1
|
Zhang X, Yin Y, Chen Y, Lin L, Shen S, Fang F, Wang Q. Gut microbiota contributes to obstructive sleep apnea-induced hypertension by gut-heart axis in mice. Int Immunopharmacol 2025; 155:114667. [PMID: 40245774 DOI: 10.1016/j.intimp.2025.114667] [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: 02/07/2025] [Revised: 03/29/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND The gut microbiome has been closely linked to obstructive sleep apnea (OSA)-associated hypertension (HTN). However, its precise role in the pathogenesis of OSA-induced HTN remains unclear. METHODS To clarify the causal relationship between gut dysbiosis and OSA-related HTN, C57BL6J mice were randomly assigned to four groups. Each group underwent fecal microbiota transplantation from healthy individuals (control), OSA patients (OSA group), OSA patients with pre-hypertension (OSA-pHTN group), or OSA patients with HTN (OSA-HTN group). The pro-hypertensive effects of the OSA gut microbiota were verified, and the composition and function of the gut microbiota were compared using 16S rDNA gene sequencing. Additionally, the gut microbiota-related lipopolysaccharide (LPS)/ Toll-like receptor 4 (TLR4)/nuclear factor-kappaB (NF-κB) pathway in aortic tissues was investigated. RESULTS Fecal microbiota transplantation induced increased systolic blood pressure and aortic injury in mice from the OSA, OSA-pHTN and OSA-HTN groups, whereas no significant injury was observed in the control group. These three groups exhibited dysbiosis and impaired intestinal barrier function as evidenced by a reduction in Akkermansia and decreased expression of zonula occludens-1 and Occludin proteins. In addition, LPS, TLR4 and phosphorylated NF-κB expression were increased in aortic tissue from the three groups, and immunofluorescence showed a significant upregulation of TLR4 expression in aortic endothelial cells compared to controls. CONCLUSION This study demonstrates the pro-hypertensive effects of gut microbiota in OSA, mediated through the gut-derived LPS/TLR4/NF-κB pathway. These findings may guide the development of therapeutic strategies focused on restoring gut microbiome homeostasis.
Collapse
Affiliation(s)
- Xiaotong Zhang
- Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuang Ta East Street, Taiyuan 030001, China
| | - Yanran Yin
- Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuang Ta East Street, Taiyuan 030001, China
| | - Yongjun Chen
- Department of Pulmonary and Critical Care, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuang Ta East Street, Taiyuan 030001, China
| | - Linghang Lin
- Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuang Ta East Street, Taiyuan 030001, China
| | - Si Shen
- Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuang Ta East Street, Taiyuan 030001, China
| | - Fan Fang
- Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuang Ta East Street, Taiyuan 030001, China
| | - Qiang Wang
- Department of Infectious Disease, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuang Ta East Street, Taiyuan 030001, China.
| |
Collapse
|
2
|
Bruce JK, Li LY, Tang Y, Forster EG, Winsor NJ, Bi PY, Krustev C, Keely S, Lee JE, Rohde JR, Gaisano HY, Philpott DJ, Girardin SE. Gasdermin-D pores induce an inactivating caspase-4 cleavage that limits IL-18 production in the intestinal epithelium. Commun Biol 2025; 8:737. [PMID: 40355718 PMCID: PMC12069520 DOI: 10.1038/s42003-025-08183-9] [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: 08/21/2023] [Accepted: 05/06/2025] [Indexed: 05/14/2025] Open
Abstract
Intestinal epithelial-derived IL-18 is critical for homeostatic intestinal barrier function and is secreted through Gasdermin D (GSDMD) pores. Inflammasome activation is a prerequisite for both IL-18 maturation and GSDMD pore formation. However, GSDMD pores also cause pyroptotic cell death, which could be detrimental to the intestinal epithelial barrier. How epithelial cells balance the need to secrete IL-18 and to maintain barrier integrity remains poorly understood. In human intestinal epithelial cell lines and in primary human epithelial intestinal organoids, but not in immune cells, GSDMD plasma membrane pore formation by LPS electroporation and by gram-negative bacterial infection induced a non-conventional p37 caspase-4 fragment that was associated with reduced levels of mature IL-18. By contrast, limiting GSDMD plasma membrane pores pharmacologically and via point-mutagenesis prevented caspase-4 cleavage and increased IL-18 production, suggesting that p37 caspase-4 cleavage may regulate IL-18 maturation in the intestinal epithelium. In support, co-expression of caspase-4 cleavage mutants and IL-18 in HEK293T cells revealed that non-cleavable caspase-4 produced more mature IL-18 than cleaved caspase-4. Overall, these studies suggest that epithelial inflammasomes encode feedback pathways that control the balance between cytokine secretion and cell death. This may be an important mechanism to ensure homeostatic IL-18 production in the intestinal epithelium.
Collapse
Affiliation(s)
- J K Bruce
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- School of Biomedical Sciences & Pharmacy, University of Newcastle, Newcastle, NSW, Australia
| | - L Y Li
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Y Tang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - E G Forster
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - N J Winsor
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - P Y Bi
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - C Krustev
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - S Keely
- School of Biomedical Sciences & Pharmacy, University of Newcastle, Newcastle, NSW, Australia
| | - J E Lee
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - J R Rohde
- Department of Microbiology and Immunology Dalhousie University, Halifax, NS, Canada
| | - H Y Gaisano
- Department of Medicine and Physiology, University of Toronto, Toronto, ON, Canada
| | - D J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - S E Girardin
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Zhang X, Shi B, Zhao Z, Deng Y, Zhou X, Hu J. Deciphering the Transcriptomic Complexity of Yak Skin Across Different Ages and Body Sites. Int J Mol Sci 2025; 26:4601. [PMID: 40429746 PMCID: PMC12111109 DOI: 10.3390/ijms26104601] [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: 02/24/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Differences in skin and hair phenotypes between the scapular and ventral regions of yaks (Bos grunniens) are obvious and become more prominent with age. However, the genetic mechanism that causes differences in yak skin at different ages has not been reported. In this study, we investigated the transcriptomic profile of yak skin across different ages (0.5 years, 2.5 years, and 4.5 years) and body sites (scapular and ventral regions). Differential gene expression analysis was initially conducted to explore the transcriptomic differences in skin at different ages and different body sites. Subsequently, weighted gene co-expression network analysis (WGCNA) was employed to analyze the transcriptomic data comprehensively. The results showed that, among all comparison groups, the Y2.5_S vs. Y2.5_V group (regional comparison) exhibited the highest number of DEGs, with 491 genes (179 upregulated and 312 downregulated), followed by the Y2.5_V vs. Y0.5_V group (age comparison), which had 370 DEGs (103 upregulated and 267 downregulated). DEGs such as IGF2BP3, ADCY8, FOSL1, and S100A8 were found in all comparison groups of different ages, and multiple members of the HOX gene family including HOXC10, HOXA9, HOXA6, HOXB9, and HOXB6 were differentially expressed in comparison groups at different sites. Functional enrichment analysis showed that there were more obvious differences in immune function between different ages of skin and more obvious differences in endocrine function between different parts of skin. WGCNA revealed that genes related with immunity such as GLYATL2, ACSL5, and SPDEF were the core genes of the co-expression module associated with the scapula region, and multiple genes related to hair follicle development such as FOXN1, OVOL1, DLX3, HOXC13, and TCHH were found to be the hub genes of the co-expression module associated with the ventral region. Overall, our study provides valuable insights into the transcriptomic complexity of yak skin across different ages and body sites. The differential gene expression patterns and co-expression network modules identified in this study lay the foundation for further research on skin biology and adaptation mechanisms in yaks.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiang Hu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (X.Z.); (B.S.); (Z.Z.); (Y.D.); (X.Z.)
| |
Collapse
|
4
|
Min R, Bai Y, Wang NR, Liu X. Gasdermins in pyroptosis, inflammation, and cancer. Trends Mol Med 2025:S1471-4914(25)00090-5. [PMID: 40307076 DOI: 10.1016/j.molmed.2025.04.003] [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: 11/23/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Pyroptosis is a type of programmed inflammatory cell death characterized by balloon-like swelling, membrane rupture, and the release of inflammatory cytokines and danger signals. Pyroptosis is directly triggered by activated gasdermins (GSDMs) which bind to membrane phospholipids, oligomerize, and form pores in cell membranes. GSDM activation is mediated by various effector proteases via cleavage of the linker region or post-translational modification to release the active N-terminal fragment in response to a variety of pathogenic or intrinsic danger signals. GSDM-mediated pyroptosis is involved in the pathogenesis of an array of infectious and inflammatory diseases and cancers. This review discusses recent advances related to the physiological and pathological functions of GSDM-mediated pyroptosis, as well as therapeutic strategies targeting pyroptosis.
Collapse
Affiliation(s)
- Rui Min
- National Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Bai
- National Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ning-Rui Wang
- School of Laboratory Medicine, Nanchang Medical College, Nanchang, Jiangxi 330052, China
| | - Xing Liu
- National Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; Shanghai Academy of Natural Sciences (SANS), Shanghai 200031, China.
| |
Collapse
|
5
|
Bai Y, Pan Y, Liu X. Mechanistic insights into gasdermin-mediated pyroptosis. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00837-0. [PMID: 40128620 DOI: 10.1038/s41580-025-00837-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2025] [Indexed: 03/26/2025]
Abstract
Pyroptosis, a novel mode of inflammatory cell death, is executed by membrane pore-forming gasdermin (GSDM) family members in response to extracellular or intracellular injury cues and is characterized by a ballooning cell morphology, plasma membrane rupture and the release of inflammatory mediators such as interleukin-1β (IL-1β), IL-18 and high mobility group protein B1 (HMGB1). It is a key effector mechanism for host immune defence and surveillance against invading pathogens and aberrant cancerous cells, and contributes to the onset and pathogenesis of inflammatory and autoimmune diseases. Manipulating the pore-forming activity of GSDMs and pyroptosis could lead to novel therapeutic strategies. In this Review, we discuss the current knowledge regarding how GSDM-mediated pyroptosis is initiated, executed and regulated, its roles in physiological and pathological processes, and the crosstalk between different modes of programmed cell death. We also highlight the development of drugs that target pyroptotic pathways for disease treatment.
Collapse
Affiliation(s)
- Yang Bai
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Youdong Pan
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xing Liu
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China.
- Shanghai Academy of Natural Sciences (SANS), Shanghai, China.
| |
Collapse
|
6
|
Zhao T, Chi Z, Wang D. Versatility of gasdermin D beyond pyroptosis. Trends Cell Biol 2025:S0962-8924(25)00061-3. [PMID: 40121145 DOI: 10.1016/j.tcb.2025.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/14/2025] [Accepted: 02/21/2025] [Indexed: 03/25/2025]
Abstract
Gasdermin D (GSDMD) has garnered significant attention primarily for the pore-forming role of its p30 N-terminal fragment (NT-p30) generated during pyroptosis, a proinflammatory form of cell death. However, emerging evidence suggests that the formation of GSDMD-NT pores is reversible, and the activation of GSDMD does not necessarily lead to pyroptosis. Instead, this process may take part either in other forms of cell death, or in various state changes of living cells, including (i) inflammation regulation, (ii) endolysosomal pathway rewiring, (iii) granule exocytosis, (iv) type II immunity, (v) food tolerance maintenance, and (vi) temporary permeability alteration. This review explores the latest insights into the involvement of GSDMD in cell death and homeostasis maintenance, aiming to underscore the pleiotropic nature of GSDMD.
