1
|
Yang X, Luo Q, Wu Z, Wang C, Yang Y, Zheng L, Li K, Zhao L, Jurong Y. Tanshinone IIA reduces tubulointerstitial fibrosis by suppressing GSDMD-mediated pyroptosis. PHARMACEUTICAL BIOLOGY 2025; 63:364-373. [PMID: 40331369 PMCID: PMC12064128 DOI: 10.1080/13880209.2025.2498166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 03/02/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025]
Abstract
CONTEXT Tanshinone IIA (Tan IIA), a bioactive compound derived from the traditional Chinese herb Salvia miltiorrhiza (Family Lamiaceae, Authority Bunge), is well-known for its protective effects in various kidney diseases. However, its role in obstructive nephropathy has not been thoroughly investigated. OBJECTIVE This study aimed to explore the protective effects of Tan IIA in a mouse model of unilateral ureteral obstruction (UUO) and to elucidate the cellular and molecular mechanisms underlying these effects. MATERIALS AND METHODS Gasdermin D (GSDMD) knockout mice and their wild-type (WT) littermates underwent UUO surgery, with Tan IIA treatment administered 24 h prior. Human proximal tubular cells (HK-2 cells) were treated with TGF-β1 to induce fibrosis (50 ng/mL for 24 h), followed by Tan IIA treatment (5 μM) for an additional 3 h. RESULTS Tan IIA significantly reduced the expression of extracellular matrix (ECM) components, including collagen I, α-smooth muscle actin (α-SMA), vimentin and fibronectin, in UUO mice. Tan IIA attenuated GSDMD-mediated pyroptosis. However, in GSDMD knockout mice subjected to UUO, the protective effects of Tan IIA on ECM gene expression and collagen deposition in the tubular interstitium were reduced. In vitro studies showed that Tan IIA reduced GSDMD activation and fibronectin protein expression in HK-2 cells. DISCUSSION AND CONCLUSIONS Tan IIA may mitigate GSDMD-mediated pyroptosis in renal tubular epithelial cells (RTECs) and reduce kidney fibrosis, highlighting its potential as a therapeutic strategy to prevent the progression of kidney disease after ureteral obstruction.
Collapse
Affiliation(s)
- Xueling Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinglin Luo
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhifen Wu
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunxuan Wang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanjing Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Luquan Zheng
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ke Li
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Lei Zhao
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Jurong
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Yao M, Miao L, Wang X, Han Y. Targeting programmed cell death pathways in gastric cancer: a focus on pyroptosis, apoptosis, necroptosis and PANoptosis. Gene 2025; 960:149546. [PMID: 40334955 DOI: 10.1016/j.gene.2025.149546] [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: 01/25/2025] [Revised: 04/05/2025] [Accepted: 05/02/2025] [Indexed: 05/09/2025]
Abstract
Gastric cancer (GC) is recognized as one of the most prevalent and serious malignancies, distinguished by its high incidence and fatality rates. Given the considerable mortality rate associated with GC, it is imperative to clarify the related pathways of GC development and further identify feasible targets for rational targeted therapy. Accumulating evidence reveals that programmed cell death (PCD) is a crucial element in both the progression and treatment of cancer. Pyroptosis, apoptosis, and necroptosis are three well-studied types of PCD, and a link between them and GC has been established in recent studies. PANoptosis, a comparatively novel type of PCD, shares key traits with pyroptosis, apoptosis, and necroptosis, yet cannot be entirely illustrated by any single model. PANoptosis has been discovered to exert an impact on multiple diseases, including cancer, infections, and inflammatory conditions, consequently offering novel perceptions into the progression and treatment of GC. This review seeks to encapsulate the emerging roles and therapeutic potential of pyroptosis, apoptosis, necroptosis, and PANoptosis in GC, laying the groundwork for the advancement of innovative treatment methods that target important signaling pathways connected with these four forms of PCD.
Collapse
Affiliation(s)
- Minghui Yao
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China
| | - Liying Miao
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China
| | - Xin Wang
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China
| | - Yangyang Han
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China; Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Xinjiang Medical University, Urumqi 830017, China.
| |
Collapse
|
3
|
Song R, Yin S, Wu J, Yan J. Neuronal regulated cell death in aging-related neurodegenerative diseases: key pathways and therapeutic potentials. Neural Regen Res 2025; 20:2245-2263. [PMID: 39104166 PMCID: PMC11759035 DOI: 10.4103/nrr.nrr-d-24-00025] [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: 01/08/2024] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 08/07/2024] Open
Abstract
Regulated cell death (such as apoptosis, necroptosis, pyroptosis, autophagy, cuproptosis, ferroptosis, disulfidptosis) involves complex signaling pathways and molecular effectors, and has been proven to be an important regulatory mechanism for regulating neuronal aging and death. However, excessive activation of regulated cell death may lead to the progression of aging-related diseases. This review summarizes recent advances in the understanding of seven forms of regulated cell death in age-related diseases. Notably, the newly identified ferroptosis and cuproptosis have been implicated in the risk of cognitive impairment and neurodegenerative diseases. These forms of cell death exacerbate disease progression by promoting inflammation, oxidative stress, and pathological protein aggregation. The review also provides an overview of key signaling pathways and crosstalk mechanisms among these regulated cell death forms, with a focus on ferroptosis, cuproptosis, and disulfidptosis. For instance, FDX1 directly induces cuproptosis by regulating copper ion valency and dihydrolipoamide S-acetyltransferase aggregation, while copper mediates glutathione peroxidase 4 degradation, enhancing ferroptosis sensitivity. Additionally, inhibiting the Xc- transport system to prevent ferroptosis can increase disulfide formation and shift the NADP + /NADPH ratio, transitioning ferroptosis to disulfidptosis. These insights help to uncover the potential connections among these novel regulated cell death forms and differentiate them from traditional regulated cell death mechanisms. In conclusion, identifying key targets and their crosstalk points among various regulated cell death pathways may aid in developing specific biomarkers to reverse the aging clock and treat age-related neurodegenerative conditions.
Collapse
Affiliation(s)
- Run Song
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Shiyi Yin
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Jiannan Wu
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Junqiang Yan
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| |
Collapse
|
4
|
Yang H, Xia Y, Ma Y, Gao M, Hou S, Xu S, Wang Y. Inhibition of the cGAS-STING pathway: contributing to the treatment of cerebral ischemia-reperfusion injury. Neural Regen Res 2025; 20:1900-1918. [PMID: 38993125 PMCID: PMC11691458 DOI: 10.4103/nrr.nrr-d-24-00015] [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: 01/05/2024] [Revised: 03/05/2024] [Accepted: 05/02/2024] [Indexed: 07/13/2024] Open
Abstract
The cGAS-STING pathway plays an important role in ischemia-reperfusion injury in the heart, liver, brain, and kidney, but its role and mechanisms in cerebral ischemia-reperfusion injury have not been systematically reviewed. Here, we outline the components of the cGAS-STING pathway and then analyze its role in autophagy, ferroptosis, cellular pyroptosis, disequilibrium of calcium homeostasis, inflammatory responses, disruption of the blood-brain barrier, microglia transformation, and complement system activation following cerebral ischemia-reperfusion injury. We further analyze the value of cGAS-STING pathway inhibitors in the treatment of cerebral ischemia-reperfusion injury and conclude that the pathway can regulate cerebral ischemia-reperfusion injury through multiple mechanisms. Inhibition of the cGAS-STING pathway may be helpful in the treatment of cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Hang Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yulei Xia
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yue Ma
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Mingtong Gao
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Shuai Hou
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Shanshan Xu
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Yanqiang Wang
- Department of Neurology II, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| |
Collapse
|
5
|
Yao J, Li Y, Liu X, Liang W, Li Y, Wu L, Wang Z, Song W. FUBP3 mediates the amyloid-β-induced neuronal NLRP3 expression. Neural Regen Res 2025; 20:2068-2083. [PMID: 39254567 PMCID: PMC11691456 DOI: 10.4103/nrr.nrr-d-23-01799] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/29/2024] [Accepted: 03/13/2024] [Indexed: 09/11/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202507000-00028/figure1/v/2024-09-09T124005Z/r/image-tiff Alzheimer's disease is characterized by deposition of amyloid-β, which forms extracellular neuritic plaques, and accumulation of hyperphosphorylated tau, which aggregates to form intraneuronal neurofibrillary tangles, in the brain. The NLRP3 inflammasome may play a role in the transition from amyloid-β deposition to tau phosphorylation and aggregation. Because NLRP3 is primarily found in brain microglia, and tau is predominantly located in neurons, it has been suggested that NLRP3 expressed by microglia indirectly triggers tau phosphorylation by upregulating the expression of pro-inflammatory cytokines. Here, we found that neurons also express NLRP3 in vitro and in vivo, and that neuronal NLRP3 regulates tau phosphorylation. Using biochemical methods, we mapped the minimal NLRP3 promoter and identified FUBP3 as a transcription factor regulating NLRP3 expression in neurons. In primary neurons and the neuroblastoma cell line Neuro2A, FUBP3 is required for endogenous NLRP3 expression and tau phosphorylation only when amyloid-β is present. In the brains of aged wild-type mice and a mouse model of Alzheimer's disease, FUBP3 expression was markedly increased in cortical neurons. Transcriptome analysis suggested that FUBP3 plays a role in neuron-mediated immune responses. We also found that FUBP3 trimmed the 5' end of DNA fragments that it bound, implying that FUBP3 functions in stress-induced responses. These findings suggest that neuronal NLRP3 may be more directly involved in the amyloid-β-to-phospho-tau transition than microglial NLRP3, and that amyloid-β fundamentally alters the regulatory mechanism of NLRP3 expression in neurons. Given that FUBP3 was only expressed at low levels in young wild-type mice and was strongly upregulated in the brains of aged mice and Alzheimer's disease mice, FUBP3 could be a safe therapeutic target for preventing Alzheimer's disease progression.
Collapse
Affiliation(s)
- Jing Yao
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuan Li
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xi Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Wenping Liang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yu Li
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Liyong Wu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Weihong Song
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang Province, China
| |
Collapse
|
6
|
Shih CC, Lin WL, Chuu CP, Lin C, Mi FL, Liu CW, Lu HY. Modified citrus pectin protects aortic dissection development involving macrophage pyroptosis. Arch Biochem Biophys 2025; 769:110428. [PMID: 40252790 DOI: 10.1016/j.abb.2025.110428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/28/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Uncontrolled aortic aneurysms can progress to aortic dissection (AD), a severe vascular disorder characterized by hematoma formation in the aortic wall, with inflammation playing a crucial role. Galectin-3 (Gal-3), a 26-kDa lectin, regulates many aspects of inflammatory cell behavior. Inhibition of Gal-3 ameliorates diabetic neuroinflammation and cardiomyopathy. Modified citrus pectin (MCP) is a PH-modified dietetic supplement produced from citrus pectin. This study investigates the therapeutic potential of MCP, which has a known affinity for Gal-3, in AD. METHODS A murine model of AD was induced by β-aminopropionitrile fumarate (BAPN)/angiotensin II (Ang-II) and treated orally with either 100 mg/kg MCP or vehicle. In vitro, H2O2 treatment was applied to RAW264.7 cells to detect macrophage death and pyroptosis. RESULTS MCP administration significantly reduced AD incidence and mortality, with decreased inflammatory cell infiltration in the aorta. MCP downregulated genes associated with inflammation and pyroptosis. In vitro, MCP mitigated macrophage death and pyroptosis induced by H2O2 treatment. The study suggests that MCP's protective effects are due to its interference with the Gal-3 and TLR4 interaction, inhibiting pyroptotic macrophage-induced inflammation. CONCLUSION MCP could improve patient outcomes and reduce progression to severe forms of AD. Clinically, MCP could serve as supportive therapy to prevent and delay aortic dissection, particularly during the acute stage of uncomplicated type B AD in patients with Marfan syndrome or abdominal aortic aneurysm.
Collapse
Affiliation(s)
- Chun-Che Shih
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, 11031, Taiwan; Taipei Heart Institute, Taipei Medical University, Taipei, 11031, Taiwan
| | - Wei-Lun Lin
- Department of Medical Research, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 407219, Taiwan; Institute of Biomedical Sciences, MacKay Medical College, No. 46, Section 3, Zhongzheng Rd, New Taipei City, 252, Taiwan
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Chi Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Fwu-Long Mi
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chen-Wei Liu
- Department of Basic Medical Science, College of Medicine, University of Arizona, Phoenix, AZ, 85721, USA
| | - Hsin-Ying Lu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, 11031, Taiwan; Department of Physical Medicine and Rehabilitation, Wan Fang Hospital, Taipei Medical University, Taipei, 11031, Taiwan; Taipei Heart Institute, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
7
|
Shawky A, Saber S, Abd El-Kader EM, El-Kashef HA. Verapamil inhibits TXNIP-dependent NLRP3 Inflammasome activation in an ulcerative colitis rat model: A new evolving role of the calcium channel blocker. Int Immunopharmacol 2025; 158:114751. [PMID: 40359884 DOI: 10.1016/j.intimp.2025.114751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025]
Abstract
Ulcerative colitis (UC) is a long-term inflammatory bowel disease (IBD) associated with significant morbidity. It is marked by inflammation and damage to the colon's mucosal lining. Studies have shown that NLRP3 inflammasome activation, apoptosis, and impaired autophagy are critical in its pathogenesis. Verapamil, a calcium channel blocker, has been found to inhibit NLRP3 inflammasome activation in various preclinical models. However, the potential influence of verapamil on the TXNIP in UC remains unexplored. This study investigates the effects of verapamil on an UC rat model induced chemically by acetic acid. Verapamil effectively inhibited the TXNIP-NLRP3-caspase-1 axis, reducing inflammasome activation and the release of IL-1β and IL-18. Additionally, verapamil suppressed NFκB, the priming step of NLRP3 activation. The drug enhanced autophagic activity, as indicated by increased expression of LC3-II and Beclin-1, along with reduced LC3-I and mTOR expression. Moreover, it demonstrated anti-apoptotic effects mediated by regulating Bax and cleaved caspase-3. These molecular changes contributed to mucosal healing and improved microscopic and macroscopic outcomes in the colitis model. Furthermore, verapamil improved the colon weight-to-length ratio and disease activity scores and mitigated oxidative stress. As verapamil has been safely used in clinics to treat hypertension, our findings suggest it may be a safe therapeutic option for ameliorating inflammation and apoptosis and activating autophagy in UC pathology. Since hypertension demonstrates a strong association with UC, the use of verapamil merits particular attention in hypertensive patients fighting against IBD.
