1
|
Broz P. Pyroptosis: molecular mechanisms and roles in disease. Cell Res 2025; 35:334-344. [PMID: 40181184 PMCID: PMC12012027 DOI: 10.1038/s41422-025-01107-6] [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: 10/28/2024] [Accepted: 03/13/2025] [Indexed: 04/05/2025] Open
Abstract
Pyroptosis is a type of programmed necrosis triggered by the detection of pathogens or endogenous danger signals in the cytosol. Pyroptotic cells exhibit a swollen, enlarged morphology and ultimately undergo lysis, releasing their cytosolic contents - such as proteins, metabolites, and nucleic acids - into the extracellular space. These molecules can function as danger-associated molecular patterns (DAMPs), triggering inflammation when detected by neighboring cells. Mechanistically, pyroptosis is initiated by members of the gasdermin protein family, which were identified a decade ago as pore-forming executors of cell death. Mammalian gasdermins consist of a cytotoxic N-terminal domain, a flexible linker, and a C-terminal regulatory domain that binds to and inhibits the N-terminus. Proteolytic cleavage within the linker releases the N-terminal domain, enabling it to target various cellular membranes, including nuclear, mitochondrial, and plasma membranes, where it forms large transmembrane pores. Gasdermin pores in the plasma membrane disrupt the electrochemical gradient, leading to water influx and cell swelling. Their formation also activates the membrane protein ninjurin-1 (NINJ1), which oligomerizes to drive complete plasma membrane rupture and the release of large DAMPs. Since their discovery as pore-forming proteins, gasdermins have been linked to pyroptosis not only in host defense but also in various pathological conditions. This review explores the history of pyroptosis, recent insights into gasdermin activation, the cellular consequences of pore formation, and the physiological roles of pyroptosis.
Collapse
Affiliation(s)
- Petr Broz
- Department of Immunobiology, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
2
|
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
|
3
|
Duan H, Chang Q, Ding H, Shao W, Wang Y, Lu K, Zhang L, Xu J. GBP1 promotes acute rejection after liver transplantation by inducing Kupffer cells pyroptosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167644. [PMID: 39732345 DOI: 10.1016/j.bbadis.2024.167644] [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: 11/02/2024] [Revised: 12/15/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Liver transplantation is currently recognized as the most effective treatment for severe liver diseases. Although survival rates after liver transplantation have improved, rejection of the transplanted liver remains a significant cause of morbidity and transplant failure in patients. Our team previously discovered a close association between high GBP1 expression and acute rejection reactions following liver transplantation. Liver biopsies were conducted on patients who experienced acute rejection or successfully achieved immune tolerance post-transplantation. We confirmed that GBP1 was highly expressed in the acute rejection group after transplantation by Immunohistochemistry. This study aims to confirm that GBP1 promotes acute rejection reactions following liver transplantation through inducing pyroptosis in rat transplanted hepatic macrophages (KCs). We knocked down GBP1 in KCs and examined the extent of pyroptosis and the severity of acute rejection in the transplanted liver post-orthotopic liver transplantation in rats and KCs. These data provide new approaches for the study of liver transplant rejection reactions and identify new targets.
Collapse
Affiliation(s)
- Haojiang Duan
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Qingyao Chang
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Huaxing Ding
- College of Basic Medicine, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Wenhao Shao
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Yan Wang
- Department of Hepatobiliary Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Kairui Lu
- Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Li Zhang
- Department of Hepatobiliary Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.
| | - Jun Xu
- Department of Hepatobiliary Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.