Collapse
Affiliation(s)
- Tianming Zhao
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 310058, China
| | - Zhexu Chi
- Center for Regeneration and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Yiwu 322000, China.
| | - Di Wang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 310058, China.
| |
Collapse
|
7
|
Fang J, Zhu W, Yu D, Zhu L, Zha H, Tang J, Li Y, Zhu X, Zhao T, Zhang W. From Inflammasomes to Pyroptosis: Molecular Mechanisms in Chronic Intestinal Diseases - Opportunity or Challenge? J Inflamm Res 2025; 18:3349-3360. [PMID: 40070928 PMCID: PMC11895680 DOI: 10.2147/jir.s498703] [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: 10/08/2024] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Pyroptosis is a unique form of programmed cell death characterized by intense inflammation. It involves the activation of Gasdermin proteins, which form membrane pores, leading to rapid cell rupture and the release of inflammatory molecules. Unlike other types of cell death, pyroptosis has distinct activation mechanisms and plays a complex role in chronic intestinal diseases, including inflammatory bowel disease, intestinal fibrosis, chronic infectious enteritis, and colorectal cancer. This review comprehensively examines how pyroptosis influences disease development and progression while exploring the therapeutic potential of targeting pyroptosis-related pathways. Moreover, the complex interplay between gut microbiota and pyroptosis is summarized, highlighting its critical role in the pathogenesis of chronic intestinal disorders. A deeper understanding of pyroptosis-related mechanisms in these diseases may provide valuable insights for future research and contribute to the development of innovative therapeutic strategies in gastroenterology.
Collapse
Affiliation(s)
- Jintao Fang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, People’s Republic of China
| | - Weihan Zhu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, People’s Republic of China
| | - Dian Yu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, People’s Republic of China
| | - Lujian Zhu
- Department of Infectious Diseases, Jinhua Municipal Central Hospital, Jinhua, 321000, People’s Republic of China
| | - Haorui Zha
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, People’s Republic of China
| | - Jingyi Tang
- Lanxi Hospital of Traditional Chinese Medicine, Jinhua, Zhejiang, 321100, People’s Republic of China
| | - Yujia Li
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, People’s Republic of China
| | - Xiaxin Zhu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, People’s Republic of China
| | - Ting Zhao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, People’s Republic of China
| | - Wei Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, People’s Republic of China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000, People’s Republic of China
| |
Collapse
|
8
|
Wu Y, Sun J, Xie W, Xue S, Li X, Guo J, Shan J, Peng G, Zheng Y. Immunomodulation of Glycyrrhiza Polysaccharides In Vivo Based on Microbiome and Metabolomics Approaches. Foods 2025; 14:874. [PMID: 40077577 PMCID: PMC11898905 DOI: 10.3390/foods14050874] [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: 02/07/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Glycyrrhiza uralensis Fisch. is a medicinal herb that can be added to food to provide therapeutic effects and reduce the burden of medications. Herein, the immunomodulatory effects of Glycyrrhiza polysaccharides (GPs) were verified and illustrated by intervening immunocompromised rats treated with different doses of GPs, which were reflected for adjusting the composition and structure of the intestinal microbiota and altering the metabolic profile. The immunomodulatory effects of GPs were exerted by regulating the intestinal microenvironment. In particular, GPs could promote the growth of probiotic bacteria Allobaculum, norank__o_Clostridia_UCG-014, Dubosiella, and g__norank_o___RF39 and curb the growth of harmful bacteria Enterococcus. The results showed that GPs had a prebiotic effect, which contributed to improving the intestinal environment and maintaining intestinal health. In addition, the content of beneficial differential metabolites was up-regulated, especially short-chain fatty acids, with alanine, aspartate, and glutamate metabolism; arginine biosynthesis; glyoxylate and dicarboxylate metabolism being the most enriched pathways. These metabolic pathways imply the metabolic process of GPs, and the metabolic pathways and differential effector metabolites of it are focused. Overall, the purpose of this article lies in providing support for the application of GPs for regulating immune function.
Collapse
Affiliation(s)
- Yixuan Wu
- Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China; (Y.W.); (J.S.); (W.X.); (S.X.); (X.L.); (G.P.)
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing 211100, China
- Jiangsu Province Engineering Research Center of Classical Prescription, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jie Sun
- Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China; (Y.W.); (J.S.); (W.X.); (S.X.); (X.L.); (G.P.)
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing 211100, China
- Jiangsu Province Engineering Research Center of Classical Prescription, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wenjie Xie
- Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China; (Y.W.); (J.S.); (W.X.); (S.X.); (X.L.); (G.P.)
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing 211100, China
- Jiangsu Province Engineering Research Center of Classical Prescription, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Simin Xue
- Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China; (Y.W.); (J.S.); (W.X.); (S.X.); (X.L.); (G.P.)
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing 211100, China
- Jiangsu Province Engineering Research Center of Classical Prescription, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xinli Li
- Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China; (Y.W.); (J.S.); (W.X.); (S.X.); (X.L.); (G.P.)
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing 211100, China
- Jiangsu Province Engineering Research Center of Classical Prescription, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jianming Guo
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China;
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Guoping Peng
- Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China; (Y.W.); (J.S.); (W.X.); (S.X.); (X.L.); (G.P.)
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing 211100, China
- Jiangsu Province Engineering Research Center of Classical Prescription, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Yunfeng Zheng
- Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China; (Y.W.); (J.S.); (W.X.); (S.X.); (X.L.); (G.P.)
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing 211100, China
- Jiangsu Province Engineering Research Center of Classical Prescription, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| |
Collapse
|
9
|
Li Y, Guo B. GSDMD-mediated pyroptosis: molecular mechanisms, diseases and therapeutic targets. MOLECULAR BIOMEDICINE 2025; 6:11. [PMID: 39994107 PMCID: PMC11850691 DOI: 10.1186/s43556-025-00249-8] [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: 08/22/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
Pyroptosis is a regulated form of inflammatory cell death in which Gasdermin D (GSDMD) plays a central role as the key effector molecule. GSDMD-mediated pyroptosis is characterized by complex biological features and considerable heterogeneity in its expression, mechanisms, and functional outcomes across various tissues, cell types, and pathological microenvironments. This heterogeneity is particularly pronounced in inflammation-related diseases and tumors. In the context of inflammatory diseases, GSDMD expression is typically upregulated, and its activation in macrophages, neutrophils, T cells, epithelial cells, and mitochondria triggers both pyroptotic and non-pyroptotic pathways, leading to the release of pro-inflammatory cytokines and exacerbation of tissue damage. However, under certain conditions, GSDMD-mediated pyroptosis may also serve a protective immune function. The expression of GSDMD in tumors is regulated in a more complex manner, where it can either promote immune evasion or, in some instances, induce tumor cell death. As our understanding of GSDMD's role continues to progress, there have been advancements in the development of inhibitors targeting GSDMD-mediated pyroptosis; however, these therapeutic interventions remain in the preclinical phase. This review systematically examines the cellular and molecular complexities of GSDMD-mediated pyroptosis, with a particular emphasis on its roles in inflammation-related diseases and cancer. Furthermore, it underscores the substantial therapeutic potential of GSDMD as a target for precision medicine, highlighting its promising clinical applications.
Collapse
Affiliation(s)
- Yujuan Li
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China.
| | - Bin Guo
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| |
Collapse
|
10
|
Gong W, Liu P, Liu J, Li Y, Jiang H, Li W, Kang J, Jiao F, Wu X, Zhao Y, Ren J. Gasdermin B modulates intestinal epithelial homeostasis via regulating hyperactive unfolded protein response in Crohn's disease. J Crohns Colitis 2025; 19:jjaf012. [PMID: 39831701 DOI: 10.1093/ecco-jcc/jjaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Impaired intestinal epithelial barrier has been considered to be associated with an increasing variety of gastrointestinal diseases, especially inflammatory bowel disease (IBD) encompassing Crohn's disease (CD) and ulcerative colitis (UC). We aimed to investigate the role of Gasdermin B (GSDMB) in modulating intestinal epithelial barrier integrity and proposed a promising therapeutic strategy. METHODS Gasdermin B expression was evaluated in adult CD samples by molecular biology means and single-cell transcriptomes. We generated GSDMB (Rosa26-lsl/lsl-GSDMB;Villin-Cre) and one of its functional missense variant rs2305480 (Rosa26-lsl/lsl-GSDMB-MU;Villin-Cre) intestinal epithelial-specific knock in mice to observe the functions of GSDMB in intestinal epithelial barrier. RNA-seq analysis as well as human and murine intestine-derived organoids were used to determine the pathogenic mechanism of GSDMB. RESULTS The expression of GSDMB was increased during active intestinal inflammation and principally localized in intestinal epithelial cells (IECs). Rosa26-lsl/lsl-GSDMB;Villin-Cre mice developed enterocolitis and exhibited aberrant intestinal barrier integrity. Mechanistically, epithelial GSDMB modulated hyperactive unfolded protein response of IECs by up-regulating BHLHA15 to mediate intestinal barrier injury. Rosa26-lsl/lsl-GSDMB-MU;Villin-Cre mice with the mutant rs2305480 of GSDMB aggravated such inflammatory effects. CONCLUSION We have uncovered an important and previously unrecognized role of GSDMB in intestinal homeostasis, which represents a potential therapeutic target for intestinal inflammation.
Collapse
Affiliation(s)
- Wenbin Gong
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Peizhao Liu
- Medical School of Nanjing University, Department of General Surgery, Jinling Hospital, Nanjing 210002, China
| | - Juanhan Liu
- Medical School of Nanjing University, Department of General Surgery, Jinling Hospital, Nanjing 210002, China
| | - Yangguang Li
- Medical School of Nanjing University, Department of General Surgery, Jinling Hospital, Nanjing 210002, China
| | - Haiyang Jiang
- BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210019, China
| | - Weizhen Li
- Medical School of Nanjing University, Department of General Surgery, Jinling Hospital, Nanjing 210002, China
- The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jiaqi Kang
- Medical School of Nanjing University, Department of General Surgery, Jinling Hospital, Nanjing 210002, China
| | - Fan Jiao
- BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210019, China
| | - Xiuwen Wu
- Medical School of Nanjing University, Department of General Surgery, Jinling Hospital, Nanjing 210002, China
| | - Yun Zhao
- BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210019, China
| | - Jianan Ren
- Medical School of Nanjing University, Department of General Surgery, Jinling Hospital, Nanjing 210002, China
| |
Collapse
|
11
|
Li L, Xu T, Qi X. Balanced regulation of ROS production and inflammasome activation in preventing early development of colorectal cancer. Immunol Rev 2025; 329:e13417. [PMID: 39523732 DOI: 10.1111/imr.13417] [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] [Indexed: 11/16/2024]
Abstract
Reactive oxygen species (ROS) production and inflammasome activation are the key components of the innate immune response to microbial infection and sterile insults. ROS are at the intersection of inflammation and immunity during cancer development. Balanced regulation of ROS production and inflammasome activation serves as the central hub of innate immunity, determining whether a cell will survive or undergo cell death. However, the mechanisms underlying this balanced regulation remain unclear. Mitochondria and NADPH oxidases are the two major sources of ROS production. Recently, NCF4, a component of the NADPH oxidase complex that primarily contributes to ROS generation in phagocytes, was reported to balance ROS production and inflammasome activation in macrophages. The phosphorylation and puncta distribution of NCF4 shifts from the membrane-bound NADPH complex to the perinuclear region, promoting ASC speck formation and inflammasome activation, which triggers downstream IL-18-IFN-γ signaling to prevent the progression of colorectal cancer (CRC). Here, we review ROS signaling and inflammasome activation studies in colitis-associated CRC and propose that NCF4 acts as a ROS sensor that balances ROS production and inflammasome activation. In addition, NCF4 is a susceptibility gene for Crohn's disease (CD) and CRC. We discuss the evidence demonstrating NCF4's crucial role in facilitating cell-cell contact between immune cells and intestinal cells, and mediating the paracrine effects of inflammatory cytokines and ROS. This coordination of the signaling network helps create a robust immune microenvironment that effectively prevents epithelial cell mutagenesis and tumorigenesis during the early stage of colitis-associated CRC.