Collapse
Affiliation(s)
- Ahmed Shawky
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt.
| | - Eman M Abd El-Kader
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt.
| | - Hassan A El-Kashef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
8
|
Senapati S, Bertolini TB, Minnier MA, Yazicioglu MN, Markusic DM, Zhang R, Wicks J, Nahvi A, Herzog RW, Walsh MC, Cejas PJ, Armour SM. Inhibition of IFNAR-JAK signaling enhances tolerability and transgene expression of systemic non-viral DNA delivery. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102502. [PMID: 40206655 PMCID: PMC11979999 DOI: 10.1016/j.omtn.2025.102502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/28/2025] [Indexed: 04/11/2025]
Abstract
Lipid nanoparticles (LNPs) have demonstrated significant therapeutic value for non-viral delivery of mRNA and siRNA. While there is considerable interest in utilizing LNPs for delivering DNA (DNA-LNPs) to address a broad range of genetic disorders, acute inflammatory responses pose significant safety concerns and limit transgene expression below therapeutically relevant levels. However, the mechanisms and immune signaling pathways underlying DNA-LNP-triggered inflammatory responses are not well characterized. Through the use of gene-targeted mouse models, we have identified cGAS-STING and interferon-α/β receptor (IFNAR) pathways as major mediators of acute inflammation triggered by systemic delivery of DNA-LNPs. cGAS-STING activation induces expression of numerous JAK-STAT-activating cytokines, and we show that treatment of mice with the JAK inhibitors ruxolitinib or baricitinib significantly improves tolerability to systemically delivered DNA-LNPs. Furthermore, specific inhibition of IFNAR signaling enhances both DNA-LNP tolerability and transgene expression. Utilization of JAK inhibitors or IFNAR blockade represent promising strategies for enhancing the safety and efficacy of non-viral DNA delivery for gene therapy.
Collapse
Affiliation(s)
| | - Thais B. Bertolini
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - David M. Markusic
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rui Zhang
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Joan Wicks
- Gene Therapy Research, Spark Therapeutics, Philadelphia, PA, USA
| | - Ali Nahvi
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Roland W. Herzog
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Pedro J. Cejas
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Sean M. Armour
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| |
Collapse
|
9
|
Fan G, Liu Y, Tao L, Wang D, Huang Y, Yang X. Sodium butyrate alleviates colitis by inhibiting mitochondrial ROS mediated macrophage pyroptosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167756. [PMID: 40044062 DOI: 10.1016/j.bbadis.2025.167756] [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: 11/20/2024] [Revised: 02/06/2025] [Accepted: 02/26/2025] [Indexed: 04/15/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory bowel disease with unclear causes and limited treatment options. Sodium butyrate (NaB), a byproduct of dietary fiber in the intestine, has demonstrated efficacy in treating inflammation. However, the precise anti-inflammatory mechanisms of NaB in colon inflammation remain largely unexplored. This study aims to investigate the effects of NaB on dextran sulfate sodium (DSS)-induced colitis in rats. The findings indicate that oral administration of NaB effectively prevent colitis and reduce levels of serum or colon inflammatory factors. Additionally, NaB demonstrated in vitro inhibition of RAW264.7 inflammation cytokines induced by LPS, along with suppression of the ERK and NF-κB signaling pathway activation. Moreover, NaB mitigated LPS and Nigericin-induced RAW264.7 pyroptosis by reducing indicators of mitochondrial damage, including increased mitochondrial membrane potential (JC-1) levels and decreased Mito-ROS production. NaB increases ZO-1 and Occludin expression in CaCo2 cells by inhibiting RAW264.7 pyroptosis. These results suggest that NaB could be utilized as a therapeutic agent or dietary supplement to alleviate colitis.
Collapse
Affiliation(s)
- Guoqiang Fan
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yaxin Liu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Limei Tao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Danping Wang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yizhu Huang
- Singao Xiamen Company, Xiamen 361006, PR China
| | - Xiaojing Yang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
10
|
Lippincott MJ, Tomkinson J, Bunten D, Mohammadi M, Kastl J, Knop J, Schwandner R, Huang J, Ongo G, Robichaud N, Dagher M, Mansilla-Soto A, Saravia-Estrada C, Tsuboi M, Basualto-Alarcón C, Way GP. A morphology and secretome map of pyroptosis. Mol Biol Cell 2025; 36:ar63. [PMID: 40202832 DOI: 10.1091/mbc.e25-03-0119] [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: 04/11/2025] Open
Abstract
Pyroptosis represents one type of programmed cell death. It is a form of inflammatory cell death that is canonically defined by caspase-1 cleavage and Gasdermin-mediated membrane pore formation. Caspase-1 initiates the inflammatory response (through IL-1β processing), and the N-terminal cleaved fragment of Gasdermin D polymerizes at the cell periphery forming pores to secrete proinflammatory markers. Cell morphology also changes in pyroptosis, with nuclear condensation and membrane rupture. However, recent research challenges canon, revealing a more complex secretome and morphological response in pyroptosis, including overlapping molecular characterization with other forms of cell death, such as apoptosis. Here, we take a multimodal, systems biology approach to characterize pyroptosis. We treated human peripheral blood mononuclear cells (PBMCs) with 36 different combinations of stimuli to induce pyroptosis or apoptosis. We applied both secretome profiling (nELISA) and high-content fluorescence microscopy (Cell Painting). To differentiate apoptotic, pyroptotic, and control cells, we used canonical secretome markers and modified our Cell Painting assay to mark the N-terminus of Gasdermin D. We trained hundreds of machine learning (ML) models to reveal intricate morphology signatures of pyroptosis that implicate changes across many different organelles and predict levels of many proinflammatory markers. Overall, our analysis provides a detailed map of pyroptosis which includes overlapping and distinct connections with apoptosis revealed through a mechanistic link between cell morphology and cell secretome.
Collapse
Affiliation(s)
- Michael J Lippincott
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045
| | - Jenna Tomkinson
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045
| | - Dave Bunten
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045
| | | | | | | | | | | | - Grant Ongo
- Nomic Bio, Montreal, Québec, Canada H2T 1C1
| | | | | | | | | | - Masafumi Tsuboi
- Department of Chemistry and Biotechnology, University of Tokyo, Tokyo, Japan 113-0033
| | - Carla Basualto-Alarcón
- Health Sciences Department, University of Aysén, Coyhaique, Chile
- Anatomy and Legal Medicine Department, University of Chile, Santiago, Chile
| | - Gregory P Way
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045
| |
Collapse
|
11
|
Khemraj P, Kuznyetsova A, Hood DA. Effect of aging, endurance training, and denervation on innate immune signaling in skeletal muscle. J Appl Physiol (1985) 2025; 138:1341-1356. [PMID: 40338209 DOI: 10.1152/japplphysiol.00038.2025] [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: 01/16/2025] [Revised: 02/11/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
Skeletal muscle function relies on mitochondria for energy and for mediating its unique adaptive plasticity. The NLRP3 inflammasome complex is an innate immune mechanism that responds to mitochondrial damage-associated molecular patterns (DAMPs); however, its activity relative to mitochondrial dysfunction in muscle requires exploration. The purpose of this study was to characterize immune signaling and mitochondrial function in muscle during aging, endurance training, and disuse induced by denervation. Denervation led to decreases in muscle mass, mitochondrial content, and impaired respiration. Protein analyses revealed increases in NF-κB p65 and downstream inflammatory markers, including NLRP3, caspase-1, GSDMD-N, STING, and IL-1β, along with pro-apoptotic BAX and AIF. When assessing potential DAMPS, denervation led to increased ROS production but no changes in cytosolic mtDNA levels, relative to total mtDNA. Since we hypothesized that inflammasome activation would be increased with age, we studied young (6-8 mo) and aged (21-22 mo) mice that remained sedentary or underwent a 6-wk voluntary running protocol. Aging resulted in marked increases in the expression of multiple pro-inflammatory and pro-apoptotic proteins. Remarkably, training uniformly attenuated age-related increases in BAX, NLRP3, caspase-1, STING, and gasdermin-D (GSDMD) protein expression and tended to reduce the elevated level of cytosolic mtDNA evident in aged muscle. Training adaptations were also evident in the aged animals by the preservation of muscle mass and improvements in oxygen consumption and endurance performance, and were achieved despite a lower training distance than in young animals. Our results strongly implicate endurance training as a promising therapeutic for combating disuse and age-related inflammation in skeletal muscle.NEW & NOTEWORTHY This study aims to further elucidate the role between skeletal muscle activity, mitochondria, and innate immune signaling, specifically looking at the NLRP3 inflammasome complex. Our results demonstrate that disuse conditions such as aging and denervation were associated with a pro-inflammatory phenotype and impairments in mitochondrial functioning, whereas chronic endurance training demonstrated anti-inflammatory effects. Notably, this work highlights the benefits of exercise in attenuating age-related inflammation in aged skeletal muscle.
Collapse
Affiliation(s)
- Priyanka Khemraj
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Anastasiya Kuznyetsova
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - David A Hood
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Bano N, Khan S, Ahamad S, Dar NJ, Alanazi HH, Nazir A, Bhat SA. Microglial NOX2 as a therapeutic target in traumatic brain injury: Mechanisms, consequences, and potential for neuroprotection. Ageing Res Rev 2025; 108:102735. [PMID: 40122395 DOI: 10.1016/j.arr.2025.102735] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability worldwide, with secondary injury mechanisms, including neuroinflammation and oxidative stress, driving much of its chronic pathology. While NADPH oxidase 2 (NOX2)-mediated reactive oxygen species (ROS) production is a recognized factor in TBI, the specific role of microglial NOX2 in perpetuating oxidative and inflammatory damage remains underexplored. Addressing this gap is critical, as current therapeutic approaches primarily target acute symptoms and fail to interrupt the persistent neuroinflammation that contributes to progressive neurodegeneration. Besides NOX, other ROS-generating enzymes, such as CYP1B1, COX2, and XO, also play crucial roles in triggering oxidative stress and neuroinflammatory conditions in TBI. However, this review highlights the pathophysiological role of microglial NOX2 in TBI, focusing on its activation following injury and its impact on ROS generation, neuroinflammatory signaling, and neuronal loss. These insights reveal NOX2 as a critical driver of secondary injury, linked to worsened outcomes, particularly in aged individuals where NOX2 activation is more pronounced. In addition, this review evaluates emerging therapeutic approaches targeting NOX2, such as GSK2795039 and other selective NOX2 inhibitors, which show potential in reducing ROS levels, limiting neuroinflammation, and preserving neurological functions. By highlighting the specific role of NOX2 in microglial ROS production and secondary neurodegeneration, this study advocates for NOX2 inhibition as a promising strategy to improve TBI outcomes by addressing the unmet need for therapies targeting long-term inflammation and neuroprotection. Our review highlights the potential of NOX2-targeted interventions to disrupt the cycle of oxidative stress and inflammation, ultimately offering a pathway to mitigate the chronic impact of TBI.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al Jouf University 77455, Saudi Arabia
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research, New Delhi, India.
| | - Shahnawaz Ali Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
13
|
Abdelaziz AM. Alpha-Synuclein drives NURR1 and NLRP3 Inflammasome dysregulation in Parkinson's disease: From pathogenesis to potential therapeutic strategies. Int Immunopharmacol 2025; 156:114692. [PMID: 40267723 DOI: 10.1016/j.intimp.2025.114692] [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: 03/10/2025] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
Parkinson's disease (PD), a progressive neurodegenerative disorder, is characterized by the loss of dopaminergic neurons and pathological aggregation of α-synuclein (α-Syn). Emerging evidence highlights the interplay between genetic susceptibility, neuroinflammation, and transcriptional dysregulation in driving PD pathogenesis. This review brings together the latest information on three important players: α-Syn, the transcription factor Orphan nuclear receptor (NURR1), and the NOD-like receptor 3 (NLRP3) inflammasome. Pathogenic α-syn aggregates cause damage to neurons by disrupting mitochondria and lysosomes and spreading in a way similar to prion proteins. They also turn on the NLRP3 inflammasome, which is a key player in neuroinflammation. NLRP3-driven release of pro-inflammatory cytokines exacerbates neurodegeneration and creates a self-sustaining inflammatory milieu. Meanwhile, reduced NURR1 activity, a pivotal modulator of dopaminergic neuron survival and development, exposes neurons to oxidative stress, neuroinflammation, and α-Syn toxicity, hence exacerbating disease progression. So, targeting this trio exhibits transformative potential against PD pathogenesis.