| |
Collapse
|
4
|
Balahura Stămat LR, Dinescu S. Inhibition of NLRP3 inflammasome contributes to paclitaxel efficacy in triple negative breast cancer treatment. Sci Rep 2024; 14:24753. [PMID: 39433537 PMCID: PMC11494052 DOI: 10.1038/s41598-024-75805-3] [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: 08/05/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
Chronic inflammation and NLRP3 inflammasome activation are among the determining factors of breast malignancies. Paclitaxel (PTX) is a drug used in breast cancer treatment which sustains prolonged inflammation, reducing the effectiveness of chemotherapy. Considering the impact of inflammatory processes in cancer progression, there is a strong concern to develop therapeutic strategy targeting NLRP3 inflammasome for triple-negative breast cancer (TNBC) treatment. Therefore, the aim of this study was to evaluate the potential of PTX and NLRP3 inflammasome modulation to counterbalance TNBC by inducing programmed cell death and inhibiting the activity of pro-inflammatory cytokines. The obtained results suggested the strong interaction between NLRP3 inflammasome and TNBC and revealed that pharmacological inhibition, using NLRP3-specific inhibitor MCC950, and genetic silencing of NLRP3 inflammasome using specific small interfering RNA, reduced inflammatory responses and facilitated PTX-determined tumor cell death. Thus, NLRP3 inflammasome manipulation in combination with anti-tumor drugs opens up new therapeutic perspectives for TNBC therapy.
Collapse
Affiliation(s)
- Liliana-Roxana Balahura Stămat
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, 050095, Romania
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, 050095, Romania.
- Research Institute of the University of Bucharest, Bucharest, 050663, Romania.
| |
Collapse
|
5
|
Jiao Y, Li W, Yang W, Wang M, Xing Y, Wang S. Icaritin Exerts Anti-Cancer Effects through Modulating Pyroptosis and Immune Activities in Hepatocellular Carcinoma. Biomedicines 2024; 12:1917. [PMID: 39200381 PMCID: PMC11351763 DOI: 10.3390/biomedicines12081917] [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: 06/26/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 09/02/2024] Open
Abstract
Icaritin (ICT), a natural compound extracted from the dried leaves of the genus Epimedium, possesses antitumor and immunomodulatory properties. However, the mechanisms through which ICT modulates pyroptosis and immune response in hepatocellular carcinoma (HCC) remain unclear. This study demonstrated that ICT exhibits pyroptosis-inducing and anti-hepatocarcinoma effects. Specifically, the caspase1-GSDMD and caspase3-GSDME pathways were found to be involved in ICT-triggered pyroptosis. Furthermore, ICT promoted pyroptosis in co-cultivation of HepG2 cells and macrophages, regulating the release of inflammatory cytokines and the transformation of macrophages into a proinflammatory phenotype. In the Hepa1-6+Luc liver cancer model, ICT treatment significantly increased the expression of cleaved-caspase1, cleaved-caspase3, and granzyme B, modulated cytokine secretion, and stimulated CD8+ T cell infiltration, resulting in a reduction in tumor growth. In conclusion, the findings in this research suggested that ICT may modulate cell pyroptosis in HCC and subsequently regulate the immune microenvironment of the tumor. These observations may expand the understanding of the pharmacological mechanism of ICT, as well as the therapy of liver cancer.
Collapse
Affiliation(s)
- Yuanyuan Jiao
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Poyanghu Road, Jinghai District, Tianjin 301617, China;
- Bioinformatics Center of AMMS, Taiping Road, Haidian District, Beijing 100850, China; (W.L.); (W.Y.); (M.W.)
| | - Wenqian Li
- Bioinformatics Center of AMMS, Taiping Road, Haidian District, Beijing 100850, China; (W.L.); (W.Y.); (M.W.)
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Daxue Road, Jinan 250355, China
| | - Wen Yang
- Bioinformatics Center of AMMS, Taiping Road, Haidian District, Beijing 100850, China; (W.L.); (W.Y.); (M.W.)
| | - Mingyu Wang
- Bioinformatics Center of AMMS, Taiping Road, Haidian District, Beijing 100850, China; (W.L.); (W.Y.); (M.W.)
| | - Yaling Xing
- Bioinformatics Center of AMMS, Taiping Road, Haidian District, Beijing 100850, China; (W.L.); (W.Y.); (M.W.)
| | - Shengqi Wang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Poyanghu Road, Jinghai District, Tianjin 301617, China;
- Bioinformatics Center of AMMS, Taiping Road, Haidian District, Beijing 100850, China; (W.L.); (W.Y.); (M.W.)