Collapse
Affiliation(s)
- Longjun Li
- Key Laboratory for Experimental Teratology of the Ministry of Education, Advanced Medical Research Institute, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tao Xu
- Key Laboratory for Experimental Teratology of the Ministry of Education, Advanced Medical Research Institute, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiaopeng Qi
- Key Laboratory for Experimental Teratology of the Ministry of Education, Advanced Medical Research Institute, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
12
|
Li Y, Chen J, Liang H, Du Q, Shen J, Wang X. Gasdermin D regulates the activation of EGFR in colorectal cancer. J Transl Med 2024; 22:1170. [PMID: 39741309 DOI: 10.1186/s12967-024-05984-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/13/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Gasdermin D (GSDMD) is a key effector molecule that activates pyroptosis through its N terminal domain (GSDMD-NT). However, the roles of GSDMD in colorectal cancer (CRC) have not been fully explored. The role of the full-length GSDMD (GSDMD-FL) is also not clear. In this study, we observed that GSDMD modulates CRC progression through other mechanisms in addition to activating GSDMD-NT. METHODS Clinical CRC samples and human-derived CRC cell lines were used in this study. GSDMD expression was evaluated by RT-qPCR, Western blot and immunohistochemical (IHC) analysis. GSDMD knockdown and overexpression stable cell lines were established by Lentiviral transduction. CCK-8 assay, flow cytometry analysis for cell cycle, Transwell assay, and cell scratch assay were performed in vitro to explore the impact of GSDMD on CRC progression. Mouse subcutaneous transplantation tumor models were constructed to assess the role of GSDMD in vivo. Intestinal epithelial cell (IEC)-specific knockout of Gsdmd mice (GsdmdΔIEC) was used to evaluate the effect of GSDMD on intestinal adenoma formation in AOM-DSS and Apcmin/+ mouse models. RNA sequencing was performed to explore the regulatory pathways associated with the role of GSDMD in CRC cells. Co-Immunoprecipitation (CO-IP), Western blot and immunofluorescence (IF) were conducted to investigate the interactions between GSDMD and EGFR. Exogenous addition of Gefitinib was used to evaluate the effect of GSDMD on autophosphorylation of EGFR at the Tyr1068 site. RESULTS GSDMD was highly expressed in clinical CRC tissues and human-derived CRC cell lines. GSDMD knockdown inhibited the viability, cell cycle changes, invasion ability and migration ability of CRC cell lines in vitro and vivo, whereas GSDMD overexpression had the opposite effects. Intestinal adenoma development was reduced in GsdmdΔIEC mice in both AOM-DSS and Apcmin/+ mouse models. GSDMD-FL interacted with EGFR and promoted CRC progression by inducing autophosphorylation of EGFR at the Tyr1068 site, subsequently activating ERK1/2. Exogenous Gefitinib abrogated the tumorigenic properties of GSDMD. CONCLUSIONS GSDMD-FL promotes CRC progression by inducing EGFR autophosphorylation at the Tyr1068 site, subsequently activating the downstream ERK1/2. Inhibition of GSDMD is a potential strategy for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Ying Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Jiayao Chen
- Department of Oncology, Zhangjiagang Third People's Hospital, Suzhou, 215611, China
| | - Huijun Liang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Qindan Du
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Jingjie Shen
- The Ninth People's Hospital of Suzhou City, Suzhou, China
| | - Xiaoying Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of medicine, Jiangnan University, Wuxi, China.
| |
Collapse
|
13
|
Li S, Wang Y, Xie Z, Wang Y, Feng Z, Xu J, Yuan B, Zhang Y, Yang G, Wang J, Yuan Y. NLRP3 activation maintains intestinal epithelial barrier and reduces liver injury in alcoholic liver disease mice. Clin Transl Med 2024; 14:e70099. [PMID: 39605303 PMCID: PMC11602754 DOI: 10.1002/ctm2.70099] [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: 09/18/2024] [Revised: 10/27/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Alcoholic liver disease (ALD) patients with bacterial infections usually exhibit high mortality rates. Infections frequently involve bacteria such as Vibrio vulnificus and Enterococcus faecalis. Nevertheless, the mechanisms predisposing ALD patients to bacterial infections and the role of the NLRP3 inflammasome in the intestinal epithelial barrier in ALD remain unclear. METHODS We established ALD mice models of WT, Nlrp3-/- and Gsdmd-/- through chronic alcohol consumption feeding and acute alcohol induction. We compared alterations in gut microbiota, ileitis, and adhesion protein expression, to analyze the role and potential mechanism of NLRP3 in the early onset of ALD. Concurrently, we examined the changes in inflammation and liver damage in the ileum of ALD and healthy mice following foodborne infection with V. vulnificus. RESULTS Compared with the control group, the expression levels of ZO-1, Claudin-1 and E-cadherin were reduced in the ileum of ALD mice, while those of NLRP3, caspase-1(p20), GSDMD-N and IL-1β were elevated. Nlrp3-/- and Gsdmd-/- ALD mice showed an increased gut bacterial load, decreased ileal expression of E-cadherin, more severe ileitis, pronounced liver damage, steatosis and higher plasma levels of FITC-dextran, D-LA and ZO-1 compared with WT mice. Notably, Nlrp3-/- ALD mice exhibited a higher presence of Deferribacterota and Enterobacteriaceae. Furthermore, ALD mice infected with V. vulnificus infection exhibited no further activation of NLRP3 in the ileum, leading to increased intestinal permeability and bloodstream infections. CONCLUSIONS This study indicated that NLRP3 activation in the ileum of ALD mice stabilizes the inflammation-related gut microbiota, preserves the intestinal epithelial barrier, and diminishes inflammation and liver injury. Furthermore, the compromised immune defence in ALD mice may contribute to their heightened susceptibility to bacterial pathogens. KEY POINTS Activation of the NLRP3-GSDMD pathway in the ileum of Alcoholic liver disease (ALD) mice. NLRP3 activation maintains homeostasis of gut microbiota and intestinal epithelial barrier in ALD mice. ALD mice infected with V. vulnificus infection exhibited no further activation of NLRP3 in the ileum, leading to increased intestinal permeability and bloodstream infections.
Collapse
Affiliation(s)
- Shi‐Qing Li
- State Key Laboratory of Pathogen and BiosecurityAcademy of Military Medical Sciences (AMMS)BeijingChina
- State Key Laboratory of Respiratory DiseaseThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Ya‐Ru Wang
- State Key Laboratory of Pathogen and BiosecurityAcademy of Military Medical Sciences (AMMS)BeijingChina
- Hainan Medical Products AdministrationHainan Center for Drug InspectionHaikouChina
| | - Zhong‐Liang Xie
- State Key Laboratory of Pathogen and BiosecurityAcademy of Military Medical Sciences (AMMS)BeijingChina
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijingChina
| | - Yan Wang
- State Key Laboratory of Pathogen and BiosecurityAcademy of Military Medical Sciences (AMMS)BeijingChina
| | - Zi‐Han Feng
- Department of Disease Control and PreventionThe No. 96609 Hospital of Chinese People's Liberation ArmyYinchuanNingxiaChina
| | - Jian‐Hao Xu
- State Key Laboratory of Pathogen and BiosecurityAcademy of Military Medical Sciences (AMMS)BeijingChina
| | - Bing Yuan
- State Key Laboratory of Pathogen and BiosecurityAcademy of Military Medical Sciences (AMMS)BeijingChina
| | - Yi‐Tong Zhang
- State Key Laboratory of Pathogen and BiosecurityAcademy of Military Medical Sciences (AMMS)BeijingChina
| | - Guan Yang
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijingChina
| | - Jing‐Lin Wang
- State Key Laboratory of Pathogen and BiosecurityAcademy of Military Medical Sciences (AMMS)BeijingChina
| | - Yuan Yuan
- State Key Laboratory of Pathogen and BiosecurityAcademy of Military Medical Sciences (AMMS)BeijingChina
| |
Collapse
|
14
|
Vahkal B, Altosaar I, Ariana A, Jabbour J, Pantieras F, Daniel R, Tremblay É, Sad S, Beaulieu JF, Côté M, Ferretti E. Human milk extracellular vesicles modulate inflammation and cell survival in intestinal and immune cells. Pediatr Res 2024:10.1038/s41390-024-03757-5. [PMID: 39609615 DOI: 10.1038/s41390-024-03757-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 11/30/2024]
Abstract
Human milk contains extracellular vesicles (EVs) that carry bioactive molecules such as microRNA, to the newborn intestine. The downstream effects of EV cargo on signaling and immune modulation may shield neonates against inflammatory diseases, including necrotizing enterocolitis. Premature infants are especially at risk, while human milk-feeding may offer protection. The effect of gestational-age specific term and preterm EVs from transitional human milk was characterized on human intestinal epithelial cells (HIECs and Caco-2), primary macrophages, and THP-1 monocytes. We hypothesized that term and preterm EVs differentially influence immune-related cytokines and cell death. We found that preterm EVs were enriched in CD14 surface marker, while both term and preterm EVs increased epidermal growth factor secretion. Following inflammatory stimuli, only term EVs inhibited secretion of IL-6 in HIECs, and reduced expression of pro-inflammatory cytokine IL-1β in macrophages. Term and preterm EVs inhibited secretion of IL-1β and reduced inflammasome related cell death. We proposed that human milk EVs regulate immune-related signaling via their conserved microRNA cargo, which could promote tolerance and a homeostatic immune response. These findings provide basis for further studies into potential therapeutic supplementation with EVs in vulnerable newborn populations by considering functional, gestational age-specific effects. IMPACT: This study reveals distinct functional differences between term and preterm transitional human milk extracellular vesicles (EVs) highlighting the importance of gestational age in their bioactivity. Term EVs uniquely inhibited IL-6 secretion, IL-1β expression, and apoptosis following inflammatory stimuli. Both term and preterm human milk EVs reduced IL-1β secretion and inflammasome-induced cell death. Conserved human milk extracellular vesicle microRNA cargo could be a mediator of the anti-inflammatory effects, particularly targeting cytokine production, the inflammasome, and programmed cell death. These findings underscore the importance of considering gestational age in future research exploring the therapeutic potential of human milk extracellular vesicles to prevent or treat intestinal inflammatory diseases in neonates.
Collapse
Affiliation(s)
- Brett Vahkal
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation (CI3), University of Ottawa, Ottawa, ON, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Illimar Altosaar
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Ardeshir Ariana
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation (CI3), University of Ottawa, Ottawa, ON, Canada
| | - Josie Jabbour
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Falia Pantieras
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Redaet Daniel
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation (CI3), University of Ottawa, Ottawa, ON, Canada
| | - Éric Tremblay
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Subash Sad
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation (CI3), University of Ottawa, Ottawa, ON, Canada
| | - Jean-François Beaulieu
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.
- Centre for Infection, Immunity and Inflammation (CI3), University of Ottawa, Ottawa, ON, Canada.
| | - Emanuela Ferretti
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada.
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada.
| |
Collapse
|
15
|
Zhu JH, Ouyang SX, Zhang GY, Cao Q, Xin R, Yin H, Wu JW, Zhang Y, Zhang Z, Liu Y, Fu JT, Chen YT, Tong J, Zhang JB, Liu J, Shen FM, Li DJ, Wang P. GSDME promotes MASLD by regulating pyroptosis, Drp1 citrullination-dependent mitochondrial dynamic, and energy balance in intestine and liver. Cell Death Differ 2024; 31:1467-1486. [PMID: 39009654 PMCID: PMC11519926 DOI: 10.1038/s41418-024-01343-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
Dysregulated metabolism, cell death, and inflammation contribute to the development of metabolic dysfunction-associated steatohepatitis (MASH). Pyroptosis, a recently identified form of programmed cell death, is closely linked to inflammation. However, the precise role of pyroptosis, particularly gasdermin-E (GSDME), in MASH development remains unknown. In this study, we observed GSDME cleavage and GSDME-associated interleukin-1β (IL-1β)/IL-18 induction in liver tissues of MASH patients and MASH mouse models induced by a choline-deficient high-fat diet (CDHFD) or a high-fat/high-cholesterol diet (HFHC). Compared with wild-type mice, global GSDME knockout mice exhibited reduced liver steatosis, steatohepatitis, fibrosis, endoplasmic reticulum stress, lipotoxicity and mitochondrial dysfunction in CDHFD- or HFHC-induced MASH models. Moreover, GSDME knockout resulted in increased energy expenditure, inhibited intestinal nutrient absorption, and reduced body weight. In the mice with GSDME deficiency, reintroduction of GSDME in myeloid cells-rather than hepatocytes-mimicked the MASH pathologies and metabolic dysfunctions, as well as the changes in the formation of neutrophil extracellular traps and hepatic macrophage/monocyte subclusters. These subclusters included shifts in Tim4+ or CD163+ resident Kupffer cells, Ly6Chi pro-inflammatory monocytes, and Ly6CloCCR2loCX3CR1hi patrolling monocytes. Integrated analyses of RNA sequencing and quantitative proteomics revealed a significant GSDME-dependent reduction in citrullination at the arginine-114 (R114) site of dynamin-related protein 1 (Drp1) during MASH. Mutation of Drp1 at R114 reduced its stability, impaired its ability to redistribute to mitochondria and regulate mitophagy, and ultimately promoted its degradation under MASH stress. GSDME deficiency reversed the de-citrullination of Drp1R114, preserved Drp1 stability, and enhanced mitochondrial function. Our study highlights the role of GSDME in promoting MASH through regulating pyroptosis, Drp1 citrullination-dependent mitochondrial function, and energy balance in the intestine and liver, and suggests that GSDME may be a potential therapeutic target for managing MASH.