Collapse
Affiliation(s)
- Ahmed M Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish 45511, Egypt.
| |
Collapse
|
14
|
Wang L, Chen H, Lu C, Ding Y, He Y, Xu J, Xu J, Zhang Z. Rebastinib attenuates acute lung injury by promoting NLRP3 ubiquitination and blocking NLRP3/GSDMD signaling pathway in macrophages and protecting alveolar epithelial cells. Int Immunopharmacol 2025; 159:114819. [PMID: 40403501 DOI: 10.1016/j.intimp.2025.114819] [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: 10/20/2024] [Revised: 04/24/2025] [Accepted: 05/06/2025] [Indexed: 05/24/2025]
Abstract
Acute lung injury (ALI) is a devastating inflammatory lung disease with high morbidity and mortality. Characterized by diffuse alveolar damage, macrophages infiltration, and pulmonary edema, ALI currently lacks effective therapeutic strategies. Rebastinib is a small molecule inhibitor of the Tie2 receptor and an antineoplastic drug. This study investigated the effects of Rebastinib on lipopolysaccharide (LPS)-induced ALI and GSDMD-mediated pyroptosis and NLRP3 inflammasome activation in vitro and in vivo. Our results revealed that Rebastinib significantly attenuated GSDMD-dependent pyroptosis in macrophages, leading to reduced production of caspase-1, LDH and IL-1β. Mechanistically, Rebastinib promoted NLRP3 ubiquitination, thereby disrupting the connection between ASC and NLRP3 and effectively suppressing NLRP3 inflammasome assembly. Additionally, Rebastinib exhibited effective protection function on alveolar epithelial cells in a co-culture system. Furthermore, Rebastinib administration alleviated lung inflammatory damage in LPS-induced ALI mouse model. These findings suggest that Rebastinib holds promise as a therapeutic candidate for ALI by inhibiting the activation of pyroptosis and NLRP3 inflammasome on macrophages.
Collapse
Affiliation(s)
- Lingqiao Wang
- Department of Emergency, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health
| | - Hao Chen
- Department of Orthopedics Surgery, Hangzhou First People's Hospital, Hangzhou, Zhejiang Province 31000, China
| | - Congcong Lu
- Department of Orthopedics Surgery, Hangzhou First People's Hospital, Hangzhou, Zhejiang Province 31000, China; The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou 310053, PR China
| | - Yi Ding
- Department of Orthopedics Surgery, Hangzhou First People's Hospital, Hangzhou, Zhejiang Province 31000, China
| | - Ying He
- Department of Cardiac Ultrasound Center, Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou City, Zhejiang Province 310000, China
| | - Jian Xu
- Department of Laboratory Medicine, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University
| | - Jiani Xu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| | - Zhen Zhang
- Department of Orthopedics Surgery, Hangzhou First People's Hospital, Hangzhou, Zhejiang Province 31000, China.
| |
Collapse
|
15
|
Li X, Liu B, Huang D, Ma N, Xia J, Zhao X, Duan Y, Li F, Lin S, Tang S, Li Q, Rao J, Zhang X. Chidamide triggers pyroptosis in T-cell lymphoblastic lymphoma/leukemia via the FOXO1/GSDME axis. Chin Med J (Engl) 2025; 138:1213-1224. [PMID: 39445538 PMCID: PMC12091596 DOI: 10.1097/cm9.0000000000003214] [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: 02/26/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND T-cell lymphoblastic lymphoma/acute lymphoblastic leukemia (T-LBL/ALL) is an aggressive form of hematological malignancy associated with poor prognosis in adult patients. Histone deacetylases (HDACs) are aberrantly expressed in T-LBL/ALL and are considered potential therapeutic targets. Here, we investigated the antitumor effect of a novel HDAC inhibitor, chidamide, on T-LBL/ALL. METHODS HDAC1, HDAC2 and HDAC3 levels in T-LBL/ALL cell lines and patient samples were compared with those in normal controls. Flow cytometry, transmission electron microscopy, and lactate dehydrogenase release assays were conducted in Jurkat and MOLT-4 cells to assess apoptosis and pyroptosis. A specific forkhead box O1 (FOXO1) inhibitor was used to rescue pyroptosis and upregulated gasdermin E (GSDME) expression caused by chidamide treatment. The role of the FOXO1 transcription factor was evaluated by dual-luciferase reporter and chromatin immunoprecipitation assays. The efficacy of chidamide in vivo was evaluated in a xenograft mouse. RESULTS The expression of HDAC1, HDAC2 and HDAC3 was significantly upregulated in T-LBL/ALL. Cell viability was obviously inhibited after chidamide treatment. Pyroptosis, characterized by cell swelling, pore formation on the plasma membrane and lactate dehydrogenase leakage, was identified as a new mechanism of chidamide treatment. Chidamide triggered pyroptosis through caspase 3 activation and GSDME transcriptional upregulation. Chromatin immunoprecipitation assays confirmed that chidamide led to the increased transcription of GSDME through a more relaxed chromatin structure at the promoter and the upregulation of FOXO1 expression. Moreover, we identified the therapeutic effect of chidamide in vivo . CONCLUSIONS This study suggested that chidamide exerts an antitumor effect on T-LBL/ALL and promotes a more inflammatory form of cell death via the FOXO1/GSDME axis, which provides a novel choice of targeted therapy for patients with T-LBL/ALL.
Collapse
Affiliation(s)
- Xinlei Li
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Jinfeng Laboratory, Chongqing 400037, China
| | - Bangdong Liu
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Jinfeng Laboratory, Chongqing 400037, China
| | - Dezhi Huang
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Jinfeng Laboratory, Chongqing 400037, China
| | - Naya Ma
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Jinfeng Laboratory, Chongqing 400037, China
| | - Jing Xia
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Jinfeng Laboratory, Chongqing 400037, China
| | - Xianlan Zhao
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Jinfeng Laboratory, Chongqing 400037, China
| | - Yishuo Duan
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Jinfeng Laboratory, Chongqing 400037, China
| | - Fu Li
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Jinfeng Laboratory, Chongqing 400037, China
| | - Shijia Lin
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Jinfeng Laboratory, Chongqing 400037, China
| | - Shuhan Tang
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Jinfeng Laboratory, Chongqing 400037, China
| | - Qiong Li
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Jinfeng Laboratory, Chongqing 400037, China
| | - Jun Rao
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Jinfeng Laboratory, Chongqing 400037, China
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Jinfeng Laboratory, Chongqing 400037, China
| |
Collapse
|
16
|
Chen D, Guo Z, Yao L, Sun Y, Dian Y, Zhao D, Ke Y, Zeng F, Zhang C, Deng G, Li L. Targeting oxidative stress-mediated regulated cell death as a vulnerability in cancer. Redox Biol 2025; 84:103686. [PMID: 40424719 DOI: 10.1016/j.redox.2025.103686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Reactive oxygen species (ROS), regulators of cellular behaviors ranging from signaling to cell death, have complex production and control mechanisms to maintain a dynamic redox balance under physiological conditions. Redox imbalance is frequently observed in tumor cells, where ROS within tolerable limits promote oncogenic transformation, while excessive ROS induce a range of regulated cell death (RCD). As such, targeting ROS-mediated regulated cell death as a vulnerability in cancer. However, the precise regulatory networks governing ROS-mediated cancer cell death and their therapeutic applications remain inadequately characterized. In this Review, we first provide a comprehensive overview of the mechanisms underlying ROS production and control within cells, highlighting their dynamic balance. Next, we discuss the paradoxical nature of the redox system in tumor cells, where ROS can promote tumor growth or suppress it, depending on the context. We also systematically explored the role of ROS in tumor signaling pathways and revealed the complex ROS-mediated cross-linking networks in cancer cells. Following this, we focus on the intricate regulation of ROS in RCD and its current applications in cancer therapy. We further summarize the potential of ROS-induced RCD-based therapies, particularly those mediated by drugs targeting specific redox balance mechanisms. Finally, we address the measurement of ROS and oxidative damage in research, discussing existing challenges and future prospects of targeting ROS-mediated RCD in cancer therapy. We hope this review will offer promise for the clinical application of targeting oxidative stress-mediated regulated cell death in cancer therapy.
Collapse
Affiliation(s)
- Danyao Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China; Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Lei Yao
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Deze Zhao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yizhe Ke
- The First Affliated Hospital of Shihezi University, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China.
| | - Linfeng Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
17
|
Zhang Y, Guan Y, Dai M, Yang Y, Yang F. Microcystin-LR induces lung injury in mice through the NF-κB/NLRP3 pathway. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:385-394. [PMID: 39773316 DOI: 10.1080/15287394.2024.2443525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Microcystin-LR (MC-LR) a cyclic toxin produced by cyanobacterial species is known to exert detrimental effects on various organs, including lung. Several investigators demonstrated that MC-LR exerts pulmonary toxicity, but the underlying mechanisms remain unclear. This study aimed to investigate whether exposure to MC-LR-induced lung inflammation and examine the underlying mechanisms. Thirty specific pathogen-free (SPF) male mice were allocated into control and MC-LR treatment groups. Mice were intraperitoneally injected with physiological saline or MC-LR (20 μg/kg) daily for a total of 21 days. Our findings indicated that exposure to MC-LR-produced histopathological changes in lung tissue, including thickening of alveolar walls and inflammatory infiltration. MC-LR was found to upregulate mRNA expression levels of pro-inflammatory cytokines TNFα, IL-6, IL-1β, and IL-18. Further, MC-LR significantly elevated the expression levels of proteins associated with the NF-κB/NLRP3 pathway p-NF-κB, NLRP3, Caspase-1, ASC. The activation of NF-κB/NLRP3 pathway further promoted the release of inflammatory cytokine IL-1β and cleavage of pyroptosis-associated GSDMD protein. These findings indicate that MC-LR may induce lung inflammation by promoting cell pyroptosis via the activation of the NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Yin Zhang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ying Guan
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Manni Dai
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yue Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Department of Public Health, The Central Hospital of Shaoyang, Shaoyang, China
| | - Fei Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| |
Collapse
|
18
|
Sun C, Gui J, Sheng Y, Huang L, Zhu X, Huang K. Specific signaling pathways mediated programmed cell death in tumor microenvironment and target therapies. Discov Oncol 2025; 16:776. [PMID: 40377777 PMCID: PMC12084487 DOI: 10.1007/s12672-025-02592-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
Increasing evidence has shown that programmed cell death (PCD) plays a crucial role in tumorigenesis and cancer progression. The components of PCD are complex and include various mechanisms such as apoptosis, necroptosis, alkaliptosis, oxeiptosis, and anoikis, all of which are interrelated in their functions and regulatory pathways. Given the significance of these processes, it is essential to conduct a comprehensive study on PCD to elucidate its multifaceted nature. Key signaling pathways, particularly the caspase signaling pathway, the RIPK1/RIPK3/MLKL pathway, and the mTOR signaling pathway, are pivotal in regulating PCD and influencing tumor progression. In this review, we briefly describe the generation mechanisms of different PCD components and focus on the regulatory mechanisms of these three major signaling pathways within the context of global PCD. Furthermore, we discuss various tumor therapeutic compounds that target different signaling axes of these pathways, which may provide novel strategies for effective tumor therapy and help improve patient outcomes in cancer treatment.
Collapse
Affiliation(s)
- Chengpeng Sun
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Jiawei Gui
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Yilei Sheng
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Le Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China
| | - Xingen Zhu
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Nanchang, 330006, Jiangxi, China.
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Kai Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Nanchang, 330006, Jiangxi, China.
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
19
|
Zeng X, Yuan Y, Li Y, Hu Z, Hu S. Deciphering the NLRP3 inflammasome in diabetic encephalopathy: Molecular insights and emerging therapeutic targets. Exp Neurol 2025; 391:115304. [PMID: 40383363 DOI: 10.1016/j.expneurol.2025.115304] [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: 12/17/2024] [Revised: 05/01/2025] [Accepted: 05/11/2025] [Indexed: 05/20/2025]
Abstract
Diabetic encephalopathy (DE) is a neurological complication characterized by neuroinflammation, cognitive impairment, and memory decline, with its pathogenesis closely linked to the activation of the NLRP3 inflammasome. As a central regulator of the innate immune system, the NLRP3 inflammasome plays a pivotal role in DE progression by mediating neuroinflammation, pyroptosis, mitochondrial dysfunction, oxidative stress, endoplasmic reticulum (ER) stress, and microglial polarization. This review systematically explores the molecular mechanisms by which the NLRP3 inflammasome contributes to DE, focusing on its role in neuroinflammatory cascades and neuronal damage, as well as the diabetes-associated physiological changes that exacerbate DE pathogenesis. Furthermore, we summarize emerging therapeutic strategies targeting the NLRP3 inflammasome, including small-molecule inhibitors and bioactive compounds derived from traditional herbal medicine, highlighting their potential for DE treatment. These findings not only advance our understanding of DE but also provide a foundation for developing NLRP3-targeted pharmacological interventions.
