1
|
Zhang C, Wang D, Shen Y, Zhang Y, Liu J. Nuclear translocation of CDK5RAP3 regulated by NXF3 promotes the progression of gastric cancer. Cell Mol Life Sci 2025; 82:100. [PMID: 40032765 PMCID: PMC11876495 DOI: 10.1007/s00018-025-05630-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Nuclear-cytoplasmic transport proteins (NCTPs) impact the transport of proteins and RNA molecules between the nucleus and cytoplasm in tumor cells, making them promising targets for cancer therapy. Currently, the molecular mechanism and function of Nuclear RNA export factor 3 (NXF3) in gastric cancer (GC) remains unclear. METHODS We used Univariate Cox regression analysis and LASSO regression analysis, Receiver Operating Characteristic (ROC) curves to construct and evaluate a NCTP prognosis risk scoring model (NCTP model). Moreover, we identified the key NCTP (NXF3) affecting GC through differential expression and prognosis analysis. Subsequently, we introduced NXF3 shRNA into GC cells to investigate the impact of NXF3 on the cell proliferation, cell migration, invasion, and cell cycle and apoptosis and tumor growth by CCK-8 assay, transwell, wound healing assay, Flow cytometry, and nude mice subcutaneous tumor in vitro and in vivo. Furthermore, we investigated the key molecules influenced by NXF3 through piRNA-Seq, RNA-Seq, RIP-Seq, IP-MS, and Nuclear-cytoplasmic transcriptomics. RESULTS We constructed a prognostic risk model related to 3 NCTPs, including NXF3, GLE1 and RANGAP. The NCTP model effectively predicts the prognosis of GC patients. The low-risk group exhibited a significantly higher overall survival rate than that of the high-risk group. Notably, NXF3 is identified as a crucial NCTP in GC, and its high expression is associated with poor prognosis of GC patients. Knocking down of NXF3 significantly inhibited the proliferation, invasion, migration, cell cycle, tumor growth and induced cell apoptosis of GC cells in vitro and in vivo. Mechanistically, NXF3 modulates the cell cycle, cellular senescence related oncogenic pathways via piRNA-target network. Specifically, our findings highlighted several piRNA-related signaling pathways in GC, such as piRNA_3457319-CCND1/CDKN1A-p53, piRNA_2847077-TGFB3/TGFBR2-Cellular senescence, piRNA_448895-IGF1/PDGFRA/ACTB/MAP2K6-Rap1. Moreover, NXF3 was shown to facilitate the nuclear export of CDK5RAP3 mRNA, thereby promoting cell cycle progression and increasing cancer cell proliferation in gastric cancer. CONCLUSION Our study demonstrates that NXF3 modulates cell cycle progression and promotes gastric cancer development through piRNA-related pathways and the nuclear export of CDK5RAP3 mRNA. Targeting NXF3 represents a promising strategy for developing novel therapeutic approaches for gastric cancer.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated, Shanghai Jiaotong University School of Medicine, No.160, Pujian Road, Pudong New Area, Shanghai, 200127, China
| | - Dongyang Wang
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated, Shanghai Jiaotong University School of Medicine, No.160, Pujian Road, Pudong New Area, Shanghai, 200127, China
| | - Yuguang Shen
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated, Shanghai Jiaotong University School of Medicine, No.160, Pujian Road, Pudong New Area, Shanghai, 200127, China
| | - Yuanruohan Zhang
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated, Shanghai Jiaotong University School of Medicine, No.160, Pujian Road, Pudong New Area, Shanghai, 200127, China
| | - Jiahua Liu
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated, Shanghai Jiaotong University School of Medicine, No.160, Pujian Road, Pudong New Area, Shanghai, 200127, China.
