrogenase levels of the treatment group increased to varying degrees. In total, 100.11 GB of clean data, 4774 DEGs, and 138 differentially expressed genes were obtained from the transcriptome data. Differentially expressed genes were classified into 187 signalling pathways, and the circadian rhythm signalling pathway, the JAK-STAT signalling pathway, the p53 signalling pathway and the PPAR signalling pathway were the top enriched pathways. The qRT-PCR results confirmed that the key genes ifnar1, socs3l, epoa, ghra, cMyc, mcl-1, shisa4, and gtse1 involved in balancing cell proliferation and apoptosis were enriched in these pathways. We discovered that the JAK-STAT and p53 pathways are important targets of koumine. Such information contributes to a better understanding of the potential mechanism by which koumine regulates hepatic immunity as well as to lays the theoretical foundation for its application.
Collapse
Affiliation(s)
- Dongjie Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Qiujie Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Zhiheng Zuo
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Zaijie Dong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China.
| | - Jiayang He
- College of Environmental Science and Engineering, Guilin University of Technology, Guilin, 541004, China
| | - Xiangchen Ye
- Aquatic Species Introduction and Breeding Centre of Guangxi Zhuang Autonomous Region, Nanning, 530000, China
| | - Huijuan Tang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jixing Zou
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| |
Collapse
|
49
|
Zhan G, Liu J, Lin J, Chen J, Sun S, Maimaitiyiming Y, Hsu CH. Multifaceted Functions of RNA m6A Modification in Modulating Regulated Cell Death. RNA TECHNOLOGIES 2023:539-573. [DOI: 10.1007/978-3-031-36390-0_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
50
|
Zhang Y, Lu H, Ji H, Li Y. p53 upregulated by HIF-1α promotes gastric mucosal epithelial cells apoptosis in portal hypertensive gastropathy. Dig Liver Dis 2023; 55:81-92. [PMID: 35780066 DOI: 10.1016/j.dld.2022.06.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Portal hypertensive gastropathy (PHG) is a serious complication of liver cirrhosis and a potential cause of gastrointestinal bleeding. Mucosal apoptosis is an essential pathological feature of PHG. However, whether HIF-1α and p53 are involved in mucosal apoptosis and whether HIF-1α induces PHG by mediating p53 remains unclear. METHODS Gastric mucosal injury and apoptosis were examined in PHG patients and animal models. The mechanisms of HIF-1α- and p53-mediated apoptosis were analyzed. The GES-1 cell line was used to elucidate the underlying mechanisms using siRNA knockdown of HIF-1α and p53 in a hypoxic environment in vitro. RESULTS Epithelial apoptosis, HIF-1α, and p53 were markedly induced in the gastric mucosa of PHG. Apoptosis was attenuated in mice with HIF-1α- and p53-specific inhibitors. Apoptotic signaling factors were markedly induced in the gastric mucosa of PHG. Inhibition of p53 demonstrably attenuated the mucosal apoptosis; however, it did not affect HIF-1α expression. Conversely, targeted deletion of HIF-1α significantly inhibited p53 expression and attenuated the injury and p53-mediated apoptosis. Bax and Bcl-2 expression can be upregulated and downregulated by p53, respectively, to increasecleaved caspase-3 expression, which can be regulated by HIF-1α. CONCLUSIONS These results indicate that HIF-1α regulates the p53-induced mucosal epithelial apoptotic signaling pathway and that HIF-1α and p53 are potential therapeutic targets for PHG.
Collapse
Affiliation(s)
- Yafei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Hongwei Lu
- Department of General Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hong Ji
- Department of General Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yiming Li
- Department of General Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|