1
|
Xia J, Zhuo W, Deng L, Yin S, Tang S, Yi L, Feng C, Zhong X, He Z, Sun B, Zhang C. BDNF is a prognostic biomarker involved in the immune infiltration of lung adenocarcinoma and associated with programmed cell death. Oncol Lett 2025; 29:191. [PMID: 40041412 PMCID: PMC11877015 DOI: 10.3892/ol.2025.14937] [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: 09/25/2024] [Accepted: 01/15/2025] [Indexed: 03/06/2025] Open
Abstract
It is well established that genes associated with cell death can serve as prognostic markers for patients with cancer. Programmed cell death (PCD) is known to play a role in cancer cell apoptosis and antitumor immunity. With the continuous discovery of new forms of PCD, the roles of PCD in lung adenocarcinoma (LUAD) require ongoing evaluation. In the present study, mRNA expression data and clinical information associated with 15 forms of PCD were extracted from publicly available databases and systematically analyzed. Utilizing these data, a robust risk prediction model was established that incorporates six PCD-related genes (PRGs). Datasets from the Gene Expression Omnibus database were employed to validate the six genes exhibiting risk-associated characteristics. The PRG-based model reliably predicted the prognosis of patients with LUAD, with the high-risk group showing a poor prognosis, reduced levels of immune infiltration molecules and diminished expression of human leukocyte antigens. Additionally, the relationships among PRGs, somatic mutations, tumor stemness index and immune infiltration were assessed. Based on these risk characteristics, a nomogram was constructed, patient stratification was performed, small-molecule drug candidates were predicted, and somatic mutations and chemotherapy responses were analyzed. Furthermore, reverse transcription-quantitative PCR was used to assess the expression of PDGs in vitro, and the critical role of brain-derived neurotrophic factor in LUAD development was identified through Mendelian randomization, gene knockdown, wound healing, western blot and colony formation assays. These findings offer new insights into the development of targeted therapies for LUAD, particularly in patients with high BDNF expression.
Collapse
Affiliation(s)
- Jiangnan Xia
- College of Pharmacy, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412012, P.R. China
| | - Wei Zhuo
- College of Pharmacy, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412012, P.R. China
| | - Lilan Deng
- College of Pharmacy, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412012, P.R. China
| | - Sheng Yin
- College of Pharmacy, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412012, P.R. China
| | - Shuangqin Tang
- College of Pharmacy, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412012, P.R. China
| | - Lijuan Yi
- College of Pharmacy, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412012, P.R. China
| | - Chuanping Feng
- College of Pharmacy, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412012, P.R. China
| | - Xiangyun Zhong
- College of Pharmacy, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412012, P.R. China
| | - Zhijun He
- College of Pharmacy, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412012, P.R. China
| | - Biqiang Sun
- College of Pharmacy, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412012, P.R. China
| | - Chi Zhang
- Department of Oncology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, P.R. China
| |
Collapse
|
2
|
Li NR, Zeng YX, Gu YF, Xie P, Deng BY, Lu SF, Li WA, Liu Y. Aspartame increases the risk of liver cancer through CASP1 protein: A comprehensive network analysis insights. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 294:118089. [PMID: 40139029 DOI: 10.1016/j.ecoenv.2025.118089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Aspartame is a widely used artificial sweetener in food and beverages. Its safety concerns and potential carcinogenic risks have garnered increasing attention. This study aims to systematically explore the carcinogenic potential and mechanisms of aspartame on the liver through a comprehensive analysis based on network toxicology, mendelian randomization, molecular dynamics and single-cell RNA sequencing. METHODS ProTox 3.0 and ADMEtlab 2.0 platforms were used to predict the toxicity and drug metabolism levels of aspartame. Network toxicology methods were employed to investigate the pathogenic pathways and mechanisms of aspartame in liver cancer. Mendelian randomization (MR) was used to verify the causal relationship between aspartame's carcinogenic targets and liver cancer. Furthermore, molecular docking and molecular dynamics (MD) simulations were conducted to explore the binding efficiency and stability of aspartame with the validated targets from MR. Single-cell technology further explores which types of liver cells have the highest expression of CASP1. RESULTS Combining the results from two prediction platforms, it was found that aspartame exhibits significant neurological, nephrotoxic, and hepatotoxic