Collapse
Affiliation(s)
- Jia-Hui Zhu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shen-Xi Ouyang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guo-Yan Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qi Cao
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, China
- The National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University, Shanghai, China
| | - Rujuan Xin
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hang Yin
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jing-Wen Wu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yan Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhen Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Liu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiang-Tao Fu
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yi-Ting Chen
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Jie Tong
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jia-Bao Zhang
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, China
| | - Jian Liu
- Department of Hepatic Surgery, The Eastern Hepatobiliary Surgery Hospital, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Pei Wang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, China.
- The National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University, Shanghai, China.
| |
Collapse
|
16
|
Zhang J, Tian R, Liu J, Yuan J, Zhang S, Chi Z, Yu W, Yu Q, Wang Z, Chen S, Li M, Yang D, Hu T, Deng Q, Lu X, Yang Y, Zhou R, Zhang X, Liu W, Wang D. A two-front nutrient supply environment fuels small intestinal physiology through differential regulation of nutrient absorption and host defense. Cell 2024; 187:6251-6271.e20. [PMID: 39427662 DOI: 10.1016/j.cell.2024.08.012] [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: 09/08/2023] [Revised: 03/11/2024] [Accepted: 08/07/2024] [Indexed: 10/22/2024]
Abstract
The small intestine contains a two-front nutrient supply environment created by luminal dietary and microbial metabolites (enteral side) and systemic metabolites from the host (serosal side). Yet, it is unknown how each side contributes differentially to the small intestinal physiology. Here, we generated a comprehensive, high-resolution map of the small intestinal two-front nutrient supply environment. Using in vivo tracing of macronutrients and spatial metabolomics, we visualized the spatiotemporal dynamics and cell-type tropism in nutrient absorption and the region-specific metabolic heterogeneity within the villi. Specifically, glutamine from the enteral side fuels goblet cells to support mucus production, and the serosal side loosens the epithelial barrier by calibrating fungal metabolites. Disorganized feeding patterns, akin to the human lifestyle of skipping breakfast, increase the risk of metabolic diseases by inducing epithelial memory of lipid absorption. This study improves our understanding of how the small intestine is spatiotemporally regulated by its unique nutritional environment.
Collapse
Affiliation(s)
- Jian Zhang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311113, China
| | - Ruonan Tian
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jia Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Yuan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Siwen Zhang
- Department of Endocrinology & Metabolism, The First Hospital of Jilin University, Changchun 130061, China
| | - Zhexu Chi
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Weiwei Yu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qianzhou Yu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhen Wang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Sheng Chen
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mobai Li
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Dehang Yang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Tianyi Hu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qiqi Deng
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoyang Lu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yidong Yang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Rongbin Zhou
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Xue Zhang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wanlu Liu
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh EH8 9YL, UK.
| | - Di Wang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311113, China.
| |
Collapse
|
17
|
Lei L, Deng D, Xu W, Yue M, Wu D, Fu K, Shi Z. Increased intestinal permeability and lipopolysaccharide contribute to swainsonine-induced systemic inflammation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116912. [PMID: 39181073 DOI: 10.1016/j.ecoenv.2024.116912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
Long-term consumption of swainsonine could be poisonous to livestock, including facilitating apoptosis by impairing lysosomal function and inhibiting autophagic degradation, leading to liver inflammation and even death in livestock. However, the mechanism by swainsonine induced systemic inflammatory responses remained unclear, especially the effects of swainsonine on intestinal permeability, lipopolysaccharide (LPS) level and oxidative stress response were unknown. In this study, swainsonine increased intestinal permeability as evidenced by the significant down-regulation of colonic goblet cells, Akkermansia muciniphila and intestinal tight junction protein Occludin, Claudin 1 and ZO-1, and the significant up-regulation of mRNA expression level of the intestinal permeability indicator protein tyrosine phosphatase receptor type H (Ptprh) in the ileum of mice. Simultaneously, the elevated LPS biosynthetic genes in intestinal microbiota and increased intestinal permeability facilitated more bacterial endotoxin LPS to enter the blood. High concentration of free-form LPS induced high levels of proinflammatory cytokines and oxidative stress response, thereby causing the systemic inflammation. These findings provided a new perspective on swainsonine-induced systemic inflammation, suggesting that intestinal permeability and free-form LPS level may be the potential trigger factors.
Collapse
Affiliation(s)
- Ling Lei
- Clinical Psychology, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, Nanning, China
| | - Dazhi Deng
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning 530021, China
| | - Wenqian Xu
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Mingyuan Yue
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Dandan Wu
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Keyi Fu
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730000, China.
| | - Zunji Shi
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
18
|
Wu J, Wang H, Gao P, Ouyang S. Pyroptosis: Induction and inhibition strategies for immunotherapy of diseases. Acta Pharm Sin B 2024; 14:4195-4227. [PMID: 39525577 PMCID: PMC11544194 DOI: 10.1016/j.apsb.2024.06.026] [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: 03/13/2024] [Revised: 05/15/2024] [Accepted: 06/20/2024] [Indexed: 11/16/2024] Open
Abstract
Cell death is a central process for organismal health. Pyroptosis, namely pyroptotic cell death, is recognized as a critical type that disrupts membrane and triggers pro-inflammatory cytokine secretion via gasdermins, providing a robust form of cytolysis. Meanwhile, along with the thorough research, a great deal of evidence has demonstrated the dual effects of pyroptosis in host defense and inflammatory diseases. More importantly, the recent identification of abundant gasdermin-like proteins in bacteria and fungi suggests an ancient origin of pyroptosis-based regulated cell death in the life evolution. In this review, we bring a general overview of pyroptosis pathways focusing on gasdermin structural biology, regulatory mechanisms, and recent progress in induction and inhibition strategies for disease treatment. We look forward to providing an insightful perspective for readers to comprehend the frame and challenges of the pyroptosis field, and to accelerating its clinical application.
Collapse
Affiliation(s)
- Junjun Wu
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, the Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Hong Wang
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, the Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Pu Gao
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Songying Ouyang
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, the Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| |
Collapse
|
19
|
Chi Z, Chen S, Yang D, Cui W, Lu Y, Wang Z, Li M, Yu W, Zhang J, Jiang Y, Sun R, Yu Q, Hu T, Lu X, Deng Q, Yang Y, Zhao T, Chang M, Li Y, Zhang X, Shang M, Xiao Q, Ding K, Wang D. Gasdermin D-mediated metabolic crosstalk promotes tissue repair. Nature 2024; 634:1168-1177. [PMID: 39260418 DOI: 10.1038/s41586-024-08022-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 09/05/2024] [Indexed: 09/13/2024]
Abstract
The establishment of an early pro-regenerative niche is crucial for tissue regeneration1,2. Gasdermin D (GSDMD)-dependent pyroptosis accounts for the release of inflammatory cytokines upon various insults3-5. However, little is known about its role in tissue regeneration followed by homeostatic maintenance. Here we show that macrophage GSDMD deficiency delays tissue recovery but has little effect on the local inflammatory milieu or the lytic pyroptosis process. Profiling of the metabolite secretome of hyperactivated macrophages revealed a non-canonical metabolite-secreting function of GSDMD. We further identified 11,12-epoxyeicosatrienoic acid (11,12-EET) as a bioactive, pro-healing oxylipin that is secreted from hyperactive macrophages in a GSDMD-dependent manner. Accumulation of 11,12-EET by direct supplementation or deletion of Ephx2, which encodes a 11,12-EET-hydrolytic enzyme, accelerated muscle regeneration. We further demonstrated that EPHX2 accumulated within aged muscle, and that consecutive 11,12-EET treatment rejuvenated aged muscle. Mechanistically, 11,12-EET amplifies fibroblast growth factor signalling by modulating liquid-liquid phase separation of fibroblast growth factors, thereby boosting the activation and proliferation of muscle stem cells. These data depict a GSDMD-guided metabolite crosstalk between macrophages and muscle stem cells that governs the repair process, which offers insights with therapeutic implications for the regeneration of injured or aged tissues.
Collapse
Affiliation(s)
- Zhexu Chi
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.
- Center for Regeneration and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Yiwu, China.
| | - Sheng Chen
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, China
| | - Dehang Yang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Wenyu Cui
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yang Lu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhen Wang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mobai Li
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weiwei Yu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, China
| | - Jian Zhang
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Yu Jiang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruya Sun
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qianzhou Yu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianyi Hu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyang Lu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiqi Deng
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yidong Yang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianming Zhao
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengfei Chang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuying Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xue Zhang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Min Shang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Xiao
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, China
| | - Di Wang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.
| |
Collapse
|
20
|
Liu Y, Pan R, Ouyang Y, Gu W, Xiao T, Yang H, Tang L, Wang H, Xiang B, Chen P. Pyroptosis in health and disease: mechanisms, regulation and clinical perspective. Signal Transduct Target Ther 2024; 9:245. [PMID: 39300122 DOI: 10.1038/s41392-024-01958-2] [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: 05/08/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024] Open
Abstract
Pyroptosis is a type of programmed cell death characterized by cell swelling and osmotic lysis, resulting in cytomembrane rupture and release of immunostimulatory components, which play a role in several pathological processes. Significant cellular responses to various stimuli involve the formation of inflammasomes, maturation of inflammatory caspases, and caspase-mediated cleavage of gasdermin. The function of pyroptosis in disease is complex but not a simple angelic or demonic role. While inflammatory diseases such as sepsis are associated with uncontrollable pyroptosis, the potent immune response induced by pyroptosis can be exploited as a therapeutic target for anti-tumor therapy. Thus, a comprehensive review of the role of pyroptosis in disease is crucial for further research and clinical translation from bench to bedside. In this review, we summarize the recent advancements in understanding the role of pyroptosis in disease, covering the related development history, molecular mechanisms including canonical, non-canonical, caspase 3/8, and granzyme-mediated pathways, and its regulatory function in health and multiple diseases. Moreover, this review also provides updates on promising therapeutic strategies by applying novel small molecule inhibitors and traditional medicines to regulate pyroptosis. The present dilemmas and future directions in the landscape of pyroptosis are also discussed from a clinical perspective, providing clues for scientists to develop novel drugs targeting pyroptosis.
Collapse
Affiliation(s)
- Yifan Liu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
- Department of Oncology, Xiangya Hospital, Central South University, 87th Xiangya road, Changsha, 410008, Hunan province, China
| | - Renjie Pan
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Yuzhen Ouyang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
- Department of Neurology, Xiangya Hospital, Central South University, 87th Xiangya road, Changsha, 410008, Hunan province, China
| | - Wangning Gu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Tengfei Xiao
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Hongmin Yang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Ling Tang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Hui Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China.
| | - Bo Xiang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China.
| | - Pan Chen
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China.
| |
Collapse
|
21
|
Chen KW, Broz P. Gasdermins as evolutionarily conserved executors of inflammation and cell death. Nat Cell Biol 2024; 26:1394-1406. [PMID: 39187689 DOI: 10.1038/s41556-024-01474-z] [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: 02/01/2024] [Accepted: 07/04/2024] [Indexed: 08/28/2024]
Abstract
The gasdermins are a family of pore-forming proteins that have recently emerged as executors of pyroptosis, a lytic form of cell death that is induced by the innate immune system to eradicate infected or malignant cells. Mammalian gasdermins comprise a cytotoxic N-terminal domain, a flexible linker and a C-terminal repressor domain. Proteolytic cleavage in the linker releases the cytotoxic domain, thereby allowing it to form β-barrel membrane pores. Formation of gasdermin pores in the plasma membrane eventually leads to a loss of the electrochemical gradient, cell death and membrane rupture. Here we review recent work that has expanded our understanding of gasdermin biology and function in mammals by revealing their activation mechanism, their regulation and their roles in autoimmunity, host defence and cancer. We further highlight fungal and bacterial gasdermin pore formation pointing to a conserved mechanism of cell death induction.