| |
Collapse
|
6
|
Fang Q, Xu Y, Tan X, Wu X, Li S, Yuan J, Chen X, Huang Q, Fu K, Xiao S. The Role and Therapeutic Potential of Pyroptosis in Colorectal Cancer: A Review. Biomolecules 2024; 14:874. [PMID: 39062587 PMCID: PMC11274949 DOI: 10.3390/biom14070874] [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: 05/09/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related mortality worldwide. The unlimited proliferation of tumor cells is one of the key features resulting in the malignant development and progression of CRC. Consequently, understanding the potential proliferation and growth molecular mechanisms and developing effective therapeutic strategies have become key in CRC treatment. Pyroptosis is an emerging type of regulated cell death (RCD) that has a significant role in cells proliferation and growth. For the last few years, numerous studies have indicated a close correlation between pyroptosis and the occurrence, progression, and treatment of many malignancies, including CRC. The development of effective therapeutic strategies to inhibit tumor growth and proliferation has become a key area in CRC treatment. Thus, this review mainly summarized the different pyroptosis pathways and mechanisms, the anti-tumor (tumor suppressor) and protective roles of pyroptosis in CRC, and the clinical and prognostic value of pyroptosis in CRC, which may contribute to exploring new therapeutic strategies for CRC.
Collapse
Affiliation(s)
- Qing Fang
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.F.); (Y.X.); (X.T.); (X.W.)
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yunhua Xu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.F.); (Y.X.); (X.T.); (X.W.)
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Xiangwen Tan
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.F.); (Y.X.); (X.T.); (X.W.)
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Xiaofeng Wu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.F.); (Y.X.); (X.T.); (X.W.)
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Shuxiang Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.L.); (J.Y.); (X.C.); (Q.H.)
| | - Jinyi Yuan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.L.); (J.Y.); (X.C.); (Q.H.)
| | - Xiguang Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.L.); (J.Y.); (X.C.); (Q.H.)
| | - Qiulin Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.L.); (J.Y.); (X.C.); (Q.H.)
| | - Kai Fu
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shuai Xiao
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.F.); (Y.X.); (X.T.); (X.W.)
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.L.); (J.Y.); (X.C.); (Q.H.)
| |
Collapse
|
7
|
Rashid MM, Selvarajoo K. Advancing drug-response prediction using multi-modal and -omics machine learning integration (MOMLIN): a case study on breast cancer clinical data. Brief Bioinform 2024; 25:bbae300. [PMID: 38904542 PMCID: PMC11190965 DOI: 10.1093/bib/bbae300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024] Open
Abstract
The inherent heterogeneity of cancer contributes to highly variable responses to any anticancer treatments. This underscores the need to first identify precise biomarkers through complex multi-omics datasets that are now available. Although much research has focused on this aspect, identifying biomarkers associated with distinct drug responders still remains a major challenge. Here, we develop MOMLIN, a multi-modal and -omics machine learning integration framework, to enhance drug-response prediction. MOMLIN jointly utilizes sparse correlation algorithms and class-specific feature selection algorithms, which identifies multi-modal and -omics-associated interpretable components. MOMLIN was applied to 147 patients' breast cancer datasets (clinical, mutation, gene expression, tumor microenvironment cells and molecular pathways) to analyze drug-response class predictions for non-responders and variable responders. Notably, MOMLIN achieves an average AUC of 0.989, which is at least 10% greater when compared with current state-of-the-art (data integration analysis for biomarker discovery using latent components, multi-omics factor analysis, sparse canonical correlation analysis). Moreover, MOMLIN not only detects known individual biomarkers such as genes at mutation/expression level, most importantly, it correlates multi-modal and -omics network biomarkers for each response class. For example, an interaction between ER-negative-HMCN1-COL5A1 mutations-FBXO2-CSF3R expression-CD8 emerge as a multimodal biomarker for responders, potentially affecting antimicrobial peptides and FLT3 signaling pathways. In contrast, for resistance cases, a distinct combination of lymph node-TP53 mutation-PON3-ENSG00000261116 lncRNA expression-HLA-E-T-cell exclusions emerged as multimodal biomarkers, possibly impacting neurotransmitter release cycle pathway. MOMLIN, therefore, is expected advance precision medicine, such as to detect context-specific multi-omics network biomarkers and better predict drug-response classifications.