| |
Collapse
|
2
|
Yan F, Guo Q, Zheng R, Ying J. Predictive performance of a centrosome-associated prognostic model in prognosis and immunotherapy of lung adenocarcinoma. Anal Biochem 2025; 698:115731. [PMID: 39617159 DOI: 10.1016/j.ab.2024.115731] [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: 07/26/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/11/2024]
Abstract
In recent years, mounting investigations have highlighted the pivotal role of centrosomes in cancer progression. In this study, we employed bioinformatics and statistics to establish a 13-centrosome-associated gene prognostic model for lung adenocarcinoma (LUAD) utilizing transcriptomic data from TCGA. Based on the Riskscore, patients were stratified into high- and low-risk groups. Through survival analysis and receiver operating characteristic curve analysis, our model demonstrated a consistent and robust prognostic capacity, which was further validated using the GEO database. Univariate/multivariate Cox regression analyses identified our model as an independent prognostic factor for LUAD patients. Subsequently, immunoinfiltration analysis showed that immune cell infiltration levels of aDCs, iDCs, Mast cells, and Neutrophils, as well as immune functionalities such as HLA, Type I IFN Response and Type II IFN Response, were markedly reduced in the high-risk group compared to the low-risk group. Finally, we conducted a drug screening to identify potential treatments for patients with different prognoses. We utilized the GDSC database and molecular docking techniques to identify small molecule compounds targeting the prognostic genes. In conclusion, our prognostic model exhibits robust and reliable predictive capability, and it may have important clinical implications in guiding treatment decisions for LUAD patients.
Collapse
Affiliation(s)
- Feng Yan
- Department of Medical Oncology, The First People's Hospital of Hangzhou Lin'an District, Hangzhou, 311300, Zhejiang Province, China
| | - Qian Guo
- Department of Medical Oncology, The First People's Hospital of Hangzhou Lin'an District, Hangzhou, 311300, Zhejiang Province, China
| | - Rongbing Zheng
- Academician Expert Workstation of Zhejiang Luoxi Medical Technology Co., Ltd., Hangzhou, 311215, China; Zhejiang Luoxi Medical Technology Co., Ltd., Hangzhou, 311215, China.
| | - Jiongming Ying
- Department of Medical Oncology, The First People's Hospital of Hangzhou Lin'an District, Hangzhou, 311300, Zhejiang Province, China.
| |
Collapse
|
3
|
Wang Y, Hassan HM, Nisar A, Zahara SS, Akbar A, Al-Emam A. Cardioprotective potential of tectochrysin against vanadium induced heart damage via regulating NLRP3, JAK1/STAT3 and NF-κB pathway. J Trace Elem Med Biol 2025; 87:127588. [PMID: 39787653 DOI: 10.1016/j.jtemb.2025.127588] [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: 11/28/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
BACKGROUND Vanadium (VAN) is a significant trace element, but its higher exposure is reported to cause severe organ toxicity. Tectochrysin (TEC) is a naturally derived flavonoid which demonstrates a wide range of pharmacological properties. AIM The current study was planned to assess the cardioprotective potential of TEC against VAN induced cardiotoxicity in rats via regulating biochemical, and histological profile. RESEARCH PLAN Thirty-six male Sprague Dawley rats were apportioned into four groups including the control, VAN (1.5 mg/kg) treated, VAN (1.5 mg/kg) + TEC (2.5 mg/kg) administrated as well as TEC (2.5 mg/kg) alone supplemented group. The doses were administrated for 28 days through oral gavage. The biochemical and histological parameters were evaluated by using qRT-PCR, ELISA, biochemical assays, histological as well as molecular simulation techniques. FINDINGS VAN intoxication reduced the activities of catalase (CAT) (84.25 %), glutathione peroxidase (GPx) (65.28 %), glutathione reductase (GSR) (78.52 %), heme oxygenase-1 (HO-1) (81.81 %), superoxide dismutase (SOD) (83.71 %) and glutathione (GSH) (76.86 %) contents while upregulating the levels of reactive oxygen species (ROS) (87.26 %) and malondialdehyde (MDA) (91.32 %). Moreover, VAN administration increased the gene expressions of nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) (91.47 %), monocyte chemoattractant protein-1 (MCP-1) (92.51 %), interleukin-6 (IL-6) (83.63 %), tumor necrosis factor-alpha (TNF-α) (89.43 %), janus kinase 1 (JAK1) (95.55 %), signal transducer and activator of transcription 3 (STAT3) (91.25 %), nuclear factor-kappa B (NF-κB) (81.31 %), interleukin-18 (IL-18) (93.27 %), interleukin-1 beta (IL-1β) (85.79 %) and cyclooxygenase-2 (COX-2) (82.12 %). The levels of CK-MB (89.43 %), BNP (91.73 %), NT-proBNP (93.64 %), CPK (87.56 %), LDH (92.62 %), troponin I (94.25 %), troponin T (97.53 %) and CRP (88.45 %) were increased following the VAN intoxication. Besides, VAN exposure upregulated the levels of Caspase-9 (89.52 %), Bax (95.52 %) and Caspase-3 (92.52 %) while reducing the levels of Bcl-2 (75.66 %). The structural integrity of cardiac tissues was extensively disrupted following VAN-induced intoxication. However, TEC treatment remarkably ameliorated cardiotoxicity via regulating abovementioned dysregulations induced by VAN exposure. At the end, molecular docking (MD) analysis was accomplished to confirm the potential protective effect of TEC against VAN prompted cardiac dysfunction. It was detected that TEC can strongly bind with the active site of JAK1, NF-kB and STAT3 which also confirm its cardioprotective effect against VAN provoked cardiac dysfunction. CONCLUSION VAN intoxication instigated cardiac impairments which is evident by dysregulations in biochemical as well as histological profile of cardiac tissues. Nonetheless, TEC treatment remarkably protected the cardiac tissues via regulating oxidative stress, inflammation and apoptosis. TEC could be employed as cardioprotective agent against VAN induced cardiotoxicity.
Collapse
Affiliation(s)
- Yahui Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Hesham M Hassan
- Department of Pathology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Abdullah Nisar
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Syeda Sania Zahara
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Ali Akbar
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan.
| | - Ahmed Al-Emam
- Department of Pathology, College of Medicine, King Khalid University, Asir 61421, Saudi Arabia; Department of Forensic Medicine and Clinical Toxicology, Mansoura University, Egypt
| |
Collapse
|
4
|
Yan H, Zhou T, Wang Y, Liu Z, Ali I, Sheng L, Jiang Q, Li T, Xiang M, Li P, Zhang W, Teng Y, Li H, Liu Y, Cai Y. CDK5RAP3, a key defender of udder, modulates NLRP3 inflammasome activation by regulating autophagolysosome degradation in S. agalactiae-infected mastitis. Int J Biol Macromol 2023; 234:123714. [PMID: 36806767 DOI: 10.1016/j.ijbiomac.2023.123714] [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: 12/11/2022] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/21/2023]
Abstract
Streptococcus agalactiae, as one of the main pathogens of clinical and subclinical mastitis, affects animal welfare and leads to huge economic losses to farms due to the sharp decline in milk yield. However, both the real pathogenic mechanisms of S. agalactiae-induced mastitis and the regulator which controls the inflammation and autophagy are largely unknown. Served as a substrate of ubiquitin-like proteins of E3 ligase, CDK5RAP3 is widely involved in the regulation of multiple signaling pathways. Our findings revealed that CDK5RAP3 was significantly down-regulated in mastitis infected by S. agalactiae. Surprisingly, inflammasome activation was triggered by CDK5RAP3 knockdown: up-regulated NLRP3, IL1β and IL6, and cleaved caspase1 promoting by NF-κB, thereby resulting in pyroptosis. Additionally, the accumulation of autophagy markers (LC3B and p62) after CDK5RAP3 knockdown suggested that the autophagolysosome degradation pathway was inhibited, thereby activating the NF-κB pathway and NLRP3 inflammasome. Hence, our findings suggest that downregulation or ablation of CDK5RAP3 inhibits autophagolysosome degradation, causes inflammation by activating the NF-κB /NLRP3 inflammasome, and triggers cell death. In conclusion, CDK5RAP3 holds the key to understanding the interaction between autophagy and immune responses, its anti-inflammatory role in this study will throw new light on the clinical drug discovery to cure S. agalactiae mastitis.