Collapse
Affiliation(s)
- Xinyi Zeng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The First Clinical Medical College of Nanchang University, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yi Yuan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; School of Huankui Academy, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yujia Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Ziyan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Shan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China.
| |
Collapse
|
20
|
Qin Z, Zhang H, Zhang J, Li T, Kuca K, Liu J, Wu W. Deoxynivalenol induces pyroptosis and IL-1β secretion via P2X7R signal in murine RAW264.7 macrophages. Toxicon 2025; 263:108418. [PMID: 40381923 DOI: 10.1016/j.toxicon.2025.108418] [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: 04/12/2025] [Revised: 05/11/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Deoxynivalenol (DON), a trichothecene mycotoxin, exerts pro-inflammatory and immunomodulatory activity. Interleukin (IL)-1β serves a crucial part as a gate keeper of inflammation in DON-induced macrophages, but an overview of how DON exposure elicits IL-1β secretion from RAW264.7 cells has not been fully illustrated. Here we found that the cellular phenomenon, involved with a type of programmed cell death known as pyroptosis, contains: 1) increase of pro-IL-1β expression, 2) motivation of caspase-1, 3) caspase-1-dependent maturement of IL-1β, 4) caspase-1 fragmentation of gasdermin D (GSDMD), and 5) IL-1β secretion through GSDMD pore. Mechanistically, the present study certified that DON both as first and second signals engaged in IL-1β release is mediated by purinergic P2X7 receptor (P2X7R)-Src signaling. During this process, P2X7R signal is required for GSDMD pore forming course in ASC-independent manner. Moreover, blocking of K+ efflux, ROS formation, as well as cathepsin B activity decreases IL-1β export. Our data show that exposure to DON does cause pyroptosis and IL-1β secretion via P2X7R signal in RAW264.7 macrophages. Overall, these results provide new mechanistic clue for DON as a pro-inflammatory factor in innate immune signaling events.
Collapse
Affiliation(s)
- Zihui Qin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Huayue Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Engineering Research Center of Bio-process, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Jie Zhang
- School of Biology and Food Engineering, Changshu Institute of Technology, Suzhou, 215500, China
| | - Tushuai Li
- School of Biology and Food Engineering, Changshu Institute of Technology, Suzhou, 215500, China
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Jiaguo Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Wenda Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Engineering Research Center of Bio-process, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China; Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic.
| |
Collapse
|
21
|
Antonello J, Roy P. Damage-Associated Molecular Patterns (DAMPs) In Vascular Diseases. J Biol Chem 2025:110241. [PMID: 40381697 DOI: 10.1016/j.jbc.2025.110241] [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: 03/10/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
Research into the role of chronic sterile inflammation (i.e. a prolonged inflammatory state not caused by an infectious agent), in vascular disease progression has continued to grow over the last few decades. DAMPs have a critical role in this research due to their ability to link stress-causing cardiovascular risk factors to inflammatory phenotypes seen in vascular disease. In this mini-review, we will briefly summarize the DAMPs and receptor signaling pathways that have been extensively studied in the context of vascular disease, including TLRs, RAGE, cGAS-STING, and the NLRP3 inflammasome. In particular, we will discuss how these pathways can promote the release of pro-inflammatory cytokines and chemokines as well as vascular remodeling. Next, we will summarize the results of studies which have linked the various pro-inflammatory effects of DAMPs with the phenotypes in the context of vascular diseases including atherosclerosis, fibrosis, aneurysm, ischemia, and hypertension. Finally, we will discuss some pre-clinical and clinical trials that have targeted DAMPs, their receptors, or the products of their signaling pathways, and discuss the outlook and future directions for the field at large.
Collapse
Affiliation(s)
| | - Partha Roy
- Bioengineering, University of Pittsburgh; Pathology, University of Pittsburgh.
| |
Collapse
|
22
|
Tang Y, Tong W, Peng Y, Sun S. Targeting cholesterol-driven pyroptosis: a promising strategy for the prevention and treatment of atherosclerosis. Mol Biol Rep 2025; 52:459. [PMID: 40372511 DOI: 10.1007/s11033-025-10554-8] [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: 03/16/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025]
Abstract
Funding Pyroptosis is a type of programmed cell death (PCD) pathway distinguished by inflammation. It is activated by specific inflammasomes. Once activated, it causes the physical breakdown of the cell, along with the discharge of pro-inflammatory cytokines, such as interleukin-1β (IL-1β) and interleukin-18 (IL-18). Abundant evidence has demonstrated the existence of pyroptotic cell death within atherosclerotic plaques, which has significance for the development of atherosclerosis (AS). As a result, pyroptosis has become a new and important topic in cardiovascular disease (CVD) research. Cholesterol, it is recognized to have a connection with inflammation, exerts a crucial function in the development process of AS, and has been linked to the initiation of pyroptosis. This review aims to briefly summarize the fundamental aspects of pyroptosis and the influence of cholesterol-related inflammation in AS. Additionally, this review will explore potential therapeutic approaches based on pyroptosis that could be utilized for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Yuehong Tang
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wenjuan Tong
- Department of Gynecology and Obstetrics, First Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, China
| | - Yujiao Peng
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shaowei Sun
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
23
|
Zhu X, Xu K, Ai S, Zhang Y, Chu C, Wei R, Gao S, Liu L, Li W, Zhang Y, Kikete S, Liu X, Zhang Z, Li X. miR-126-5p protects from URSA via inhibiting Caspase-1-dependent pyroptosis of trophoblast cells. Cell Mol Life Sci 2025; 82:204. [PMID: 40372489 DOI: 10.1007/s00018-025-05713-w] [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: 02/06/2025] [Revised: 04/07/2025] [Accepted: 04/11/2025] [Indexed: 05/16/2025]
Abstract
Unexplained recurrent spontaneous abortion (URSA) is a distressing pregnancy complication that seriously threat to women's reproductive health. Trophoblast pyroptosis was involved in the occurrence of URSA, but the potential mechanism remains unclear. In this work, we found CASP1 transcription and the level of pyroptosis were significantly elevated in the villous tissues of URSA patients. Suppression of cell pyroptosis by Gasdermin-D (GSDMD) or Caspase-1 inhibitors can reduce embryo resorption rate of URSA mice, while Caspase-1 over-expression in normal pregnant (NP) mice can aggravate embryo resorption. Meanwhile, a pronounced decline in the expression of microRNA-126-5p (miR-126-5p) was found in URSA patients, which was inversely related to CASP1 expression. Over-expression of miR-126-5p restrained trophoblast pyroptosis via inhibiting Caspase-1/GSDMD signaling pathway by direct binding to 3'-UTR of CASP1. Moreover, experiments in vivo substantiated that up-regulation of miR-126-5p effectively suppressed Caspase-1-mediated pyroptosis in placental tissue and significantly reduced embryo resorption rate. Collectively, these results underscored that diminished miR-126-5p expression plays a crucial role in URSA by enhancing trophoblast pyroptosis through activating Caspase-1/GSDMD signaling pathway. As a result, miR-126-5p shows significant promise as a possible biomarker for diagnosis and treatment of URSA.
Collapse
Affiliation(s)
- Xiaoxiao Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Changqing District, Jinan, 250399, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Ke Xu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Changqing District, Jinan, 250399, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Shuang Ai
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yingjie Zhang
- The First Clinical College of Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chu Chu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Changqing District, Jinan, 250399, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Ran Wei
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Changqing District, Jinan, 250399, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Shufeng Gao
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Changqing District, Jinan, 250399, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Lu Liu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Changqing District, Jinan, 250399, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Wei Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Changqing District, Jinan, 250399, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Yunhong Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Changqing District, Jinan, 250399, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Siambi Kikete
- School of Health Sciences, Department of Pharmacognosy and Pharmaceutical Chemistry, Kenyatta University, Nairobi, 00609, Kenya
| | - Xinkui Liu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Changqing District, Jinan, 250399, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Zhen Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Changqing District, Jinan, 250399, China.
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China.
| | - Xia Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Changqing District, Jinan, 250399, China.
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China.
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
24
|
Szczerba M, Ganesh A, Gil-Marqués ML, Briken V, Goldberg MB. NLRP11 is required for canonical NLRP3 and non-canonical inflammasome activation during human macrophage infection with mycobacteria. mBio 2025; 16:e0081825. [PMID: 40272180 PMCID: PMC12077127 DOI: 10.1128/mbio.00818-25] [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: 03/10/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
The NLRP11 protein is only expressed in primates and participates in the activation of the canonical NLRP3 and non-canonical NLRP3 inflammasome activation after infection with gram-negative bacteria. Here, we generated a series of defined NLRP11 deletion mutants to further analyze the role of NLRP11 in NLRP3 inflammasome activation. Like the complete NLRP11 deletion mutant (NLRP11-/-), the NLRP11 mutant lacking the NAIP, C2TA, HET-E, and TP1 (NACHT) and leucine-rich repeat (LRR) domains (NLRP11∆N_LRR) showed reduced activation of the canonical NLRP3 inflammasome, whereas a pyrin domain mutant (NLRP11∆PYD) had no effect on NLRP3 activation. The NLRP11-/- and NLRP11∆N_LRR mutants, but not the NLRP11∆PYD mutant, also displayed reduced activation of caspase-4 during infection with the intracytosolic, gram-negative pathogen Shigella flexneri. We found that the human-adapted, acid-fast pathogen Mycobacterium tuberculosis and the opportunistic pathogen Mycobacterium kansasii both activate the non-canonical NLRP11 inflammasome in a caspase-4/caspase-5-dependent pathway. In conclusion, we show that NLRP11 functions in the non-canonical caspase-4/caspase-5 inflammasome activation pathway and the canonical NLRP3 inflammasome pathway and that NLRP11 is required for full recognition of mycobacteria by each of these pathways. Our work extends the spectrum of bacterial pathogen recognition by the non-canonical NLRP11-caspase4/caspase-5 pathway beyond gram-negative bacteria.IMPORTANCEThe activation of inflammasome complexes plays a crucial role in intracellular pathogen detection. NLRP11 and caspase-4 are essential for recognizing lipopolysaccharide (LPS), a molecule found in gram-negative bacteria such as the human pathogens Shigella spp., which activate both canonical NLRP3 and non-canonical inflammasome pathways. Through a series of deletion mutants, we demonstrate that the NACHT and LRR domains of NLRP11, but not its pyrin domain, are critical for detection of S. flexneri. Notably, our research reveals that the acid-fast bacterium M. tuberculosis is also detected by NLRP11 and caspase-4, despite not producing LPS. These findings significantly expand the range of pathogens recognized by NLRP11 and caspase-4 to now include acid-fast bacteria that do not contain LPS and underscore the versatility of these innate immune components in pathogen detection.
Collapse
Affiliation(s)
- Mateusz Szczerba
- Division of Infectious Diseases, Department of Medicine, Center for Bacterial Pathogenesis, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Akshaya Ganesh
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - María Luisa Gil-Marqués
- Division of Infectious Diseases, Department of Medicine, Center for Bacterial Pathogenesis, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Volker Briken
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Marcia B. Goldberg
- Division of Infectious Diseases, Department of Medicine, Center for Bacterial Pathogenesis, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Li Y, Gao W, Qiu Y, Pan J, Guo Q, Liu X, Geng L, Shen Y, Deng Y, Hu Z, Li S, Liu S, Idris A, Huang J, Yang H, Ge B, Fan X, Chen X, Li J. RING1 dictates GSDMD-mediated inflammatory response and host susceptibility to pathogen infection. Cell Death Differ 2025:10.1038/s41418-025-01527-2. [PMID: 40369166 DOI: 10.1038/s41418-025-01527-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 04/27/2025] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
RING1 is an E3 ligase component of the polycomb repressive complex 1 (PRC1) with known roles in chromatin regulation and cellular processes such as apoptosis and autophagy. However, its involvement in inflammation and pyroptosis remains elusive. Here, we demonstrate that human RING1, not RING2, promotes K48-linked ubiquitination of Gasdermin D (GSDMD) and acts as a negative regulator of pyroptosis and bacterial infection. Indeed, we showed that loss of Ring1 increased S. typhimurium infectious load and mortality in vivo. Though RING1 deletion initially reduced M. tuberculosis (Mtb) infectious load in vivo, increased lung inflammation and impaired immune defense responses were later observed. Moreover, Ring1 knockout exacerbated acute sepsis induced by lipopolysaccharide (LPS) in vivo. Mechanistically, RING1 directly interacts with GSDMD and ubiquitinates the K51 and K168 sites of GSDMD for K48-linked proteasomal degradation, thereby inhibiting pyroptosis. Inhibition of RING1 E3 ligase activity by direct mutation or with the use of small molecule inhibitors increased GSDMD level and cell death during pyroptosis. Our findings reveal that RING1 dictates GSDMD-mediated inflammatory response and host susceptibility to pathogen infection, highlighting RING1 as a potential therapeutic target for combating infectious diseases.