Collapse
Affiliation(s)
- Kaiwen W Chen
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| | - Petr Broz
- Department of Immunobiology, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
22
|
Cheng CK, Yi M, Wang L, Huang Y. Role of gasdermin D in inflammatory diseases: from mechanism to therapeutics. Front Immunol 2024; 15:1456244. [PMID: 39253076 PMCID: PMC11381298 DOI: 10.3389/fimmu.2024.1456244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
Inflammatory diseases compromise a clinically common and diverse group of conditions, causing detrimental effects on body functions. Gasdermins (GSDM) are pore-forming proteins, playing pivotal roles in modulating inflammation. Belonging to the GSDM family, gasdermin D (GSDMD) actively mediates the pathogenesis of inflammatory diseases by mechanistically regulating different forms of cell death, particularly pyroptosis, and cytokine release, in an inflammasome-dependent manner. Aberrant activation of GSDMD in different types of cells, such as immune cells, cardiovascular cells, pancreatic cells and hepatocytes, critically contributes to the persistent inflammation in different tissues and organs. The contributory role of GSDMD has been implicated in diabetes mellitus, liver diseases, cardiovascular diseases, neurodegenerative diseases, and inflammatory bowel disease (IBD). Clinically, alterations in GSDMD levels are potentially indicative to the occurrence and severity of diseases. GSDMD inhibition might represent an attractive therapeutic direction to counteract the progression of inflammatory diseases, whereas a number of GSDMD inhibitors have been shown to restrain GSDMD-mediated pyroptosis through different mechanisms. This review discusses the current understanding and future perspectives on the role of GSDMD in the development of inflammatory diseases, as well as the clinical insights of GSDMD alterations, and therapeutic potential of GSDMD inhibitors against inflammatory diseases. Further investigation on the comprehensive role of GSDM shall deepen our understanding towards inflammation, opening up more diagnostic and therapeutic opportunities against inflammatory diseases.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Min Yi
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
23
|
Zhang H, Cui L, Si P, Zhou Y, Zhang Y, Zhang Y, Kong Q. Environmentally relevant concentrations of naphthenic acids initiate intestinal injury and gut microbiota dysbiosis in marine medaka (Oryzias melastigma). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 273:106996. [PMID: 38852546 DOI: 10.1016/j.aquatox.2024.106996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Naphthenic acids (NAs) are important pollutants in marine crude oils and have obvious toxic effects on marine organisms. However, the effects of NAs on the intestine are largely unknown. Thus, we evaluated the effects of NAs exposure in the intestines of marine medaka. Fish were experimentally exposed to NAs (0.5 mg/L, 5 mg/L, and 10 mg/L) for 96 h and monitored for changes in intestinal histology, markers of oxidative stress, and intestinal microbiome responses. Significant mucosal damage, inflammation, and oxidative stress were observed in the intestines of marine medaka after exposure to NAs. In addition, significant changes in the gut microbiota were observed. Specifically, the relative abundance of Proteobacteria decreased, while that of Verrucomicrobiota increased in the high-concentration exposure group. In addition, nutrient synthesis and metabolism in the gut were affected. The results of this study contribute to a better understanding of the ecological risk of different concentrations of NAs to marine organisms. CAPSULE ABSTRACT: Changes in the gut microbial community of marine medaka (Oryzias melastigma) caused by naphthenic acids in the marine environment were investigated through the assessment of gut inflammatory factors and comprehensive analysis using 16S rDNA high-throughput sequencing. The results indicated the induction of intestinal inflammation and changes in the structural composition of the intestinal flora.
Collapse
Affiliation(s)
- Huanxin Zhang
- College of Geography and Environment, Shandong Normal University, 88 Wenhua Donglu, Jinan 250014, Shandong, PR China.
| | - Lihua Cui
- College of Geography and Environment, Shandong Normal University, 88 Wenhua Donglu, Jinan 250014, Shandong, PR China
| | - Panpan Si
- College of Geography and Environment, Shandong Normal University, 88 Wenhua Donglu, Jinan 250014, Shandong, PR China
| | - Yumiao Zhou
- College of the Environment and Ecology, Xiamen University, Xiamen 361005, Fujian, PR China
| | - Yu Zhang
- College of Geography and Environment, Shandong Normal University, 88 Wenhua Donglu, Jinan 250014, Shandong, PR China
| | - Youru Zhang
- College of Geography and Environment, Shandong Normal University, 88 Wenhua Donglu, Jinan 250014, Shandong, PR China
| | - Qiang Kong
- College of Geography and Environment, Shandong Normal University, 88 Wenhua Donglu, Jinan 250014, Shandong, PR China; Dongying Institute, Shandong Normal University, Dongying 257092, Shandong, PR China
| |
Collapse
|
24
|
Chen J, Singh N, Ye X, Theune EV, Wang K. Gut microbiota-mediated activation of GSDMD ignites colorectal tumorigenesis. Cancer Gene Ther 2024; 31:1007-1017. [PMID: 38898209 PMCID: PMC11257976 DOI: 10.1038/s41417-024-00796-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Activation of Gasdermin D (GSDMD) results in its cleavage, oligomerization, and subsequent formation of plasma membrane pores, leading to a form of inflammatory cell death denoted as pyroptosis. The roles of GSDMD in inflammation and immune responses to infection are well documented. However, whether GSDMD also plays a role in sporadic cancer development, especially that in the gut epithelium, remains unknown. Here, we show that GSDMD is activated in colorectal tumors of both human and mouse origins. Ablation of GSDMD in a mouse model of sporadic colorectal cancer resulted in reduced tumor formation in the colon and rectum, suggesting a tumor-promoting role of the protein in the gut. Both antibiotic-mediated depletion of gut microbiota and pharmacological inhibition of NLRP3 inflammasome reduced the activation of GSDMD. Loss of GSDMD resulted in reduced infiltration of immature myeloid cells, and increased numbers of macrophages in colorectal tumors. Activation of GSDMD is also accompanied by the aggregation of the endosomal sorting complex required for transport (ESCRT) membrane repair proteins on the membrane of colorectal tumor cells, suggesting that active membrane repairment may prevent pyroptosis induced by the formation of GSDMD pore in tumor cells. Our results show that gut microbiota/NLRP3-mediated activation of GSDMD promotes the development of colorectal tumors, and supports the use of NLRP3 inhibitors to treat colon cancer.
Collapse
Affiliation(s)
- Ju Chen
- Department of Immunology, School of Medicine, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT, 06030, USA
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, 528000, China
| | - Neha Singh
- Department of Immunology, School of Medicine, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT, 06030, USA
| | - Xiaoyang Ye
- Department of Immunology, School of Medicine, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT, 06030, USA
| | - Eileen Victoria Theune
- Department of Immunology, School of Medicine, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT, 06030, USA
| | - Kepeng Wang
- Department of Immunology, School of Medicine, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT, 06030, USA.
| |
Collapse
|
25
|
Wang Y, Li W, He Z, Yin W, Chen X, Zhang J, Li H. Multichiral Mesoporous Silica Screws with Chiral Differential Mucus Penetration and Mucosal Adhesion for Oral Drug Delivery. ACS NANO 2024; 18:16166-16183. [PMID: 38867485 DOI: 10.1021/acsnano.4c01245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
In the harsh gastrointestinal tract, helical bacteria with hierarchical chiral architectures possess strong abilities. Taking inspirations from nature, we developed a multichiral mesoporous silica nanoscrew (L/D-MCNS) as an efficient oral drug delivery platform by modifying the structural chiral silica nanoscrew (CNS) with L/D-alanine (L/D-Ala) enantiomers via the sequential application of a chiral template and postmodification strategies. We demonstrated that L-MCNS showed differential biological behaviors and superior advantages in oral adsorption compared to those of CNS, D-MCNS, and DL-MCNS. During the delivery, helical L/D-MCNS presenting distinctive topological structures, including small section area, large rough external surface, and a screw-like body, displayed multiple superiorities in mucus diffusion and mucosal adhesion. Meanwhile, the grafted chiral enantiomers enabled positive or negative chiral recognition with the biosystems. Once racemic flurbiprofen (FP) was encapsulated into the nanopores of L/D-MCNS (FP@L/D-MCNS), L/D-MCNS providing highly cross-linked and mesoscopic chiral nanochannels was beneficial for controlling the drug loading/release kinetics with chiral microenvironment sensitivity. Particularly, we noticed enantioselective absorption of FP in vivo, which could be attributed to the differential biological behaviors of L/D-MCNS. By simple design and regulation of the multilevel chirality of nanocarriers, L/D-MCNS can be employed for efficient oral drug delivery from the perspectives of material science, pharmacy, and bionics.
Collapse
Affiliation(s)
- Yumei Wang
- Department of Pharmaceutics, College of Pharmacy, Army Medical University, No. 30 Gaotanyan Main St., 400038 Chongqing, People's Republic of China
| | - Wei Li
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, BinwenRD548, 10053 Hangzhou, Zhejiang Province, People's Republic of China
| | - Zhenwei He
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, No. 4 Chongshan East Road, Huanggu District, Shenyang, Liaoning Province 110032, People's Republic of China
| | - Wencai Yin
- Department of Pharmaceutics, College of Pharmacy, Army Medical University, No. 30 Gaotanyan Main St., 400038 Chongqing, People's Republic of China
| | - Xuchun Chen
- Department of Organ transplantation and Hepatobiliary surgery, First Affiliated Hospital, China Medical University, Nanjingbei Street 155, 110001 Shenyang, Liaoning Province People's Republic of China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Army Medical University, No. 30 Gaotanyan Main St., 400038 Chongqing, People's Republic of China
| | - Heran Li
- School of Pharmacy, China Medical University, Puhe RD77, 110122, Shenyang North New Area, Shenyang, Liaoning Province, People's Republic of China
| |
Collapse
|
26
|
Dong W, Zhang K, Wang X, Li J, Zou H, Yuan Y, Gu J, Zhu J, Liu G, Liu Z, Song R. SIRT1 alleviates Cd nephrotoxicity through NF-κB/p65 deacetylation-mediated pyroptosis in rat renal tubular epithelial cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 929:172392. [PMID: 38608885 DOI: 10.1016/j.scitotenv.2024.172392] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/14/2024]
Abstract
Cadmium (Cd) is a widely distributed environmental pollutant, primarily causing nephrotoxicity through renal proximal tubular cell impairment. Pyroptosis is an inflammation-related nucleotide-binding oligomerization segment-like receptor family 3 (NLRP3)-dependent pathway for programmed cell death. We previously reported that inappropriate inflammation caused by Cd is a major contributor to kidney injury. Therefore, research on Cd-induced inflammatory response and pyroptosis may clarify the mechanisms underlying Cd-induced nephrotoxicity. In this study, we observed that Cd-induced nephrotoxicity is associated with NLRP3 inflammasome activation, leading to an increase in proinflammatory cytokine expression and secretion, as well as pyroptosis-related gene upregulation, both in primary rat proximal tubular (rPT) cells and kidney tissue from Cd-treated rats. In vitro, these effects were significantly abrogated through siRNA-based Nlrp3 silencing; thus, Cd may trigger pyroptosis through an NLRP3 inflammasome-dependent pathway. Moreover, Cd exposure considerably elevated reactive oxygen species (ROS) content. N-acetyl-l-cysteine, an ROS scavenger, mitigated Cd-induced NLRP3 inflammasome activation and subsequent pyroptosis. Mechanistically, Cd hindered the expression and deacetylase activity of SIRT1, eventually leading to a decline in SIRT1-p65 interactions, followed by an elevation in acetylated p65 levels. The administration of resveratrol (a SIRT1 agonist) or overexpression of Sirt1 counteracted Cd-induced RELA/p65/NLRP3 pathway activation considerably, leading to pyroptosis. This is the first study to reveal significant contributions of SIRT1-triggered p65 deacetylation to pyroptosis and its protective effects against Cd-induced chronic kidney injury. Our results may aid in developing potential therapeutic strategies for preventing Cd-induced pyroptosis through SIRT1-mediated p65 deacetylation.