Collapse
Affiliation(s)
- Md Mamunur Rashid
- Biomolecular Sequence to Function Division, BII, (ASTAR), Singapore 138671, Republic of Singapore
| | - Kumar Selvarajoo
- Biomolecular Sequence to Function Division, BII, (ASTAR), Singapore 138671, Republic of Singapore
- Synthetic Biology Translational Research Program, Yong Loo Lin School of Medicine, NUS, Singapore 117456, Republic of Singapore
- School of Biological Sciences, Nanyang Technological University (NTU), Singapore 639798, Republic of Singapore
| |
Collapse
|
8
|
Huang H, Chen R, Deng X, Wang J, Chen J. GSDMB: A novel, independent prognostic marker and potential new therapeutic target in clear cell renal cell carcinoma. Oncol Lett 2024; 27:85. [PMID: 38249806 PMCID: PMC10797315 DOI: 10.3892/ol.2024.14219] [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: 06/12/2023] [Accepted: 11/08/2023] [Indexed: 01/23/2024] Open
Abstract
Gasdermin (GSDM) family members are involved in numerous biological processes, including pyroptosis, as well as in the initiation and progression of various types of cancer. However, the specific role of GSDM genes in clear cell renal cell carcinoma (ccRCC) has yet to be fully clarified. The present study investigated the differential expression and genetic alterations GSDM genes, their effects on prognosis and immune modulation, and their functional enrichment in ccRCC. Several bioinformatics databases were used, including UALCAN, The Cancer Genome Atlas, Gene Expression Profiling Interactive Analysis, Metascape, Tumor Immune Estimation Resource, GSCALite and cBioPortal. The results revealed that the expression levels of GSDMA, GSDMB, GSDMC and GSDMD were significantly upregulated in cancer tissues compared with those in paracancerous tissues in patients with ccRCC, whereas the expression of DFNB59 exhibited the opposite trend. The results were experimentally validated in patients with ccRCC, and it was confirmed that the expression levels of GSDMA, GSDMB, GSDMC, GSDMD and GSDME (DFNA5) were significantly enhanced, whereas (PJVK, DFNB59) expression was reduced. In addition, elevated GSDMB, GSDMD and DFNA5 expression levels were clearly associated with worse pathological characteristics of ccRCC, including a high pathological stage and high tumor grade. Furthermore, the high expression levels of GSDMB, GSDMC, GSDMD, DFNA5 and PJVK were shown to be associated with worse overall survival (OS) and progression-free interval in patients with ccRCC. Both univariate and multivariate analyses indicated that the expression of GSDMB was independently associated with the OS of patients with ccRCC. Additionally, a high mutation rate of GSDM genes (33%) was observed in patients with ccRCC, and GSDM gene mutations were also significantly associated with a poor OS in patients with ccRCC. Significant associations between GSDM genes and ccRCC immunoprofiling and drug sensitivity were also determined. In conclusion, the findings of the present study indicated that GSDMB, GSDMD and DFNA5 may be considered promising therapeutic agents and potential biomarkers for patients with ccRCC. Furthermore, GSDMB could act as an independent predictor for the OS of patients with ccRCC.
Collapse
Affiliation(s)
- Hongshuang Huang
- Department of Urology, The Affiliated Hospital of Putian University, Putian, Fujian 351100, P.R. China
| | - Ru Chen
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Xinxi Deng
- Department of Urology, Jiujiang No. 1 People's Hospital, Jiujiang, Jiangxi 332000, P.R. China
| | - Jie Wang
- Department of Ultrasound, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Jianhui Chen
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350000, P.R. China
| |
Collapse
|