Collapse
Affiliation(s)
- Yuanyuan Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Wenqing Gao
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Yuxin Qiu
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Jiasong Pan
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Qingqing Guo
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Xuehe Liu
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Lu Geng
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Yajie Shen
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Yifan Deng
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Zhidong Hu
- Shanghai Institute of Infectious Diseases and Biosecurity & Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Suhua Li
- Division of Natural Science, Duke Kunshan University, Suzhou, China
| | - Shanshan Liu
- Shanghai Key Laboratory of Tuberculosis, Department of Microbiology and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Adi Idris
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jinqing Huang
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Hua Yang
- Shanghai Key Laboratory of Tuberculosis, Department of Microbiology and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Baoxue Ge
- Shanghai Key Laboratory of Tuberculosis, Department of Microbiology and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyong Fan
- Shanghai Institute of Infectious Diseases and Biosecurity & Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xiangjun Chen
- Department of Neurology, Huashan Hospital and Institute of Neurology, Fudan University, Shanghai, China
| | - Jixi Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
26
|
Hartmann S, Radochonski L, Ye C, Martinez-Sobrido L, Chen J. SARS-CoV-2 ORF3a drives dynamic dense body formation for optimal viral infectivity. Nat Commun 2025; 16:4393. [PMID: 40355429 PMCID: PMC12069715 DOI: 10.1038/s41467-025-59475-x] [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: 05/08/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
SARS-CoV-2 hijacks multiple organelles for virion assembly, of which the mechanisms have not been fully understood. Here, we identified a SARS-CoV-2-driven membrane structure named the 3a dense body (3DB). 3DBs are unusual electron-dense and dynamic structures driven by the accessory protein ORF3a via remodeling a specific subset of the trans-Golgi network (TGN) and early endosomal membrane. 3DB formation is conserved in related bat and pangolin coronaviruses but was lost during the evolution to SARS-CoV. During SARS-CoV-2 infection, 3DB recruits the viral structural proteins spike (S) and membrane (M) and undergoes dynamic fusion/fission to maintain the optimal unprocessed-to-processed ratio of S on assembled virions. Disruption of 3DB formation resulted in virions assembled with an abnormal S processing rate, leading to a dramatic reduction in viral entry efficiency. Our study uncovers the crucial role of 3DB in maintaining maximal SARS-CoV-2 infectivity and highlights its potential as a target for COVID-19 prophylactics and therapeutics.
Collapse
Affiliation(s)
- Stella Hartmann
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Lisa Radochonski
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Jueqi Chen
- Department of Microbiology, University of Chicago, Chicago, IL, USA.
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA.
| |
Collapse
|
27
|
Bahmani F, Shayanmanesh M, Safari M, Alaei A, Yasaman Pouriafar, Rasti Z, Zaker F, Rostami S, Damerchiloo F, Safa M. Bone marrow microenvironment in myelodysplastic neoplasms: insights into pathogenesis, biomarkers, and therapeutic targets. Cancer Cell Int 2025; 25:175. [PMID: 40349084 PMCID: PMC12065391 DOI: 10.1186/s12935-025-03793-z] [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: 12/26/2024] [Accepted: 04/17/2025] [Indexed: 05/14/2025] Open
Abstract
Myelodysplastic neoplasms (MDS) represent a heterogeneous group of malignant hematopoietic stem and progenitor cell (HSPC) disorders characterized by cytopenia, ineffective hematopoiesis, as well as the potential to progress to acute myeloid leukemia (AML). The pathogenesis of MDS is influenced by intrinsic factors, such as genetic insults, and extrinsic factors, including altered bone marrow microenvironment (BMM) composition and architecture. BMM is reprogrammed in MDS, initially to prevent the development of the disease but eventually to provide a survival advantage to dysplastic cells. Recently, inflammation or age-related inflammation in the bone marrow has been identified as a key pathogenic mechanism for MDS. Inflammatory signals trigger stress hematopoiesis, causing HSPCs to emerge from quiescence and resulting in MDS development. A better understanding of the role of the BMM in the pathogenesis of MDS has opened up new avenues for improving diagnosis, prognosis, and treatment of the disease. This article provides a comprehensive review of the current knowledge regarding the significance of the BMM to MDS pathophysiology and highlights recent advances in developing innovative therapies.
Collapse
Affiliation(s)
- Forouzan Bahmani
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Shayanmanesh
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahdi Safari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirarsalan Alaei
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Yasaman Pouriafar
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Rasti
- Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Zaker
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shahrbano Rostami
- Research Institute for Oncology, Hematology and Cell Therapy, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Damerchiloo
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Berk BC, Chávez CL, George Hsu C. PDE10A Inhibition Reduces NLRP3 Activation and Pyroptosis in Sepsis and Nerve Injury. Int J Mol Sci 2025; 26:4498. [PMID: 40429643 PMCID: PMC12111586 DOI: 10.3390/ijms26104498] [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: 03/24/2025] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Cell death and inflammation are key innate immune responses, but excessive activation can cause tissue damage. The NLRP3 inflammasome is a promising target for reducing inflammation and promoting recovery. Immunometabolism regulates NLRP3 responses in neurological and inflammatory diseases through cyclic nucleotide signaling. Targeting phosphodiesterases (PDEs), which hydrolyze cAMP and cGMP, offer a novel approach to mitigate inflammation. While 14 PDE inhibitors are FDA-approved, PDE10A's role in NLRP3 inflammasome activation remains unclear. This study investigates the effects of PDE10A inhibition on inflammasome-driven inflammation using two PDE10A inhibitors, MP-10 and TP-10, in macrophage and animal models of sepsis and traumatic nerve injury. Our results show that PDE10A inhibition reduces inflammasome activation by preventing ASC speck formation and by lowering levels of cleaved caspase-1, gasdermin D, and IL-1β, which are key mediators of pyroptosis. In the sepsis model, MP-10 significantly reduced inflammation, decreased plasma IL-1β, alleviated thrombocytopenia, and improved organ damage markers. In the nerve injury model, PDE10A inhibition enhanced motor function recovery and reduced muscle atrophy-related gene expression. These findings suggest that PDE10A inhibition could be a promising therapeutic approach for inflammatory and neuromuscular injuries. Given MP-10's established safety in human trials, Phase 2 clinical studies for sepsis and nerve injury are highly promising.
Collapse
Affiliation(s)
- Bradford C. Berk
- Department of Physical Medicine and Rehabilitation, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Camila Lage Chávez
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA;
| | - Chia George Hsu
- Department of Kinesiology, The University of Texas at San Antonio, San Antonio, TX 78249, USA
- Department of Molecular Microbiology and Immunology, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| |
Collapse
|
29
|
Zhou X, Wang Z, Wang Y, Xu G, Luo M, Zhang M, Li Y. Rutin ameliorates LPS-induced acute lung injury in mice by inhibiting the cGAS-STING-NLRP3 signaling pathway. Front Pharmacol 2025; 16:1590096. [PMID: 40406492 PMCID: PMC12095315 DOI: 10.3389/fphar.2025.1590096] [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: 03/08/2025] [Accepted: 04/22/2025] [Indexed: 05/26/2025] Open
Abstract
Introduction Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), represent critical respiratory failures with high mortality rates and limited treatment options. While the flavonoid rutin exhibits documented anti-inflammatory and antioxidant properties, its therapeutic mechanisms in ALI/ARDS remain unclear. This study investigated rutin's efficacy against lipopolysaccharide (LPS)-induced ALI in mice, with a mechanistic focus on the cGAS-STING pathway and NLRP3 inflammasome activation. Methods Male C57BL/6 mice were divided into Vehicle control, LPS induction, LPS + rutin co-treatment, and Rutin monotherapy groups. ALI was induced by intratracheal LPS challenge, and rutin was administered via gavage. Proteomics analysis, histological evaluation, immunohistochemistry, TUNEL staining, immunofluorescence, RT-qPCR, western blot, ELISA, and oxidative stress assays were performed to assess the effects of rutin on ARDS. Results The proteomic profiling of lung tissues from LPS-challenged mice identified significant dysregulation of proteins integral to the cGAS-STING cascade and pyroptotic processes. Gene ontology and KEGG pathway analyses underscored the pivotal role of immune and inflammatory responses in ALI, particularly in cytosolic DNA-sensing and NOD-like receptor signaling pathways. Rutin administration significantly alleviated LPS-induced lung injury, reducing oxidative stress, apoptosis, and proinflammatory cytokine levels (IL-6, IL-1β, TNF-α). Mechanistically, rutin demonstrated dual suppression: 1) inhibiting cGAS-STING activation through decreased expression of cGAS, STING, and phosphorylation of TBK1/IRF3 (P<0.05), and 2) attenuating NLRP3-mediated pyroptosis via downregulation of NLRP3-ASC-caspase1-GSDMD signaling (P<0.05). Pharmacological STING inhibition (C-176) validated the cGAS-STING-NLRP3 regulatory hierarchy in ALI pathogenesis. Conclusion These findings elucidate rutin's novel therapeutic mechanism through coordinated suppression of the cGAS-STING-NLRP3 axis, positioning it as a promising candidate for ALI/ARDS intervention.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Respiratory Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zhibin Wang
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yuting Wang
- Department of Respiratory Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Guofeng Xu
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Min Luo
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Mengwei Zhang
- Department of Respiratory Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yuying Li
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
30
|
Chen G, Sun M, Li M, Ma J, He L, Xiong J, Gao C, Xu X. Ruxolitinib Ameliorates Neuronal Pyroptosis in the Acute Phase of Intracerebral Hemorrhage through Inhibiting the Activation of Caspase-8. ACS Chem Neurosci 2025; 16:1827-1837. [PMID: 40286335 DOI: 10.1021/acschemneuro.5c00022] [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: 04/29/2025] Open
Abstract
Intracerebral hemorrhage (ICH) is a common type of stroke with higher rates of death and neurological dysfunction than ischemic stroke. Based on previous studies, we found that reducing neuronal pyroptosis in the acute phase of ICH improved the neurological dysfunction of mice that suffered from nontraumatic parenchymal hemorrhage. Still, the mechanism must be further explored. In this study, we used ruxolitinib, a selective inhibitor of JAK1/2, to treat CD-1 mice with ICH. We found that inhibition of the JAK1/STAT1 pathway alleviated ICH-induced neuronal pyroptosis and that the activation of caspase-8 was suppressed at the same time. Given that caspase-8 is crosstalk for different types of programmed cell death and its role in the pyroptotic cell death after ICH has not yet been defined, we administered z-IETD-fmk, a selective inhibitor of caspase-8, to treat mice with ICH. We found that the downregulation of caspase-8 reversed ICH-induced neuronal pyroptosis and improved motor and cognitive functions of mice after ICH. Our results show that the JAK1/STAT1/caspase-8 axis is a critical mediator of neuronal pyroptosis in ICH. Inhibiting this axis improved neurological outcomes of mice with ICH, and we propose ruxolitinib as a potential therapeutic approach for post-ICH treatment.
Collapse
Affiliation(s)
- Guang Chen
- School of Forensic Medicine, Wannan Medical College, Wuhu 241000, China
| | - Mengbei Sun
- School of Forensic Medicine, Wannan Medical College, Wuhu 241000, China
| | - Mingming Li
- School of Forensic Medicine, Wannan Medical College, Wuhu 241000, China
| | - Jiaqi Ma
- School of Forensic Medicine, Wannan Medical College, Wuhu 241000, China
| | - Liangchao He
- School of Forensic Medicine, Wannan Medical College, Wuhu 241000, China
| | - Junjie Xiong
- School of Forensic Medicine, Wannan Medical College, Wuhu 241000, China
| | - Cheng Gao
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Xiang Xu
- School of Forensic Medicine, Wannan Medical College, Wuhu 241000, China
| |
Collapse
|
31
|
Curwin AJ, Kurokawa K, Bigliani G, Brouwers N, Nakano A, Malhotra V. The pathway of unconventional protein secretion involves CUPS and a modified trans-Golgi network. J Cell Biol 2025; 224:e202312120. [PMID: 40015244 PMCID: PMC11867701 DOI: 10.1083/jcb.202312120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 12/03/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
Compartment for unconventional protein secretion (CUPS), a compartment for secretion of signal sequence-lacking proteins, forms through COPI-independent extraction of membranes from early Golgi cisternae, lacks Golgi-specific glycosyltransferases, and requires phosphatidylinositol 4-phosphate (PI4P) for biogenesis, as well as phosphatidylinositol 3-phosphate for stability. Our findings demonstrate that Drs2, a PI4P effector from the trans-Golgi network (TGN), is essential for CUPS formation, specifically through its interaction with Rcy1, and Rcy1 is crucial for the unconventional secretion. Using 4D super-resolution confocal live imaging microscopy, we observed that CUPS interact with a modified TGN that contains Drs2 in addition to proteins Tlg2 and Snc2, which are necessary for membrane fusion. Notably, while CUPS remain stable, the modified TGN undergoes remodeling during the later stages of unconventional secretion. In summary, we suggest that CUPS and the modified TGN, without the function of COPII and COPI, participate in collecting and sorting unconventionally secreted proteins, reflecting the role of Golgi membranes in receiving cargo from the ER during conventional secretion.
Collapse
Affiliation(s)
- Amy J. Curwin
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Kazuo Kurokawa
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Wako, Japan
| | - Gonzalo Bigliani
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Nathalie Brouwers
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Akihiko Nakano
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Wako, Japan
| | - Vivek Malhotra
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- ICREA, Barcelona, Spain
| |
Collapse
|
32
|
Nadendla EK, Tweedell RE, Kasof G, Kanneganti TD. Caspases: structural and molecular mechanisms and functions in cell death, innate immunity, and disease. Cell Discov 2025; 11:42. [PMID: 40325022 PMCID: PMC12052993 DOI: 10.1038/s41421-025-00791-3] [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: 06/28/2024] [Accepted: 03/05/2025] [Indexed: 05/07/2025] Open
Abstract
Caspases are critical regulators of cell death, development, innate immunity, host defense, and disease. Upon detection of pathogens, damage-associated molecular patterns, cytokines, or other homeostatic disruptions, innate immune sensors, such as NLRs, activate caspases to initiate distinct regulated cell death pathways, including non-lytic (apoptosis) and innate immune lytic (pyroptosis and PANoptosis) pathways. These cell death pathways are driven by specific caspases and distinguished by their unique molecular mechanisms, supramolecular complexes, and enzymatic properties. Traditionally, caspases are classified as either apoptotic (caspase-2, -3, -6, -7, -8, -9, and -10) or inflammatory (caspase-1, -4, -5, and -11). However, extensive data from the past decades have shown that apoptotic caspases can also drive lytic inflammatory cell death downstream of innate immune sensing and inflammatory responses, such as in the case of caspase-3, -6, -7, and -8. Therefore, more inclusive classification systems based on function, substrate specificity, or the presence of pro-domains have been proposed to better reflect the multifaceted roles of caspases. In this review, we categorize caspases into CARD-, DED-, and short/no pro-domain-containing groups and examine their critical functions in innate immunity and cell death, along with their structural and molecular mechanisms, including active site/exosite properties and substrates. Additionally, we highlight the emerging roles of caspases in cellular homeostasis and therapeutic targeting. Given the clinical relevance of caspases across multiple diseases, improved understanding of these proteins and their structure-function relationships is critical for developing effective treatment strategies.