Collapse
Affiliation(s)
- Wenxuan Dong
- Laboratory of Animal Nutrition Metabolic and Poisoning Diseases, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China; College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Kanglei Zhang
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Xueru Wang
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Jiahui Li
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Jiaqiao Zhu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Gang Liu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China; Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Ruilong Song
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| |
Collapse
|
27
|
Xia J, Zhang Y, Zhang S, Lu C, Huan H, Guan X. Oat Dietary Fiber Delays the Progression of Chronic Kidney Disease in Mice by Modulating the Gut Microbiota and Reducing Uremic Toxin Levels. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38836841 DOI: 10.1021/acs.jafc.4c02591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Chronic kidney disease (CKD) has emerged as a significant public health concern. In this article, we investigated the mechanism of oat dietary fiber in regulating CKD. Our findings indicated that the gut microbiota of CKD patients promoted gut microbiota dysbiosis and kidney injury in CKD mice. Intervention with oat-resistant starch prepared by ultrasonic combined enzymatic hydrolysis (ORSU) and oat β-glucan with a molecular weight of 5 × 104 Da (OBGM) elevated the levels of short-chain fatty acids (SCFAs) and regulated gut dysbiosis in the gut-humanized CKD mice. ORSU and OBGM also reduced CKD-related uremic toxins such as creatinine, indoxyl sulfate (IS), and p-cresol sulfate (PCS) levels; reinforced the intestinal barrier function of the gut-humanized CKD mice; and mitigated renal inflammation and fibrosis via the NF-κB/TGF-β pathway. Therefore, ORSU and OBGM might delay the progression of CKD by modulating the gut microbiota to reduce uremic toxins levels. Our results explain the mechanism of oat dietary fiber aimed at mitigating CKD.
Collapse
Affiliation(s)
- Ji'an Xia
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yu Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| | - Suhua Zhang
- Suzhou Kowloon Hospital Shanghai Jiao Tong University School of Medicine, Suzhou, Jiangsu 215028, China
| | - Chunlai Lu
- The 905th Hospital of People's Liberation Army Navy, Shanghai 200050, China
| | - Hongdi Huan
- The 905th Hospital of People's Liberation Army Navy, Shanghai 200050, China
| | - Xiao Guan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| |
Collapse
|
28
|
Shen J, Li F, Han X, Fu D, Xu Y, Zhu C, Liang Z, Tang Z, Zheng R, Hu X, Lin R, Pei Q, Nie J, Luo N, Li X, Chen W, Mao H, Zhou Y, Yu X. Gasdermin D deficiency aborts myeloid calcium influx to drive granulopoiesis in lupus nephritis. Cell Commun Signal 2024; 22:308. [PMID: 38831451 PMCID: PMC11149269 DOI: 10.1186/s12964-024-01681-z] [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: 02/17/2024] [Accepted: 05/27/2024] [Indexed: 06/05/2024] Open
Abstract
Gasdermin D (GSDMD) is emerging as an important player in autoimmune diseases, but its exact role in lupus nephritis (LN) remains controversial. Here, we identified markedly elevated GSDMD in human and mouse LN kidneys, predominantly in CD11b+ myeloid cells. Global or myeloid-conditional deletion of GSDMD was shown to exacerbate systemic autoimmunity and renal injury in lupus mice with both chronic graft-versus-host (cGVH) disease and nephrotoxic serum (NTS) nephritis. Interestingly, RNA sequencing and flow cytometry revealed that myeloid GSDMD deficiency enhanced granulopoiesis at the hematopoietic sites in LN mice, exhibiting remarkable enrichment of neutrophil-related genes, significant increases in total and immature neutrophils as well as granulocyte/macrophage progenitors (GMPs). GSDMD-deficient GMPs and all-trans-retinoic acid (ATRA)-stimulated human promyelocytes NB4 were further demonstrated to possess enhanced clonogenic and differentiation abilities compared with controls. Mechanistically, GSDMD knockdown promoted self-renewal and granulocyte differentiation by restricting calcium influx, contributing to granulopoiesis. Functionally, GSDMD deficiency led to increased pathogenic neutrophil extracellular traps (NETs) in lupus peripheral blood and bone marrow-derived neutrophils. Taken together, our data establish that GSDMD deletion accelerates LN development by promoting granulopoiesis in a calcium influx-regulated manner, unraveling its unrecognized critical role in LN pathogenesis.
Collapse
Affiliation(s)
- Jiani Shen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Feng Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Xu Han
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Dongying Fu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Yiping Xu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Changjian Zhu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Zhou Liang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Ziwen Tang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Ruilin Zheng
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Xinrong Hu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Ruoni Lin
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Qiaoqiao Pei
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Jing Nie
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ning Luo
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Xiaoyan Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.
| | - Yi Zhou
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.
| | - Xueqing Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.
- Department of Nephrology, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China.
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
29
|
Li H, Pu J, Yang D, Liu L, Hu Y, Yang S, Wang B. GSDMD protects intestinal epithelial cells against bacterial infections through its N-terminal activity impacting intestinal immune homeostasis. J Biomed Res 2024; 38:1-12. [PMID: 38807373 PMCID: PMC11629157 DOI: 10.7555/jbr.38.20240041] [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: 02/18/2024] [Revised: 04/07/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
The intestinal mucosal barrier serves as a vital guardian for gut health, maintaining a delicate equilibrium between gut microbiota and host immune homeostasis. Recent studies have found the intricate roles of Gasdermin D (GSDMD), a key executioner of pyroptosis downstream of the inflammasome, within the intestine, including controlling colitis in intestinal macrophage and the regulatory function in goblet cell mucus secretion. Thus, the exact role and nature of GSDMD's regulatory function in maintaining intestinal immune homeostasis and defending against pathogens remain elucidation. Here, we uncover that GSDMD plays a key role in defending against intestinal Citrobacter rodentium infection, with high expression in intestinal epithelial and lamina propria myeloid cells. Our results show that GSDMD specifically acts in intestinal epithelial cells to fight the infection, independently of its effects on antimicrobial peptides or mucin secretion. Instead, the resistance is mediated through GSDMD's N-terminal fragments, highlighting its importance in intestinal immunity. However, the specific underlying mechanism of GSDMD N-terminal activity in protection against intestinal bacterial infections still needs further study to clarify in the future.
Collapse
Affiliation(s)
- Honghui Li
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jie Pu
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Dongxue Yang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Lu Liu
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yingchao Hu
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Shuo Yang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bingwei Wang
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| |
Collapse
|
30
|
Traughber CA, Timinski K, Prince A, Bhandari N, Neupane K, Khan MR, Opoku E, Opoku E, Brubaker G, Shin J, Hong J, Kanuri B, Ertugral EG, Nagareddy PR, Kothapalli CR, Cherepanova O, Smith JD, Gulshan K. Disulfiram Reduces Atherosclerosis and Enhances Efferocytosis, Autophagy, and Atheroprotective Gut Microbiota in Hyperlipidemic Mice. J Am Heart Assoc 2024; 13:e033881. [PMID: 38563369 PMCID: PMC11262521 DOI: 10.1161/jaha.123.033881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/06/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Pyroptosis executor GsdmD (gasdermin D) promotes atherosclerosis in mice and humans. Disulfiram was recently shown to potently inhibit GsdmD, but the in vivo efficacy and mechanism of disulfiram's antiatherosclerotic activity is yet to be explored. METHODS AND RESULTS We used human/mouse macrophages, endothelial cells, and smooth muscle cells and a hyperlipidemic mouse model of atherosclerosis to determine disulfiram antiatherosclerotic efficacy and mechanism. The effects of disulfiram on several atheroprotective pathways such as autophagy, efferocytosis, phagocytosis, and gut microbiota were determined. Atomic force microscopy was used to determine the effects of disulfiram on the biophysical properties of the plasma membrane of macrophages. Disulfiram-fed hyperlipidemic apolipoprotein E-/- mice showed significantly reduced interleukin-1β release upon in vivo Nlrp3 (NLR family pyrin domain containing 3) inflammasome activation. Disulfiram-fed mice showed smaller atherosclerotic lesions (~27% and 29% reduction in males and females, respectively) and necrotic core areas (~50% and 46% reduction in males and females, respectively). Disulfiram induced autophagy in macrophages, smooth muscle cells, endothelial cells, hepatocytes/liver, and atherosclerotic plaques. Disulfiram modulated other atheroprotective pathways (eg, efferocytosis, phagocytosis) and gut microbiota. Disulfiram-treated macrophages showed enhanced phagocytosis/efferocytosis, with the mechanism being a marked increase in cell-surface expression of efferocytic receptor MerTK. Atomic force microscopy analysis revealed altered biophysical properties of disulfiram-treated macrophages, showing increased order-state of plasma membrane and increased adhesion strength. Furthermore, 16sRNA sequencing of disulfiram-fed hyperlipidemic mice showed highly significant enrichment in atheroprotective gut microbiota Akkermansia and a reduction in atherogenic Romboutsia species. CONCLUSIONS Taken together, our data show that disulfiram can simultaneously modulate several atheroprotective pathways in a GsdmD-dependent as well as GsdmD-independent manner.
Collapse
Affiliation(s)
- C. Alicia Traughber
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Kara Timinski
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Ashutosh Prince
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Nilam Bhandari
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Kalash Neupane
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Mariam R. Khan
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Esther Opoku
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Emmanuel Opoku
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Gregory Brubaker
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Junchul Shin
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Junyoung Hong
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Babunageswararao Kanuri
- Department of Internal Medicine, Cardiovascular SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | - Elif G. Ertugral
- Department of Chemical & Biomedical EngineeringCleveland State UniversityClevelandOHUSA
| | - Prabhakara R. Nagareddy
- Department of Internal Medicine, Cardiovascular SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | | | - Olga Cherepanova
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Jonathan D. Smith
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Kailash Gulshan
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| |
Collapse
|
31
|
Zhu C, Xu S, Jiang R, Yu Y, Bian J, Zou Z. The gasdermin family: emerging therapeutic targets in diseases. Signal Transduct Target Ther 2024; 9:87. [PMID: 38584157 PMCID: PMC10999458 DOI: 10.1038/s41392-024-01801-8] [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: 01/15/2024] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
The gasdermin (GSDM) family has garnered significant attention for its pivotal role in immunity and disease as a key player in pyroptosis. This recently characterized class of pore-forming effector proteins is pivotal in orchestrating processes such as membrane permeabilization, pyroptosis, and the follow-up inflammatory response, which are crucial self-defense mechanisms against irritants and infections. GSDMs have been implicated in a range of diseases including, but not limited to, sepsis, viral infections, and cancer, either through involvement in pyroptosis or independently of this process. The regulation of GSDM-mediated pyroptosis is gaining recognition as a promising therapeutic strategy for the treatment of various diseases. Current strategies for inhibiting GSDMD primarily involve binding to GSDMD, blocking GSDMD cleavage or inhibiting GSDMD-N-terminal (NT) oligomerization, albeit with some off-target effects. In this review, we delve into the cutting-edge understanding of the interplay between GSDMs and pyroptosis, elucidate the activation mechanisms of GSDMs, explore their associations with a range of diseases, and discuss recent advancements and potential strategies for developing GSDMD inhibitors.