effects. Network toxicology results indicated that aspartame promotes the development of liver cancer by affecting multiple key proteins and regulatory factors PTGS2, IL1β and CASP1, in the Necroptosis, NF-κB and TNF signaling pathways. MR was used to discover that among the core targets of aspartame, REN, HLA-A, CASP1, and MME have causal relationships with liver cancer, while CASP1 is a risk factor for liver cancer. The binding affinity of aspartame to these four proteins was investigated by molecular docking, and it was found that the binding to CASP1 was the strongest at -18.45 kJ/mol. MD further verified that over a 50 ns period, the protein-target complex of aspartame and CASP1 exhibited excellent binding stability. Additionally, the single-cell sequencing found that CASP1 is most highly expressed in endothelial cells. In summary, these findings suggested that aspartame may increase the possibility of liver cancer by modulating the CASP1 protein. CONCLUSIONS This study identifies CASP1 as a potential target for aspartame-induced liver cancer, which is of a significant public health importance. The potential carcinogenic risk of aspartame and reliability need to be re-evaluated. The study provides a new method for assessing the safety of food additives and offers novel scientific insights into the toxicological effects of aspartame. Furthermore, subsequent experimental validation is crucial for further research into the carcinogenic mechanisms of aspartame.
Collapse
Affiliation(s)
- Ni-Ren Li
- Traditional Chinese Pharmacological Laboratory, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Yi-Xuan Zeng
- Traditional Chinese Pharmacological Laboratory, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Yu-Feng Gu
- Jiangmen Central Hospital, Jiangmen 529000, PR China.
| | - Pai Xie
- Traditional Chinese Pharmacological Laboratory, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Bing-Ying Deng
- Traditional Chinese Pharmacological Laboratory, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Si-Fan Lu
- Traditional Chinese Pharmacological Laboratory, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Wang-An Li
- College of Life Science and Chemistry, Hunan University of Technology, PR China
| | - Yi Liu
- Traditional Chinese Pharmacological Laboratory, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| |
Collapse
|
3
|
Wang Q, Jiang Y, Liao W, Zhu P. Comprehensive Pan-cancer Analysis Revealed CASP10 As a Promising Biomarker For Diverse Tumor Types. Int J Immunopathol Pharmacol 2025; 39:3946320251327620. [PMID: 40152300 PMCID: PMC11954456 DOI: 10.1177/03946320251327620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
We aimed to explore the comprehensive cancer landscape of Caspase-10 (CASP10). CASP10, a member of the caspase family, is located at the human chromosome locus 2q33-34. Studies have suggested its potential role in the development of certain cancers. To evaluate CASP10 expression in normal and pan-cancer tissues, we integrated data from The Cancer Genome Atlas (TCGA), GEO, Human Protein Atlas (HPA), and UALCAN databases. The diagnostic and prognostic significance of CASP10 was analyzed using Receiver Operating Characteristic (ROC), Cox regression, and Kaplan-Meier analysis. Correlations of CASP10 with clinical parameters were assessed via the Wilcoxon test, Kruskal-Wallis test, and logistic regression analysis. Genomic variations were explored with cBioPortal, GSCALite database, and UALCAN databases. LinkedOmics database was used to detect the function of CASP10 in pan-cancer. Interactions between CASP10 and the Tumor Immune Microenvironment (TIME) were investigated using TISIDB, TIMER2, and TISCH databases. The GSCALite database was utilized to assess the sensitivity of CASP10 to small-molecule drugs. In addition, Western Blotting (WB) was employed to detect the expression of the CASP10 in our clinical Liver Hepatocellular Carcinoma (LIHC) and Stomach Adenocarcinoma (STAD) cohorts. The transcription and protein expression of CASP10 significantly differ across cancer types, marking it as a biomarker for diagnosis and prognosis. Its expression correlated with certain clinical characteristics such as histological types and Alpha-Fetoprotein (AFP) levels. CASP10 gene exhibited a 2% alteration frequency across pan-cancer patients, with significant SNV and CNV profiles, and decreased methylation levels. CASP10 was closely related to the Nuclear Factor-κappa B (NF-κB), TNF, cell cycle, and JAK-STAT signal pathways. CASP10 showed correlation with immune components in the tumor microenvironment, including lymphocytes, immune stimulators, immune inhibitors, MHC molecules, chemokines, receptors, and Cancer-Associated Fibroblasts (CAFs). Importantly, CASP10 could predict the sensitivity of diverse anti-cancer drugs. Finally, WB analysis validated the overexpression of CASP10 in LIHC and STAD tissues. Our comprehensive bioinformatic analysis reveal the function of CASP10 on the diagnosis, prognosis, and progression of diverse cancer types.