Collapse
Affiliation(s)
- Eswar Kumar Nadendla
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rebecca E Tweedell
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gary Kasof
- Cell Signaling Technology, Danvers, MA, USA
| | | |
Collapse
|
33
|
Hossain MJ, Romanov KA, Jian J, Swaby LC, Bandyopadhyay S, Guan I, Thomas SM, Olive AJ, O’Connor TJ. Bacterial pathogens hijack host cell peroxisomes for replication vacuole expansion and integrity. SCIENCE ADVANCES 2025; 11:eadr8005. [PMID: 40305606 PMCID: PMC12042894 DOI: 10.1126/sciadv.adr8005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025]
Abstract
Pathogens manipulate host cell organelles to establish infection. There is extensive evidence of pathogen modulation of the endoplasmic reticulum, Golgi apparatus, mitochondria, endosomes, lysosomes, and nucleus. However, one organelle that has been largely overlooked in connection with bacterial pathogenesis is peroxisomes. Here, we demonstrate that Legionella actively recruits peroxisomes to its replication vacuole using a secreted bacterial effector protein. Defects in peroxisome metabolic function restrict expansion of the Legionella vacuole membrane and cause rupture of this compartment, inhibiting bacterial replication and leading to bacterial degradation. Similarly, peroxisome dysfunction causes Salmonella replication vacuole destabilization and reduced bacterial burden within host cells. Thus, these two intracellular bacterial pathogens exploit host cell peroxisomes to maintain their replication compartments, establishing a critical role for this organelle in disease.
Collapse
Affiliation(s)
- Mohammad J. Hossain
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Katerina A. Romanov
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeffrey Jian
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Louis C. Swaby
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Saumya Bandyopadhyay
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ivan Guan
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sean M. Thomas
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Andrew J. Olive
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Tamara J. O’Connor
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
34
|
Zhang T, Sun Y, Xia J, Fan H, Shi D, Wu Q, Huang M, Hou XY. Targeting HPK1 inhibits neutrophil responses to mitigate post-stroke lung and cerebral injuries. EMBO Mol Med 2025; 17:1018-1040. [PMID: 40169896 DOI: 10.1038/s44321-025-00220-8] [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: 09/25/2024] [Revised: 03/01/2025] [Accepted: 03/10/2025] [Indexed: 04/03/2025] Open
Abstract
Circulating neutrophils are responsible for poor neurological outcomes and have been implicated in respiratory morbidity after acute ischemic stroke (AIS). However, the molecular mechanisms regulating neutrophil responses and their pathological relevance in post-stroke complications remain unclear. In this study, we investigated the involvement of hematopoietic progenitor kinase 1 (HPK1) in neutrophil responses and mobilization, as well as subsequent lung and cerebral injuries following AIS. We found that lipopolysaccharide treatment triggered neutrophil activation in an HPK1-dependent manner. HPK1 enhanced intrinsic NF-κB/STAT3/p38-MAPK pathways and gasdermin D cleavage, leading to neutrophil hyperactivation. Following AIS, HPK1 promoted the mobilization of CXCR2high bone marrow neutrophils. HPK1 loss inhibited peripheral neutrophil hyperactivation, neutrophil infiltration, and aggregation of neutrophil extracellular traps, progressively alleviating systemic inflammation and impairments in mouse pulmonary and neurological functions. Furthermore, HPK1 pharmacological inhibition attenuated post-stroke pulmonary and neurological impairments in mice. Our findings revealed that HPK1 upregulates neutrophil mobilization and various responses, promoting post-stroke systemic inflammation and tissue injury. This study highlights HPK1 as a therapeutic target for improving pulmonary and neurological functions after AIS.
Collapse
Affiliation(s)
- Tingting Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Ying Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jing Xia
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Hongye Fan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Dingfang Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Qian Wu
- The Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Ming Huang
- Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Xiao-Yu Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
35
|
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
|
36
|
Qiu X, Huang W, Liang J, Chen H, Sha W, Lyu Y, Chen K, Yang H, Zhang Q. Predicting prognosis, immune landscape, and drug targets with a novel signature for hepatocellular carcinoma. Technol Health Care 2025; 33:1367-1380. [PMID: 40331560 DOI: 10.1177/09287329241296358] [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: 05/08/2025]
Abstract
BackgroundDespite advances in therapeutics, hepatocellular carcinoma (HCC) remains one of the most malignant types of digestive tract cancers with a poor prognosis. Pyroptosis is a form of programmed cell death induced by inflammatory caspases. Recent studies have identified pyroptosis, a form of programmed cell death induced by inflammatory caspases, as playing a role in tumorigenesis and cancer progression. However, the functions and mechanisms of pyroptosis in HCC are barely explored.MethodsGene expression and clinical data were derived from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. A prognostic signature and nomogram were constructed by on differentially expressed genes and clinical data. Pathway enrichment and immune cell infiltration were further analyzed. Potential drugs to modulate the pathways were explored.ResultsIn this study, a pyroptosis-related gene signature was developed and identified to be significantly correlated with the survival of HCC patients. Additionally, a nomogram on the basis of pyroptosis-related genes was constructed with distinct prognostic values. Furthermore, the pyroptosis-related gene signature might correlate with immune-related pathways and the regulation of the immune microenvironment, and several compounds (KIN001-220, TPCA-1, LY-303511, physostigmine, vemurafenib, etc.) could potentially reverse the pathogenic gene-expression patterns.Conclusions: Our study provides evidence that pyroptosis is involved in HCC development, progression and immune microenvironment, which is promising in predicting the prognosis and developing targeted therapy.
Collapse
Affiliation(s)
- Xinqi Qiu
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
- Cancer Prevention Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Wentao Huang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Jun Liang
- Geriatric Critical Care Medicine, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510080, China
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
- Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Weihong Sha
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
- Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Yanlin Lyu
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
- Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Kequan Chen
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510080, China
| | - Hongwei Yang
- Department of Medical Ultrasound, The First affiliated hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Qingfang Zhang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
| |
Collapse
|
37
|
Zhang XY, Li YY, Chen W, Zhou YM, Zhou L, Xie LL, Hu YQ, Huang HX, Zhao CC, Qin Y, Lan T, Sun WC. The underlying mechanism of Porcine Teschovirus 2 3C pro antagonizing the NLRP3 inflammasome. Vet Microbiol 2025; 304:110479. [PMID: 40132520 DOI: 10.1016/j.vetmic.2025.110479] [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: 12/06/2024] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025]
Abstract
Porcine teschovirus (PTV) can cause diseases such as poliomyelitis, pneumonia, and reproductive disorders in sows, but research on the pathogenesis of PTV infection is limited. In this investigation, we observed that PTV infection inhibits the activation of the NLRP3 inflammasome. PTV 3Cpro inhibits the activation of the NLRP3 inflammasome and pyroptosis by degrading NLRP3, IL-1β, and GSDMD. The degradation mechanism of 3Cpro involves the interaction of NLRP3, IL-1β, and 3Cpro, and 3Cpro degrades IL-1β through the caspase pathway. The mechanism by which PTV 3Cpro degrades GSDMD diverges from other picornavirus, remaining mechanistically elusive. Moreover, 3Cpro cannot degrade target proteins after their protease activity is lost. Our study provides new insights into the mechanism of antagonizing programmed cell death by PTV.
Collapse
Affiliation(s)
- Xin-Yu Zhang
- Institute of Virology, Wenzhou University, Wenzhou 325035, China
| | - Yu-Ying Li
- College of Animal Sciences, Institute of Preventive Veterinary Medicine, Zhejiang University, Hangzhou 310000, China
| | - Wei Chen
- Institute of Virology, Wenzhou University, Wenzhou 325035, China
| | - Yi-Min Zhou
- Institute of Virology, Wenzhou University, Wenzhou 325035, China
| | - Lin Zhou
- Institute of Virology, Wenzhou University, Wenzhou 325035, China
| | - Lu-Lu Xie
- Institute of Virology, Wenzhou University, Wenzhou 325035, China
| | - Yan-Qing Hu
- Department of Children's Respiration disease, the Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Hai-Xin Huang
- Institute of Virology, Wenzhou University, Wenzhou 325035, China; College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Chen-Chen Zhao
- Institute of Virology, Wenzhou University, Wenzhou 325035, China
| | - Yan Qin
- Institute of Virology, Wenzhou University, Wenzhou 325035, China
| | - Tian Lan
- Institute of Virology, Wenzhou University, Wenzhou 325035, China.
| | - Wen-Chao Sun
- Institute of Virology, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
38
|
Poppenborg T, Saljic A, Bruns F, Abu-Taha I, Dobrev D, Fender AC. A short history of the atrial NLRP3 inflammasome and its distinct role in atrial fibrillation. J Mol Cell Cardiol 2025; 202:13-23. [PMID: 40057301 DOI: 10.1016/j.yjmcc.2025.02.011] [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: 11/14/2024] [Revised: 01/21/2025] [Accepted: 02/24/2025] [Indexed: 04/23/2025]
Abstract
Inflammasomes are multiprotein complexes of the innate immune system that mediate inflammatory responses to infection and to local and systemic stress and tissue injury. The principal function is to facilitate caspase-1 auto-activation and subsequently maturation and release of the effectors interleukin (IL)-1β and IL-18. The atrial-specific NLRP3 inflammasome is a unifying causal feature of atrial fibrillation (AF) development, progression and recurrence after ablation. Many AF-associated risk factors and co-morbidities converge mechanistically on the activation of this central inflammatory signaling platform. This review presents the historical conceptual development of a distinct atrial inflammasome and its potential causal involvement in AF. We follow the early observations linking systemic and local inflammation with AF, to the emergence of an atrial-intrinsic NLRP3 inflammasome operating within not just immune cells but also in resident atrial fibroblasts and cardiomyocytes. We outline the key developments in understanding how the atrial NLRP3 inflammasome and its effector IL-1β contribute causally to cellular and tissue-level arrhythmogenesis in different pathological settings, and outline candidate therapeutic concepts verified in preclinical models of atrial cardiomyopathy and AF.
Collapse
Affiliation(s)
| | - Arnela Saljic
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Florian Bruns
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany
| | - Issam Abu-Taha
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA; Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
| | - Anke C Fender
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
39
|
Toscano ECB, Justo AFO, Paula MCA, Grossi LB, Neves VH, Leite REP, Paes VR, Melo RCN, Nitrini R, Pasqualucci C, Ferriolli E, Teixeira AL, Grinberg LT, Suemoto CK. Upregulation of NLRP3 Inflammasome in Specific Hippocampal Regions: Strengthening the Link Between Neuroinflammation and Selective Vulnerability in Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04975-6. [PMID: 40281298 DOI: 10.1007/s12035-025-04975-6] [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: 12/10/2024] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Neuroinflammation has emerged as an important mechanism in the early stages of neurodegenerative diseases. Experimental models have demonstrated the detrimental role of inflammasomes in the development of Alzheimer's disease (AD). However, neuropathological studies characterizing NLRP1 and NLRP3 pathways in AD are scarce. In addition, the possible association between inflammasome-induced neuroinflammation and clinicopathological outcomes is unclear. This study aimed to characterize the hippocampal expression of the inflammasome proteins in post-mortem samples of individuals with pure AD neuropathological change (ADNC) compared to controls from an admixed Latin American sample (n = 28 per group). We also investigated potential associations of inflammasome expression with neuropathological burden and cognitive abilities. The expression of NLRP1, NLRP3, caspase-1, ASC, gasdermin D, IL-1β, IL-18, amyloid β, and hyperphosphorylated tau (p-tau) was evaluated in the cornu ammonis (CA), dentate gyrus (DG), and subiculum (SUB), using immunohistochemistry and morphometry. We also performed the alignment of serial sections and 3D reconstruction of ADNC samples to verify the spatial locations of NLRP3/ASC and AD pathology across the hippocampus. We used ordinal logistic regression to investigate potential associations between inflammasome proteins and AD pathology, while linear regression assessed relationships between inflammasome and cognitive abilities. NLRP3, ASC, caspase-1, IL-1β, and IL-18 were overexpressed in CA and SUB of individuals with ADNC compared to controls. NLRP3 pathway correlated with AD pathology and CDR-SB, mainly in CA and SUB. Our results suggest that hippocampal NLRP3, but not NLRP1, inflammasome was associated with pathologic burden and cognitive impairment in AD and may contribute to the selective vulnerability to AD pathology.