Collapse
Affiliation(s)
- Chenglong Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Sheng Xu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Ruoyu Jiang
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Yizhi Yu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China.
| | - Jinjun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Zui Zou
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
32
|
Ding N, Fu X, Gui Q, Wu M, Niu Z, Du A, Liu J, Wu H, Wang Y, Yue X, Zhu L. Biomimetic Structure Hydrogel Loaded with Long-Term Storage Platelet-Rich Plasma in Diabetic Wound Repair. Adv Healthc Mater 2024; 13:e2303192. [PMID: 38011625 DOI: 10.1002/adhm.202303192] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/16/2023] [Indexed: 11/29/2023]
Abstract
Exploring the preparation of multifunctional hydrogels from a bionic perspective is an appealing strategy. Here, a multifunctional hydrogel dressing inspired by the characteristics of porous extracellular matrix produced during Acomys wound healing is prepared. These dressings are printed by digital light processing printing of hydrogels composed of gelatin methacrylate, hyaluronic acid methacrylate, and pretreated platelet-rich plasma (PRP) to shape out triply periodic minimal surface structures, which are freeze-dried for long-term storage. These dressings mimic the porous extracellular matrix of Acomys, while the freeze-drying technique effectively extends the storage duration of PRP viability. Through in vivo and in vitro experiments, the biomimetic dressings developed in this study modulate cell behavior and facilitate wound healing. Consequently, this research offers a novel approach for the advancement of regenerative wound dressings.
Collapse
Affiliation(s)
- Neng Ding
- Department of Plastic and Reconstructive Surgery, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 415 Fengyang Road, Shanghai, 200003, China
- Department of Burns and Plastic Surgery, The 74th Group Army Hospital of the PLA Army, 468 Xingang Zhong Road, Guangzhou, 510315, China
| | - Xinxin Fu
- School of Materials Science and Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai, 200082, China
| | - Qixiang Gui
- Department of Plastic and Reconstructive Surgery, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 415 Fengyang Road, Shanghai, 200003, China
- Stem Cell and Regeneration Medicine Institute, Research Center of Translational Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Minjuan Wu
- Department of Histology and Embryology, Basic Medicine College, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Zhongpu Niu
- School of Materials Science and Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai, 200082, China
| | - Antong Du
- Department of Plastic and Reconstructive Surgery, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 415 Fengyang Road, Shanghai, 200003, China
| | - Jinyue Liu
- Department of Plastic and Reconstructive Surgery, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 415 Fengyang Road, Shanghai, 200003, China
| | - Haimei Wu
- School of Materials Science and Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai, 200082, China
| | - Yue Wang
- Stem Cell and Regeneration Medicine Institute, Research Center of Translational Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China
- Department of stem cell engneering, Shanghai Institute of Stem Cell Research and Clinical Translation, 551 Pudong Nan Road, Shanghai, 200120, China
- Department of stem cell engneering, Shanghai Key Laboratory of Cell Engineering, 800 Xiangyin Road, Shanghai, 200433, China
| | - Xuezheng Yue
- School of Materials Science and Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai, 200082, China
| | - Lie Zhu
- Department of Plastic and Reconstructive Surgery, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 415 Fengyang Road, Shanghai, 200003, China
| |
Collapse
|
33
|
Wang S, Li X, Zhang B, Li Y, Chen K, Qi H, Gao M, Rong J, Liu L, Wan Y, Dong X, Yan M, Ma L, Li P, Zhao T. Tangshen formula targets the gut microbiota to treat non-alcoholic fatty liver disease in HFD mice: A 16S rRNA and non-targeted metabolomics analyses. Biomed Pharmacother 2024; 173:116405. [PMID: 38484559 DOI: 10.1016/j.biopha.2024.116405] [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: 12/26/2023] [Revised: 02/24/2024] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Tangshen formula (TSF) has an ameliorative effect on hepatic lipid metabolism in non-alcoholic fatty liver disease (NAFLD), but the role played by the gut microbiota in this process is unknown. METHOD We conducted three batches of experiments to explore the role played by the gut microbiota: TSF administration, antibiotic treatment, and fecal microbial transplantation. NAFLD mice were induced with a high-fat diet to investigate the ameliorative effects of TSF on NAFLD features and intestinal barrier function. 16S rRNA sequencing and serum untargeted metabolomics were performed to further investigate the modulatory effects of TSF on the gut microbiota and metabolic dysregulation in the body. RESULTS TSF ameliorated insulin resistance, hypercholesterolemia, lipid metabolism disorders, inflammation, and impairment of intestinal barrier function. 16S rRNA sequencing analysis revealed that TSF regulated the composition of the gut microbiota and increased the abundance of beneficial bacteria. Antibiotic treatment and fecal microbiota transplantation confirmed the importance of the gut microbiota in the treatment of NAFLD with TSF. Subsequently, untargeted metabolomics identified 172 differential metabolites due to the treatment of TSF. Functional predictions suggest that metabolisms of choline, glycerophospholipid, linoleic acid, alpha-linolenic acid, and arachidonic acid are the key metabolic pathways by which TSF ameliorates NAFLD and this may be influenced by the gut microbiota. CONCLUSION TSF treats the NAFLD phenotype by remodeling the gut microbiota and improving metabolic profile, suggesting that TSF is a functional gut microbial and metabolic modulator for the treatment of NAFLD.
Collapse
Affiliation(s)
- Shaopeng Wang
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, State Key Laboratory of Rsepiratory Health and Multimorbidity, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China; College of Pharmacy, Shandong Second Medical University, Weifang, PR China
| | - Xin Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, State Key Laboratory of Rsepiratory Health and Multimorbidity, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China
| | - Bo Zhang
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, State Key Laboratory of Rsepiratory Health and Multimorbidity, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China
| | - Yuxi Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, State Key Laboratory of Rsepiratory Health and Multimorbidity, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China
| | - Kexu Chen
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, State Key Laboratory of Rsepiratory Health and Multimorbidity, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China; College of Pharmacy, Shandong Second Medical University, Weifang, PR China
| | - Huimin Qi
- College of Pharmacy, Shandong Second Medical University, Weifang, PR China
| | - Mengqi Gao
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, State Key Laboratory of Rsepiratory Health and Multimorbidity, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China
| | - Jin Rong
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, State Key Laboratory of Rsepiratory Health and Multimorbidity, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China
| | - Lin Liu
- Zoucheng Market Supervision Administration, Jining, PR China
| | - Yuzhou Wan
- Research and Development Department, Nanjing Denovo Pharma Co., Ltd, Nanjing, PR China
| | - Xi Dong
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, State Key Laboratory of Rsepiratory Health and Multimorbidity, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China
| | - Meihua Yan
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, State Key Laboratory of Rsepiratory Health and Multimorbidity, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China
| | - Liang Ma
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, State Key Laboratory of Rsepiratory Health and Multimorbidity, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, State Key Laboratory of Rsepiratory Health and Multimorbidity, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China.
| | - Tingting Zhao
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, State Key Laboratory of Rsepiratory Health and Multimorbidity, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China.
| |
Collapse
|
34
|
Yin JT, Zhang MR, Zhang S, Yang SH, Li JP, Liu Y, Duan JA, Guo JM. Astragalus membranaceus Polysaccharide Regulates Small Intestinal Microbes and Activates IL-22 Signal Pathway to Promote Intestinal Stem Cell Regeneration in Aging Mice. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:513-539. [PMID: 38533568 DOI: 10.1142/s0192415x24500228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Aging can cause degenerative changes in multiple tissues and organs. Gastrointestinal diseases and dysfunctions are common in the elderly population. In this study, we investigated the effects of Astragalus membranaceus polysaccharide (APS) and Astragalus membranaceus ethanol extract (AEE) on age-related intestinal dysfunction and gut microbiota dysbiosis in naturally aging mice. The energy expenditure and physical activity of 23-month-old C57BL6/J mice were recorded using a metabolic cage system. Pathological changes in the intestine were evaluated using Alcian blue staining. The protein levels of leucine-rich repeats containing G protein-coupled receptor 5 (Lgr5) and Stat3 in the small intestine were determined using immunohistochemistry. The intestinal cell migration distance was assessed using bromodeoxyuridine (BrdU) immunofluorescence staining. The gene transcription levels of intestinal stem cell (ISC) markers and ISC-related signaling pathways were detected using quantitative real-time PCR (qRT-PCR). Microbiota analysis based on 16S rDNA was performed to evaluate the composition of the gut microbiota. APS and AEE improved a series of aging phenotypes in female but not in male aging mice. APS and AEE ameliorate intestinal dysfunction and histopathological changes in aging mice. APS had a more significant anti-aging effect than AEE, particularly on intestinal dysfunction. APS promotes ISC regeneration by activating the IL-22 signaling pathway. Cohousing (CH) experiments further confirmed that APS induced the IL-22 signaling pathway by increasing the abundance of Lactobacillus, thereby promoting the regeneration of ISCs. Our results show that APS may serve as a promising agent for improving age-related intestinal dysfunction.
Collapse
Affiliation(s)
- Jia-Ting Yin
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM, Formulae Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Ming-Ruo Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM, Formulae Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Shu Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM, Formulae Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Shu-Hui Yang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM, Formulae Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Jian-Ping Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM, Formulae Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Yun Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM, Formulae Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM, Formulae Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Jian-Ming Guo
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM, Formulae Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| |
Collapse
|
35
|
Shkarina K, Broz P. Selective induction of programmed cell death using synthetic biology tools. Semin Cell Dev Biol 2024; 156:74-92. [PMID: 37598045 DOI: 10.1016/j.semcdb.2023.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 08/21/2023]
Abstract
Regulated cell death (RCD) controls the removal of dispensable, infected or malignant cells, and is thus essential for development, homeostasis and immunity of multicellular organisms. Over the last years different forms of RCD have been described (among them apoptosis, necroptosis, pyroptosis and ferroptosis), and the cellular signaling pathways that control their induction and execution have been characterized at the molecular level. It has also become apparent that different forms of RCD differ in their capacity to elicit inflammation or an immune response, and that RCD pathways show a remarkable plasticity. Biochemical and genetic studies revealed that inhibition of a given pathway often results in the activation of back-up cell death mechanisms, highlighting close interconnectivity based on shared signaling components and the assembly of multivalent signaling platforms that can initiate different forms of RCD. Due to this interconnectivity and the pleiotropic effects of 'classical' cell death inducers, it is challenging to study RCD pathways in isolation. This has led to the development of tools based on synthetic biology that allow the targeted induction of RCD using chemogenetic or optogenetic methods. Here we discuss recent advances in the development of such toolset, highlighting their advantages and limitations, and their application for the study of RCD in cells and animals.
Collapse
Affiliation(s)
- Kateryna Shkarina
- Institute of Innate Immunity, University Hospital Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| | - Petr Broz
- Department of Immunobiology, University of Lausanne, Switzerland.
| |
Collapse
|
36
|
Fu SL, Qian YY, Dai AN, Li HY, Jin XH, He WT, Kang S, Ding PH. Casp11 Deficiency Alters Subgingival Microbiota and Attenuates Periodontitis. J Dent Res 2024; 103:298-307. [PMID: 38197150 DOI: 10.1177/00220345231221712] [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: 01/11/2024] Open
Abstract
Periodontitis (PD) is the primary cause of tooth loss in adults. Porphyromonas gingivalis (P.g), a keystone pathogen, has been identified as a crucial contributor to this process. Pyroptosis activation in PD is acknowledged, with accumulating evidence underscoring the crucial role of Caspase-11 (described as Caspase-4/5 in humans)-mediated noncanonical pyroptosis. However, the mechanism behind its impact on PD remains unclear. In this study, we delved into the interplay between the Caspase-11-mediated noncanonical pyroptosis, subgingival microbiota alteration, and macrophage polarization. Clinical samples from PD patients revealed heightened expression of Caspase-4, gasdermin-D, and their active fragments, pointing to the activation of the noncanonical pyroptosis. Single-cell sequencing analysis linked Caspase-4 with gingival macrophages, emphasizing their involvement in PD. In vitro cell experiments confirmed that P.g-induced pyroptosis was activated in macrophages, with Casp11 deficiency attenuating these effects. In an experimental PD mouse model, Casp11 deficiency led to an alteration in subgingival microbiota composition and reduced alveolar bone resorption. Casp11-/- mice cohousing with wild-type mice confirmed the alteration of the subgingival microbiota and aggravated the alveolar bone resorption. Notably, Casp11 deficiency led to decreased M1-polarized macrophages, corresponding with reduced alveolar bone resorption, uncovering a connection between subgingival microbiota alteration, macrophage M1 polarization, and alveolar bone resorption. Taken together, we showed that Caspase-11 fulfilled a crucial role in the noncanonical pyroptosis in PD, potentially influencing the subgingival microbiota and linking to M1 polarization, which was associated with alveolar bone resorption. These findings underscored the pivotal role of the Caspase-11-mediated noncanonical pyroptosis in PD pathogenesis and may provide critical insights into potential therapeutic avenues for mitigating PD.