Collapse
Affiliation(s)
- Qian Wang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- The Precision Medical Institute, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yaping Jiang
- Department of Clinical Laboratory, Xi’an NO. 3 Hospital, Xi’an, China
| | - Weijia Liao
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Pengpeng Zhu
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
4
|
Hu CY, Yin YF, Xu DP, Xu Y, Yang JY, Xu YN, Hua R. Construction and validation of immunogenic cell death-related molecular clusters, signature, and immune landscape in pancreatic cancer. Clin Exp Med 2024; 25:19. [PMID: 39708151 DOI: 10.1007/s10238-024-01533-7] [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: 07/18/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024]
Abstract
Pancreatic cancer (PC) is a malignancy of the gastrointestinal tract that is characterized by a poor prognosis. This study investigates the roles of immunogenic cell death (ICD) genes in the prognosis and progression of PC. Expression data for PC patients were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets, while ICD genes were sourced from published literature. We explored the expression patterns and identified two distinct clusters based on ICD genes. Kaplan-Meier analysis, differential expression analysis, tumor mutational burden analysis, and immune cell infiltration analysis were performed on these clusters. An ICD gene-based risk model was developed, categorizing samples from the TCGA and GEO datasets into low- and high-risk groups. Additionally, we investigated the expression levels of the genes included in the risk model within the TCGA cohort and our own samples. Finally, a loss-of-function assay was conducted to assess the role of MYD88 in PC. Two clusters of PC samples were identified, patients in the ICD-low cluster exhibited a higher degree of immune cell enrichment. The survival time of patients in the low-risk group was longer than that of those in the high-risk group. The genes included in the risk model (CASP1, MYD88, and PIK3CA) showed upregulated expression levels in tumor samples. Furthermore, the predictive accuracy of our risk model was validated using our own samples. Genetic inhibition of MYD88 led to significantly decreased proliferation and migration of PC cells in the loss-of-function assay. There were disparities in survival time and tumor immune microenvironment (TIME) between two ICD gene clusters. Additionally, we developed an ICD-related risk model that was validated as an independent prognostic indicator for patients with PC.
Collapse
Affiliation(s)
- Cheng-Yu Hu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Yi-Fan Yin
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Da-Peng Xu
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation, Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, People's Republic of China.