Collapse
Affiliation(s)
- Eliana C B Toscano
- Laboratory of Pathology, Department of Pathology, Federal University of Juiz de Fora Medical School, Eugênio Do Nascimento, S/No.-Dom Bosco, Juiz de Fora, MG, 36038 - 330, Brazil.
- Physiopathology in Aging Laboratory (LIM- 22), Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil.
| | - Alberto F O Justo
- Physiopathology in Aging Laboratory (LIM- 22), Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | - Michelle C A Paula
- Laboratory of Pathology, Department of Pathology, Federal University of Juiz de Fora Medical School, Eugênio Do Nascimento, S/No.-Dom Bosco, Juiz de Fora, MG, 36038 - 330, Brazil
| | - Laura B Grossi
- Laboratory of Pathology, Department of Pathology, Federal University of Juiz de Fora Medical School, Eugênio Do Nascimento, S/No.-Dom Bosco, Juiz de Fora, MG, 36038 - 330, Brazil
| | - Vitor H Neves
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Renata E P Leite
- Physiopathology in Aging Laboratory (LIM- 22), Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | - Vitor R Paes
- Physiopathology in Aging Laboratory (LIM- 22), Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | - Rossana C N Melo
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Ricardo Nitrini
- Department of Neurology, University of São Paulo Medical School, São Paulo, Brazil
| | - Carlos Pasqualucci
- Physiopathology in Aging Laboratory (LIM- 22), Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | - Eduardo Ferriolli
- Division of Geriatrics, Department of Internal Medicine, University of São Paulo Medical School, São Paulo, Brazil
| | - Antonio L Teixeira
- The Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, USA
| | - Lea T Grinberg
- Memory and Aging Center, University of California San Francisco, San Francisco, USA
| | - Claudia K Suemoto
- Physiopathology in Aging Laboratory (LIM- 22), Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
- Division of Geriatrics, Department of Internal Medicine, University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
40
|
Wu W, Lan W, Jiao X, Wang K, Deng Y, Chen R, Zeng R, Li J. Pyroptosis in sepsis-associated acute kidney injury: mechanisms and therapeutic perspectives. Crit Care 2025; 29:168. [PMID: 40270016 PMCID: PMC12020238 DOI: 10.1186/s13054-025-05329-3] [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: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 04/25/2025] Open
Abstract
Sepsis-associated acute kidney injury (S-AKI) is a severe complication characterized by high morbidity and mortality, driven by multi-organ dysfunction. Recent evidence suggests that pyroptosis, a form of programmed cell death distinct from apoptosis and necrosis, plays a critical role in the pathophysiology of S-AKI. This review examines the mechanisms of pyroptosis, focusing on inflammasome activation (e.g., NLRP3), caspase-mediated processes, and the role of Gasdermin D in renal tubular damage. We also discuss the contributions of inflammatory mediators, oxidative stress, and potential therapeutic strategies targeting pyroptosis, including inflammasome inhibitors, caspase inhibitors, and anti-inflammatory therapies. Lastly, we highlight the clinical implications and challenges in translating these findings into effective treatments, underscoring the need for personalized medicine approaches in managing S-AKI.
Collapse
Affiliation(s)
- Wenyu Wu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, 510405, China
| | - Wanning Lan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xin Jiao
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
| | - Kai Wang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yawen Deng
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
| | - Rui Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Research On Emergency in TCM, Guangzhou, Guangdong, China.
| | - Ruifeng Zeng
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Research On Emergency in TCM, Guangzhou, Guangdong, China.
| | - Jun Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
41
|
Horbach N, Kalinka M, Ćwilichowska-Puślecka N, Al Mamun A, Mikołajczyk-Martinez A, Turk B, Snipas SJ, Kasperkiewicz P, Groborz KM, Poręba M. Visualization of calpain-1 activation during cell death and its role in GSDMD cleavage using chemical probes. Cell Chem Biol 2025; 32:603-619.e7. [PMID: 40157358 DOI: 10.1016/j.chembiol.2025.03.002] [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: 09/07/2024] [Revised: 01/30/2025] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
Calpain-1, a calcium-dependent cysteine protease, plays a vital role in cellular processes such as cell death, cytoskeletal remodeling, signal transduction, and cell cycle progression. While its role in apoptosis, including substrate cleavage for orderly disassembly, is well established, its involvement in pyroptosis remains less understood. This study focused on developing chemical tools to detect calpain-1 activity. Using the hybrid combinatorial substrate library (HyCoSuL) approach with unnatural amino acids, we designed fluorescent substrates, inhibitors, and fluorescent activity-based probe (ABP) specific to calpain-1, enabling its visualization in living cells. We further investigated calpain-1's expression alongside cell death proteins in immune cells using mass cytometry and observed strong colocalization with gasdermin D (GSDMD). Additionally, we demonstrated that calpain-1 can hydrolyze GSDMD in vitro. Through fluorescence-based substrate assays and mass spectrometry, we identified putative cleavage sites within the GSDMD sequence that may promote pyroptosis. These findings underscore calpain-1's multifaceted role in cell death pathways, extending beyond apoptosis.
Collapse
Affiliation(s)
- Natalia Horbach
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | - Małgorzata Kalinka
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | | | - Abdulla Al Mamun
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | - Agata Mikołajczyk-Martinez
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland; Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| | - Boris Turk
- Jozef Stefan Institute, Sl-1000 Ljubljana, Slovenia
| | - Scott J Snipas
- SBP Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Paulina Kasperkiewicz
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | - Katarzyna M Groborz
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland; SBP Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Marcin Poręba
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland; Faculty of Medicine, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland.
| |
Collapse
|
42
|
Nie X, Miao S, Hou Y, Ma Y, Li M, Liu Y, Yang Y, Xu J, Wang Y. TLR4-mediated endoplasmic reticulum stress regulates pyroptosis in macrophages infected with the Bacillus Calmette-Guérin mycobacterial. Int Immunopharmacol 2025; 152:114346. [PMID: 40064059 DOI: 10.1016/j.intimp.2025.114346] [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: 11/13/2024] [Revised: 02/11/2025] [Accepted: 02/18/2025] [Indexed: 03/24/2025]
Abstract
Tuberculosis results from Mycobacterium tuberculosis (Mtb) infection. Immune responses controlled by Toll-like receptor 4 (TLR4) are closely associated with the host response to pathogens, including Mtb. NLRP3 inflammasome-mediated pyroptosis forms a significant part of the inflammatory response during Mtb infection, and endoplasmic reticulum stress (ERS) is implicated in the activation of the NLRP3 inflammasome. Here, the function of TLR4 in macrophage pyroptosis induced by infection with the Bacillus Calmette-Guérin (BCG) mycobacterial strain was investigated. It was found that infection with BCG activated TLR4 signaling, induced ERS and subsequent NLRP3 inflammasome activation, leading to pyroptosis in mouse lung tissues. The TLR4 inhibitor TAK 242 inhibited the ERS onset, NLRP3 inflammasome stimulation, and pyroptosis, while the ERS inhibitor TUDCA blocked both inflammasome activation and pyroptosis, and the NLRP3 inhibitor MCC950 specifically inhibited pyroptosis. Furthermore, TAK 242, TUDCA, and MCC950 all exacerbated lung injury caused by BCG infection and promoted BCG survival. Similarly, after in BCG-infected THP-1 macrophages, TLR4 signaling was found to mediate NLRP3 inflammasome activation through ERS, thereby inducing pyroptosis. In summary, BCG infection leads to macrophage pyroptosis via the TLR4/ERS/NLRP3 inflammasome signaling axis, providing new insights for further research into the pathogenesis and treatment of tuberculosis.
Collapse
Affiliation(s)
- Xueyi Nie
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Shen'ao Miao
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Yuxin Hou
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Yabo Ma
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Mengyuan Li
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Yueyang Liu
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Yi Yang
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Jinrui Xu
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China.
| | - Yujiong Wang
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China.
| |
Collapse
|
43
|
Mu XY, Chen SB, Yang SY, Wang WS, Zhou HM, Wang YX, Chen XY, Peng XP, Li WJ. Ganoderma atrum polysaccharide inhibits ROS/NLRP3/pyroptosis axis by fixing mitochondrial dynamics disorder in PD-1 inhibitors-induced carditis of Lewis lung carcinoma mice. Int J Biol Macromol 2025; 310:143163. [PMID: 40246098 DOI: 10.1016/j.ijbiomac.2025.143163] [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: 10/28/2024] [Revised: 03/25/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025]
Abstract
Mitochondria were critical for pathogenesis of PD-1 inhibitors-induced carditis, which was demonstrated to be the mechanism for Ganoderma atrum polysaccharide (PSG) against cardiomyopathy. Hence, the present study aimed to determine the role of PSG in controlling mitochondrial homeostasis in PD-1 inhibitors-induced carditis of Lewis lung carcinoma mice. Results showed that PSG significantly alleviated PD-1 inhibitors-induced cardiotoxicity without compromising their anti-tumor effects, as evidenced by inhibiting cardiac histopathological disorders, creatine kinase (CK) release, and tumor growth. PSG administration significantly ameliorated inflammation by reducing pro-inflammatory cytokine IL-1β release and NLRP3 expression. Meanwhile, the reduction of pyroptosis was demonstrated to be implicated in PSG-inhibited carditis evidenced by the decrease in Caspase-1, gasdermin D (GSDMD). Mechanistically, mitochondria were sites of ROS generation and NLRP3 inflammasome activation. Our results showed that PSG suppressed NLRP3-induced pyroptosis, which was associated with inhibition of ROS attack and mitochondrial protection by maintaining mitochondrial membrane potential, reversing a deficiency in mitochondrial fission, suppressing mitochondrial hyper-fusion, suggesting that ROS/NLRP3/pyroptosis axis was a vital process in avoiding mitochondrial dysfunction during PSG-mediated cardioprotection. Additionally, the modification of the redox system was also shown in the context of cardioprotection of PSG, by elevating antioxidant enzyme activities and suppressing lipid oxidation.
Collapse
Affiliation(s)
- Xiao-Yu Mu
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Sheng-Bin Chen
- Affiliated Rehabilitation Hospital of Nanchang University, 999 Xuefu Boulevard, Nanchang 330031, China
| | - Song-Yu Yang
- Affiliated Rehabilitation Hospital of Nanchang University, 999 Xuefu Boulevard, Nanchang 330031, China
| | - Wen-Sheng Wang
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Hong-Mei Zhou
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Yi-Xuan Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Xuan-Ying Chen
- Department of Pharmacy, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Xiao-Ping Peng
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Wen-Juan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| |
Collapse
|
44
|
Kusunoki Y, Li C, Long H, Watanabe-Kusunoki K, Kuang M, Marschner JA, Linkermann A, Steiger S, Anders HJ. Gasdermin D deficiency aggravates nephrocalcinosis-related chronic kidney disease with rendering macrophages vulnerable to necroptosis. Cell Death Dis 2025; 16:283. [PMID: 40221396 PMCID: PMC11993636 DOI: 10.1038/s41419-025-07620-1] [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: 12/12/2024] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
Several forms of regulated necrosis contribute to the pathogenesis of crystal nephropathy, however, the role of pyroptosis, an inflammatory form of cell death involving the formation of gasdermin-D pores in internal and external cell membranes, in this condition remains unknown. Our transcriptional and histological analyses suggest that Gsdmd in tubulointerstitital cells may contribute to the pathogenesis of chronic oxalate nephropathy. However, genetic deletion of Gsdmd exacerbated oxalate nephropathy in mice in association with enhanced CaOx crystal deposition and accelerated tubular epithelial cell injury. Pharmacological inhibition of necroptosis reversed this effect. Indeed, Gsdmd-/- bone marrow-derived macrophages were more prone to undergo necroptosis when stimulated with CaOx crystals compared to their wildtype counterparts. We conclude that gasdermin D suppresses the necroptosis pathway, which determines the outcome of oxalate nephropathy-related nephrocalcinosis.
Collapse
Affiliation(s)
- Yoshihiro Kusunoki
- Renal Division, Department of Medicine IV, Hospital of the Ludwig-Maximilians-University, Munich, Germany
- Department of Rheumatology, Endocrinology, and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Chenyu Li
- Renal Division, Department of Medicine IV, Hospital of the Ludwig-Maximilians-University, Munich, Germany
- Department of Medicine, Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hao Long
- Renal Division, Department of Medicine IV, Hospital of the Ludwig-Maximilians-University, Munich, Germany
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Kanako Watanabe-Kusunoki
- Renal Division, Department of Medicine IV, Hospital of the Ludwig-Maximilians-University, Munich, Germany
- Department of Rheumatology, Endocrinology, and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Meisi Kuang
- Renal Division, Department of Medicine IV, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | | | - Andreas Linkermann
- Department of Medicine V, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Stefanie Steiger
- Renal Division, Department of Medicine IV, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Hans-Joachim Anders
- Renal Division, Department of Medicine IV, Hospital of the Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
45
|
Carnazzo V, Rigante D, Restante G, Basile V, Pocino K, Basile U. The entrenchment of NLRP3 inflammasomes in autoimmune disease-related inflammation. Autoimmun Rev 2025; 24:103815. [PMID: 40233890 DOI: 10.1016/j.autrev.2025.103815] [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: 02/17/2025] [Revised: 03/24/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025]
Abstract
Autoinflammation and autoimmunity are almost "opposite" phenomena characterized by chronic activation of the immune system, 'innate' in the first and 'adaptive' in the second, leading to inflammation of several tissues with specific protean effectors of tissue damage. The mechanism of involvement of multiprotein complexes called 'inflammasomes' within autoimmune pictures, differently from autoinflammatory conditions, is yet undeciphered. In this review we provide a comprehensive overview on NLRP3 inflammasome contribution into the pathogenesis of some autoimmune diseases. In response to autoantibodies against nucleic acids or tissue-specific antigens the NLRP3 inflammasome is activated within dendritic cells and macrophages of patients with systemic lupus erythematosus. Crucial is NLRP3 inflammasome to amplify tissue inflammation with interleukin-1 overexpression and matrix metalloproteinase production at the joint level in rheumatoid arthritis. A deregulated NLRP3 inflammasome activation occurs in the serous acini of salivary and lacrimal glands prone to Sjogren's syndrome, but also in the inflammatory process involving endothelial cells, leucocyte recruitment, and platelet plugging of vasculitides. Furthermore, organ-specific autoimmune diseases such as thyroiditis and hepatitis may display hyperactive NLRP3 inflammasomes at the level of resident immune cells within thyroid or liver, respectively. Therefore, it is not unexpected that preclinical studies have shown how specific inflammasome inhibitors may significantly overthrow the severity of different autoimmune diseases and slow down their trend towards an ominous progression. Specific markers of inflammasome activation could also reveal subclinical inflammatory components escaping conventional diagnostic approaches or improve monitoring of autoimmune diseases and personalizing their treatment.