Collapse
Affiliation(s)
- Hongchen Yan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Tianci Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yongsheng Wang
- Department of Respiratory Medicine, Nanjing Drum Tower Hospital Affiliated to Medical school of Nanjing University, Nanjing 210008, China
| | - Zhengcheng Liu
- Department of Cardiovascular and Thoracic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Ilyas Ali
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Le Sheng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Qiang Jiang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Tao Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Minghui Xiang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ping Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wei Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Honglin Li
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yang Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yafei Cai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
5
|
Stem Cell-Based Therapeutic Strategies for Premature Ovarian Insufficiency and Infertility: A Focus on Aging. Cells 2022; 11:cells11233713. [PMID: 36496972 PMCID: PMC9738202 DOI: 10.3390/cells11233713] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/14/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Reproductive aging is on the rise globally and inseparable from the entire aging process. An extreme form of reproductive aging is premature ovarian insufficiency (POI), which to date has mostly been of idiopathic etiology, thus hampering further clinical applications and associated with enormous socioeconomic and personal costs. In the field of reproduction, the important functional role of inflammation-induced ovarian deterioration and therapeutic strategies to prevent ovarian aging and increase its function are current research hotspots. This review discusses the general pathophysiology and relative causes of POI and comprehensively describes the association between the aging features of POI and infertility. Next, various preclinical studies of stem cell therapies with potential for POI treatment and their molecular mechanisms are described, with particular emphasis on the use of human induced pluripotent stem cell (hiPSC) technology in the current scenario. Finally, the progress made in the development of hiPSC technology as a POI research tool for engineering more mature and functional organoids suitable as an alternative therapy to restore infertility provides new insights into therapeutic vulnerability, and perspectives on this exciting research on stem cells and the derived exosomes towards more effective POI diagnosis and treatment are also discussed.
Collapse
|
6
|
Ali I, Li C, Kuang M, Shah AU, Shafiq M, Ahmad MA, Abdalmegeed D, Li L, Wang G. Nrf2 Activation and NF-Kb & caspase/bax signaling inhibition by sodium butyrate alleviates LPS-induced cell injury in bovine mammary epithelial cells. Mol Immunol 2022; 148:54-67. [PMID: 35671559 DOI: 10.1016/j.molimm.2022.05.121] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/17/2022] [Accepted: 05/29/2022] [Indexed: 12/23/2022]
Abstract
Mastitis, an inflammation of the mammary gland, is a complex disease that affects the health of dairy cows worldwide. Sodium butyrate (SB) is a short-chain fatty acid that has recently been shown to have antioxidant, anti-inflammatory and anti-apoptotic potential in various cells types, although its role in bovine mammary epithelial cells (bMECs) has not been comprehensively reported. Therefore, the aim of this study was to assess the protective effect of sodium butyrate on Lipopolysaccharide (LPS)-induced mastitis model in vitro and to elucidate the possible underlying molecular mechanisms. The in vitro mastitis model was designed to investigate the regulatory effect of SB on LPS-induced inflammatory conditions in bMECs, with particular emphasis on oxidative stress, inflammatory response, apoptosis, and mitochondrial dysfunction. The results showed that SB co-treatment markedly prevented LPS-induced death of bMECs in a concentration-dependent manner. In addition, SB attenuated LPS-induced oxidative stress (OS) (Increased Intracellular ROS, MDA, and decreased SOD, GSH-Px and CAT activity), thereby reduced inflammation (increased expression of IL-6, IL-Iβ, and TNF-α), and apoptosis (Increased the expression of caspases and Bax and decreased Bcl-2) via inhibiting NF-kB and caspase/bax signaling pathways. Furthermore, the protective effect of SB was also associated with the activation of endogenous antioxidant system (Nrf2, Keap1, NQO-1 and HO-1). Nrf2 silencing significantly abolished the protective effect of SB on bMECs. In conclusion, our findings suggest that SB has a significant protective effect on LPS-induced OS, inflammatory responses and apoptosis by activating Nrf2 and inhibiting NF-kB and ROS-mediated mitochondrial dysfunction. These results propose that SB may be an important regulator of OS and its subsequent inflammatory responses, and thus could be used as a therapeutic agent for bovine mastitis.
Collapse
Affiliation(s)
- Ilyas Ali
- Department of Medical Cell Biology and Genetics, Health Science Center, Shenzhen University, Shenzhen 518060, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chengmin Li
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China
| | - Meqian Kuang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Abid Ullah Shah
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Muhammad Shafiq
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Muhammad Arslan Ahmad
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Dyaaaldin Abdalmegeed
- Laboratory Center of Life Sciences, College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China; Microbiology section, Botany Department, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Lian Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Genlin Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|