| | - Yu Xu
- Department of Hepatopancreatobiliary Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Jian-Yu Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Yan-Nan Xu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Rong Hua
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
5
|
Zeng X, Nie D, Liu Z, Peng X, Wang X, Qiu K, Zhong S, Liao Z, Zha X, Li Y, Zeng C. Aptamer sgc8-Modified PAMAM Nanoparticles for Targeted siRNA Delivery to Inhibit BCL11B in T-Cell Acute Lymphoblastic Leukemia. Int J Nanomedicine 2024; 19:12297-12309. [PMID: 39583320 PMCID: PMC11585994 DOI: 10.2147/ijn.s477597] [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: 07/08/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024] Open
Abstract
Introduction T-cell acute lymphoblastic leukemia (T-ALL) is a malignant hematological disease with limited targeted therapy options. Overexpression of B-cell lymphoma/leukemia 11B is frequently observed in T-ALL and contributes to leukemogenesis. Knockdown of BCL11B inhibits T-ALL cell proliferation and induces apoptosis, making it a potential therapeutic target. However, the clinical application of siRNA therapies is hindered by challenges such as poor delivery efficiency and limited clinical outcomes. Methods We developed a targeted delivery system for BCL11B siRNA (siBCL11B) using generation 5 polyamidoamine (G5-PAMAM) dendrimers conjugated with the sgc8 aptamer, which specifically binds to the T-ALL cell membrane protein PTK7. This nanoparticle, designated G5-sgc8-siBCL11B, was designed to selectively deliver siRNA to T-ALL cells. In vitro and in vivo experiments were conducted to evaluate its therapeutic efficacy and safety. Results We demonstrate that sgc8-conjugated siBCL11B nanoparticles selectively and efficiently target BCL11B-overexpressing T-ALL cells, significantly inhibiting cell viability and promoting apoptosis while exhibiting minimal impact on the viability of normal T cells. In T-ALL mouse model studies, G5-sgc8-siBCL11B and G5-siBCL11B significantly inhibited the progression of T-ALL in vivo, extending the survival of mice compared to the control (CTR), G5, and G5-sgc8 groups. Although there was no significant difference in survival between the G5-sgc8-siBCL11B and G5-siBCL11B groups, a trend towards improved survival was observed (p = 0.0993). Conclusion The G5-sgc8-siBCL11B nanoparticle system demonstrated efficient delivery and significant therapeutic efficacy, highlighting its potential as a promising novel approach for the treatment of T-ALL.
Collapse
Affiliation(s)
- Xiangbo Zeng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, People’s Republic of China
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Dingrui Nie
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, People’s Republic of China
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Zhen Liu
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Xueting Peng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Xianfeng Wang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Kangjie Qiu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Shuxin Zhong
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Ziwei Liao
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510180, People’s Republic of China
| | - Xianfeng Zha
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, People’s Republic of China
- Department of Clinical Laboratory, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, People’s Republic of China
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Chengwu Zeng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, People’s Republic of China
| |
Collapse
|
6
|
Bourne CM, Taabazuing CY. Harnessing Pyroptosis for Cancer Immunotherapy. Cells 2024; 13:346. [PMID: 38391959 PMCID: PMC10886719 DOI: 10.3390/cells13040346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024] Open
Abstract
Cancer immunotherapy is a novel pillar of cancer treatment that harnesses the immune system to fight tumors and generally results in robust antitumor immunity. Although immunotherapy has achieved remarkable clinical success for some patients, many patients do not respond, underscoring the need to develop new strategies to promote antitumor immunity. Pyroptosis is an immunostimulatory type of regulated cell death that activates the innate immune system. A hallmark of pyroptosis is the release of intracellular contents such as cytokines, alarmins, and chemokines that can stimulate adaptive immune activation. Recent studies suggest that pyroptosis promotes antitumor immunity. Here, we review the mechanisms by which pyroptosis can be induced and highlight new strategies to induce pyroptosis in cancer cells for antitumor defense. We discuss how pyroptosis modulates the tumor microenvironment to stimulate adaptive immunity and promote antitumor immunity. We also suggest research areas to focus on for continued development of pyroptosis as an anticancer treatment. Pyroptosis-based anticancer therapies offer a promising new avenue for treating immunologically 'cold' tumors.
Collapse
Affiliation(s)
| | - Cornelius Y. Taabazuing
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| |
Collapse
|