Collapse
Affiliation(s)
- Valeria Carnazzo
- Department of Clinical Pathology, Santa Maria Goretti Hospital, Latina, Italy.
| | - Donato Rigante
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Università Cattolica Sacro Cuore, Rome, Italy.
| | - Giuliana Restante
- Department of Experimental Medicine, University "La Sapienza", Rome, Italy
| | - Valerio Basile
- Clinical Pathology Unit and Cancer Biobank, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Krizia Pocino
- Unit of Clinical Pathology, Ospedale San Pietro Fatebenefratelli, Rome, Italy
| | - Umberto Basile
- Department of Clinical Pathology, Santa Maria Goretti Hospital, Latina, Italy.
| |
Collapse
|
46
|
Zhang Z, Rana I, Nam J. Metal coordination polymer nanoparticles for cancer therapy. Essays Biochem 2025; 69:EBC20253012. [PMID: 40209056 DOI: 10.1042/ebc20253012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/24/2025] [Indexed: 04/12/2025]
Abstract
Metal ions are essential elements in biological processes and immune homeostasis. They can regulate cancer cell death through multiple distinct molecular pathways and stimulate immune cells implicated in antitumor immune responses, suggesting opportunities to design novel metal ion-based cancer therapies. However, their small size and high charge density result in poor target cell uptake, uncontrolled biodistribution, and rapid clearance from the body, reducing therapeutic efficacy and increasing potential off-target toxicity. Metal coordination polymer nanoparticles (MCP NPs) are nanoscale polymer networks composed of metal ions and organic ligands linked via noncovalent coordination interactions. MCP NPs offer a promising nanoplatform for reshaping metal ions into more drug-like formulations, improving their in vivo pharmacological performance and therapeutic index for cancer therapy applications. This review provides a comprehensive overview of the inherent biological functions of metal ions in cancer therapy, showcasing examples of MCP NP systems designed for preclinical cancer therapy applications where drug delivery principles play a critical role in enhancing therapeutic outcomes. MCP NPs offer versatile metal ion engineering approaches using selected metal ions, various organic ligands, and functional payloads, enabling on-demand nano-drug designs that can significantly improve therapeutic efficacy and reduce side effects for effective cancer therapy.
Collapse
Affiliation(s)
- Zhengzheng Zhang
- College of Pharmacy, Chonnam National University, Gwanju 61186, South Korea
| | - Isra Rana
- College of Pharmacy, Chonnam National University, Gwanju 61186, South Korea
| | - Jutaek Nam
- College of Pharmacy, Chonnam National University, Gwanju 61186, South Korea
| |
Collapse
|
47
|
Zhang H, Li X, Lin L. Biological Functions and Clinical Implications of CFLAR: From Cell Death Mechanisms to Therapeutic Targeting in Immune Regulation. J Inflamm Res 2025; 18:4911-4928. [PMID: 40224389 PMCID: PMC11994107 DOI: 10.2147/jir.s519885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025] Open
Abstract
Since its initial functional characterization in the late 1990s, CASP-8 and FADD-like apoptosis regulator (CFLAR) has been recognized as a crucial regulator of both apoptosis and immune responses. CFLAR inhibits caspase-8 activation by forming heterodimers with procaspase-8 at the death-inducing signaling complex (DISC), thereby preventing its proteolytic maturation. In addition to its role in cell death, CFLAR is integral to immune regulation, modulating NF-κB-dependent cytokine production (eg, IL-1β, TNF-α) and effector functions of T cells and macrophages. Recent studies underscore the pathological significance of dysregulated CFLAR expression in a variety of diseases, including cancers and inflammatory conditions. Within the tumor microenvironment, elevated CFLAR expression confers resistance to therapy, while in infectious and inflammatory diseases, its expression levels modulate the magnitude and direction of the immune response. This review provides an in-depth exploration of CFLAR's structural and functional properties, focusing on its involvement in apoptosis, autophagy, and immune modulation. Moreover, we examine its translational potential as a therapeutic target, evidenced by ongoing preclinical studies targeting CFLAR isoforms in cancer immunotherapy. By synthesizing recent advances in CFLAR's dual roles in cell death and immune surveillance, this review highlights actionable targets for overcoming therapy resistance and immune dysregulation.
Collapse
Affiliation(s)
- Haiyang Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| | - Xin Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| | - Liangkang Lin
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Department of Pediatrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| |
Collapse
|
48
|
Cabrera Ranaldi EDLRM, Bramlett HM, Umland O, Levine LI, Keane RW, de Rivero Vaccari JP, Dietrich WD, Kerr NA. Gasdermin-D Genetic Knockout Reduces Inflammasome-Induced Disruption of the Gut-Brain Axis After Traumatic Brain Injury. Int J Mol Sci 2025; 26:3512. [PMID: 40331993 PMCID: PMC12027180 DOI: 10.3390/ijms26083512] [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: 02/21/2025] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Traumatic brain injury (TBI) pathology is significantly mediated by an inflammatory response involving inflammasome activation, resulting in the release of interleukin (IL)-1β and pyroptotic cell death through gasdermin-D (GSDMD) cleavage. Inflammasome components are transported through extracellular vesicles (EVs) to mediate systemic inflammation in peripheral organs, including the gut. The purpose of this study was to determine the protective effect of GSDMD knockout (KO) on TBI-induced inflammasome activation, EV signaling, and gut function. GSDMD-KO and C57BL6 (WT) mice were subjected to the controlled cortical impact model of TBI. Cytokine expression was assessed with electrochemiluminescent immunoassay and immunoblotting of the cerebral cortex and gut. EVs were examined for pathology-associated markers using flow cytometry, and gut permeability was determined. GSDMD-KO attenuated IL-1β and IL-6 expression in the cerebral cortex and reduced IL-1β and IL-18 in the gut 3 days post-injury. GSDMD-KO mice had decreased neuronal- and gut-derived EVs compared to WT mice post-TBI. GSDMD-KO EVs also had decreased IL-1β and different surface marker expression post-TBI. GSDMD-KO mice had decreased gut permeability after TBI. These data demonstrate that GSDMD ablation improves post-TBI inflammation and gut pathology, suggesting that GSDMD may serve as a potential therapeutic target for the improvement of TBI-associated pathologies.
Collapse
Affiliation(s)
- Erika d. l. R. M. Cabrera Ranaldi
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.d.l.R.M.C.R.); (H.M.B.); (L.I.L.); (R.W.K.); (J.P.d.R.V.); (W.D.D.)
| | - Helen M. Bramlett
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.d.l.R.M.C.R.); (H.M.B.); (L.I.L.); (R.W.K.); (J.P.d.R.V.); (W.D.D.)
- Bruce W. Carter Department of Veteran Affairs Medical Center, Miami, FL 33136, USA
| | - Oliver Umland
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Leo I. Levine
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.d.l.R.M.C.R.); (H.M.B.); (L.I.L.); (R.W.K.); (J.P.d.R.V.); (W.D.D.)
| | - Robert W. Keane
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.d.l.R.M.C.R.); (H.M.B.); (L.I.L.); (R.W.K.); (J.P.d.R.V.); (W.D.D.)
- Department of Cellular Physiology and Molecular Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.d.l.R.M.C.R.); (H.M.B.); (L.I.L.); (R.W.K.); (J.P.d.R.V.); (W.D.D.)
- Department of Cellular Physiology and Molecular Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - W. Dalton Dietrich
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.d.l.R.M.C.R.); (H.M.B.); (L.I.L.); (R.W.K.); (J.P.d.R.V.); (W.D.D.)
| | - Nadine A. Kerr
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.d.l.R.M.C.R.); (H.M.B.); (L.I.L.); (R.W.K.); (J.P.d.R.V.); (W.D.D.)
| |
Collapse
|
49
|
Xu H, Wu M, Wang Y, Jiao Y, Chen Y, Yuan Z, Sun L. Teleost GSDMEc regulates GSDMEa-mediated pyroptosis. J Adv Res 2025:S2090-1232(25)00226-7. [PMID: 40210150 DOI: 10.1016/j.jare.2025.04.007] [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: 01/20/2025] [Revised: 03/24/2025] [Accepted: 04/04/2025] [Indexed: 04/12/2025] Open
Abstract
INTRODUCTION Gasdermin (GSDM) is a family of proteins that execute pyroptosis after being activated by caspase (CASP) cleavage. Mammals possess five GSDM members (A - E) with pyroptotic ability. Teleosts possess only one pyroptotic GSDM, GSDME, that exists in three orthologs, GSDMEa, b, and c. GSDMEa and GSDMEb are known to induce pyroptosis, but the function of GSDMEc is unknown. OBJECTIVES The present study aimed to elucidate the function of teleost GSDMEc and examine the interplay among teleost GSDME orthologs by using snakehead Channa argus as a representative species. METHODS Pyroptosis was assessed via microscopy and biochemical assays. GSDME cleavage, oligomerization, and membrane translocation were examined via immunoblotting. The interactions of GSDME products were examined using confocal microscopy and co-immunoprecipitation. GSDME knockdown in fish and in vivo bacterial infection were performed. RESULTS C. argus possessed three GSDME variants (CaGSDMEa, CaGSDMEc1, and CaGSDMEc2). CaGSDMEa was cleaved by C. argus CASP (CaCASP) 1/8 to produce an N-terminal fragment (NT), NT261, that induced pyroptosis. CaGSDMEc1 and CaGSDMEc2 were also cleaved by CaCASP1/8, but the resulting NTs, NT123 and NT108, respectively, were unable to induce pyroptosis. However, both NT123 and NT108 could bind and promote the pyroptotic activity of NT261 by facilitating NT261 oligomerization and membrane translocation. The interaction between NT261 and NT123/NT108 depended on a positively charged motif that is conserved in the metazoan GSDME and is essential to the membrane localization of NT123 and the pyroptotic activity of NT261. Bacterial infection induced CaGSDMEa/CaCASP8 activation and CaGSDMEc1/c2 cleavage in snakehead cells, resulting in pyroptosis, IL-1β/18 maturation cleavage, and extracellular DNA-net formation. CaGSDMEa/c1 knockdown significantly increased bacterial dissemination in fish tissues and reduced fish survival. CONCLUSIONS Our results revealed the functions and interactive mechanism of teleost GSDME orthologs, and provided new insights into the regulation of pyroptosis in lower vertebrates.
Collapse
Affiliation(s)
- Hang Xu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Meng Wu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; College of Marine Sciences, University of Chinese Academy of Sciences, Qingdao, China
| | - Yujian Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Yaoming Jiao
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; College of Marine Sciences, University of Chinese Academy of Sciences, Qingdao, China
| | - Yuan Chen
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; College of Marine Sciences, University of Chinese Academy of Sciences, Qingdao, China
| | - Zihao Yuan
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; College of Marine Sciences, University of Chinese Academy of Sciences, Qingdao, China.
| | - Li Sun
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; College of Marine Sciences, University of Chinese Academy of Sciences, Qingdao, China.
| |
Collapse
|
50
|
Zhang N, Xu D. Controlling pyroptosis through post-translational modifications of gasdermin D. Dev Cell 2025; 60:994-1007. [PMID: 40199241 DOI: 10.1016/j.devcel.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/15/2024] [Accepted: 02/05/2025] [Indexed: 04/10/2025]
Abstract
Pyroptosis, a lytic and programmed cell death pathway, is mediated by gasdermins (GSDMs), with GSDMD playing an important role in innate immunity and pathology. Upon activation, GSDMD is cleaved to release the active N-terminal fragment that oligomerizes into membrane pores, which promote pyroptosis and cytokine secretion, leading to inflammation. Emerging evidence indicates that post-translational modification (PTM) is an important regulatory mechanism of GSDMD activity. This review explores how PTMs, aside from proteolytic cleavage, control GSDMD activity and link biological contexts to pyroptosis in innate immunity and inflammation, which could inform future studies and therapeutic solutions for treating inflammatory conditions.
Collapse
Affiliation(s)
- Na Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Daichao Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China; Shanghai Key Laboratory of Aging Studies, Shanghai 201210, China; Shanghai Academy of Natural Sciences (SANS), Fudan University, Shanghai 200031, China.
| |
Collapse
|