Collapse
Affiliation(s)
- S L Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Y Y Qian
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - A N Dai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - H Y Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - X H Jin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - W T He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - S Kang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - P H Ding
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| |
Collapse
|
37
|
Chao L, Zhang W, Feng Y, Gao P, Ma J. Pyroptosis: a new insight into intestinal inflammation and cancer. Front Immunol 2024; 15:1364911. [PMID: 38455052 PMCID: PMC10917886 DOI: 10.3389/fimmu.2024.1364911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/09/2024] [Indexed: 03/09/2024] Open
Abstract
Pyroptosis is an innate immune response triggered by the activation of inflammasomes by various influencing factors, characterized by cell destruction. It impacts the immune system and cancer immunotherapy. In recent years, the roles of pyroptosis and inflammasomes in intestinal inflammation and cancer have been continuously confirmed. This article reviews the latest progress in pyroptosis mechanisms, new discoveries of inflammasomes, mutual regulation between inflammasomes, and their applications in intestinal diseases. Additionally, potential synergistic treatment mechanisms of intestinal diseases with pyroptosis are summarized, and challenges and future directions are discussed, providing new ideas for pyroptosis therapy.
Collapse
Affiliation(s)
| | | | | | | | - Jinyou Ma
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| |
Collapse
|
38
|
Shen S, Guo H, Li Y, Zhang L, Tang Y, Li H, Li X, Wang PH, Yu XF, Wei W. SARS-CoV-2 and oncolytic EV-D68-encoded proteases differentially regulate pyroptosis. J Virol 2024; 98:e0190923. [PMID: 38289118 PMCID: PMC10878271 DOI: 10.1128/jvi.01909-23] [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: 12/05/2023] [Accepted: 01/03/2024] [Indexed: 02/21/2024] Open
Abstract
Pyroptosis, a pro-inflammatory programmed cell death, has been implicated in the pathogenesis of coronavirus disease 2019 and other viral diseases. Gasdermin family proteins (GSDMs), including GSDMD and GSDME, are key regulators of pyroptotic cell death. However, the mechanisms by which virus infection modulates pyroptosis remain unclear. Here, we employed a mCherry-GSDMD fluorescent reporter assay to screen for viral proteins that impede the localization and function of GSDMD in living cells. Our data indicated that the main protease NSP5 of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) blocked GSDMD-mediated pyroptosis via cleaving residues Q29 and Q193 of GSDMD. While another SARS-CoV-2 protease, NSP3, cleaved GSDME at residue G370 but activated GSDME-mediated pyroptosis. Interestingly, respiratory enterovirus EV-D68-encoded proteases 3C and 2A also exhibit similar differential regulation on the functions of GSDMs by inactivating GSDMD but initiating GSDME-mediated pyroptosis. EV-D68 infection exerted oncolytic effects on human cancer cells by inducing pyroptotic cell death. Our findings provide insights into how respiratory viruses manipulate host cell pyroptosis and suggest potential targets for antiviral therapy as well as cancer treatment.IMPORTANCEPyroptosis plays a crucial role in the pathogenesis of coronavirus disease 2019, and comprehending its function may facilitate the development of novel therapeutic strategies. This study aims to explore how viral-encoded proteases modulate pyroptosis. We investigated the impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and respiratory enterovirus D68 (EV-D68) proteases on host cell pyroptosis. We found that SARS-CoV-2-encoded proteases NSP5 and NSP3 inactivate gasdermin D (GSDMD) but initiate gasdermin E (GSDME)-mediated pyroptosis, respectively. We also discovered that another respiratory virus EV-D68 encodes two distinct proteases 2A and 3C that selectively trigger GSDME-mediated pyroptosis while suppressing the function of GSDMD. Based on these findings, we further noted that EV-D68 infection triggers pyroptosis and produces oncolytic effects in human carcinoma cells. Our study provides new insights into the molecular mechanisms underlying virus-modulated pyroptosis and identifies potential targets for the development of antiviral and cancer therapeutics.
Collapse
Affiliation(s)
- Siyu Shen
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Haoran Guo
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Yan Li
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Lili Zhang
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Yubin Tang
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Huili Li
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Xiaohan Li
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Pei-Hui Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao-Fang Yu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Wei
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Translational Medicine, First Hospital, Jilin University, Changchun, Jilin, China
| |
Collapse
|
39
|
Lin Z, Chen Q, Ruan HB. To die or not to die: Gasdermins in intestinal health and disease. Semin Immunol 2024; 71:101865. [PMID: 38232665 PMCID: PMC10872225 DOI: 10.1016/j.smim.2024.101865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
Intestinal homeostasis is achieved by the balance among intestinal epithelium, immune cells, and gut microbiota. Gasdermins (GSDMs), a family of membrane pore forming proteins, can trigger rapid inflammatory cell death in the gut, mainly pyroptosis and NETosis. Importantly, there is increasing literature on the non-cell lytic roles of GSDMs in intestinal homeostasis and disease. While GSDMA is low and PJVK is not expressed in the gut, high GSDMB and GSDMC expression is found almost restrictively in intestinal epithelial cells. Conversely, GSDMD and GSDME show more ubiquitous expression among various cell types in the gut. The N-terminal region of GSDMs can be liberated for pore formation by an array of proteases in response to pathogen- and danger-associated signals, but it is not fully understood what cell type-specific mechanisms activate intestinal GSDMs. The host relies on GSDMs for pathogen defense, tissue tolerance, and cancerous cell death; however, pro-inflammatory milieu caused by pyroptosis and excessive cytokine release may favor the development and progression of inflammatory bowel disease and cancer. Therefore, a thorough understanding of spatiotemporal mechanisms that control gasdermin expression, activation, and function is essential for the development of future therapeutics for intestinal disorders.
Collapse
Affiliation(s)
- Zhaoyu Lin
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China.
| | - Qianyue Chen
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
40
|
Lin X, Zhao Z, Sun SP, Liu W. Scinderin promotes glioma cell migration and invasion via remodeling actin cytoskeleton. World J Clin Oncol 2024; 15:32-44. [PMID: 38292665 PMCID: PMC10823943 DOI: 10.5306/wjco.v15.i1.32] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/20/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Glioma is one of the most common intracranial tumors, characterized by invasive growth and poor prognosis. Actin cytoskeletal rearrangement is an essential event of tumor cell migration. The actin dynamics-related protein scinderin (SCIN) has been reported to be closely related to tumor cell migration and invasion in several cancers. AIM To investigate the role and mechanism of SCIN in glioma. METHODS The expression and clinical significance of SCIN in glioma were analyzed based on public databases. SCIN expression was examined using real-time quantitative polymerase chain reaction and Western blotting. Gene silencing was performed using short hairpin RNA transfection. Cell viability, migration, and invasion were assessed using cell counting kit 8 assay, wound healing, and Matrigel invasion assays, respectively. F-actin cytoskeleton organization was assessed using F-actin staining. RESULTS SCIN expression was significantly elevated in glioma, and high levels of SCIN were associated with advanced tumor grade and wild-type isocitrate dehydrogenase. Furthermore, SCIN-deficient cells exhibited decreased proliferation, migration, and invasion in U87 and U251 cells. Moreover, knockdown of SCIN inhibited the RhoA/focal adhesion kinase (FAK) signaling to promote F-actin depolymerization in U87 and U251 cells. CONCLUSION SCIN modulates the actin cytoskeleton via activating RhoA/FAK signaling, thereby promoting the migration and invasion of glioma cells. This study identified the cancer-promoting effect of SCIN and provided a potential therapeutic target for the treatment of glioma.
Collapse
Affiliation(s)
- Xin Lin
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300000, China
| | - Zhao Zhao
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300000, China
| | - Shu-Peng Sun
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300000, China
| | - Wei Liu
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300000, China
| |
Collapse
|
41
|
Zhou L, Li Y, You J, Wu C, Zuo L, Chen Y, Kang L, Zhou Z, Huang R, Wu S. Salmonella spvC gene suppresses macrophage/neutrophil antibacterial defense mediated by gasdermin D. Inflamm Res 2024; 73:19-33. [PMID: 38135851 DOI: 10.1007/s00011-023-01818-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/15/2023] [Accepted: 11/06/2023] [Indexed: 12/24/2023] Open
Abstract
OBJECTIVE Salmonella enterica serovar Typhimurium (S. Typhimurium) is a representative model organism for investigating host-pathogen interactions. It was reported that S. Typhimurium spvC gene alleviated intestinal inflammation to aggravate systemic infection, while the precise mechanisms remain unclear. In this study, the influence of spvC on the antibacterial defense of macrophage/neutrophil mediated by gasdermin D (GSDMD) was investigated. METHODS Mouse macrophage-like cell lines J774A.1 and RAW264.7, neutrophil-like cells derived from HL-60 cells (human promyletic leukemia cell lines) were infected with S. Typhimurium wild type, spvC deletion and complemented strains. Cell death was evaluated by LDH release and Annexin V-FITC/PI staining. Macrophage pyroptosis and neutrophil NETosis were detected by western blotting, live cell imaging and ELISA. Flow cytometry was used to assess the impact of spvC on macrophage-neutrophil cooperation in macrophage (dTHP-1)-neutrophil (dHL-60) co-culture model pretreated with GSDMD inhibitor disulfiram. Wild-type and Gsdmd-/- C57BL/6J mice were utilized for in vivo assay. The degree of phagocytes infiltration and inflammation were analyzed by immunofluorescence and transmission electron microscopy. RESULTS Here we find that spvC inhibits pyroptosis in macrophages via Caspase-1/Caspase-11 dependent canonical and non-canonical pathways, and restrains neutrophil extracellular traps extrusion in GSDMD-dependent manner. Moreover, spvC could ameliorate macrophages/neutrophils infiltration and cooperation in the inflammatory response mediated by GSDMD to combat Salmonella infection. CONCLUSIONS Our findings highlight the antibacterial activity of GSDMD in phagocytes and reveal a novel pathogenic mechanism employed by spvC to counteract this host defense, which may shed new light on designing effective therapeutics to control S. Typhimurium infection.
Collapse
Affiliation(s)
- Liting Zhou
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
- Center of Clinical Laboratory, Dushu Lake Hospital, Affiliated to Soochow University, Suzhou, China
| | - Yuanyuan Li
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Medical Microbiology, Experimental Center, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jiayi You
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chaoyi Wu
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Lingli Zuo
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
- Medical Research Center, The People's Hospital of Suzhou New District, Suzhou, China
| | - Yilin Chen
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Li Kang
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Zhengyu Zhou
- Laboratory Animal Center, Suzhou Medical College of Soochow University, Suzhou, China
| | - Rui Huang
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Shuyan Wu
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
42
|
Feng Y, Wang S, Liu X, Han Y, Xu H, Duan X, Xie W, Tian Z, Yuan Z, Wan Z, Xu L, Qin S, He K, Huang J. Geometric constraint-triggered collagen expression mediates bacterial-host adhesion. Nat Commun 2023; 14:8165. [PMID: 38071397 PMCID: PMC10710423 DOI: 10.1038/s41467-023-43827-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Cells living in geometrically confined microenvironments are ubiquitous in various physiological processes, e.g., wound closure. However, it remains unclear whether and how spatially geometric constraints on host cells regulate bacteria-host interactions. Here, we reveal that interactions between bacteria and spatially constrained cell monolayers exhibit strong spatial heterogeneity, and that bacteria tend to adhere to these cells near the outer edges of confined monolayers. The bacterial adhesion force near the edges of the micropatterned monolayers is up to 75 nN, which is ~3 times higher than that at the centers, depending on the underlying substrate rigidities. Single-cell RNA sequencing experiments indicate that spatially heterogeneous expression of collagen IV with significant edge effects is responsible for the location-dependent bacterial adhesion. Finally, we show that collagen IV inhibitors can potentially be utilized as adjuvants to reduce bacterial adhesion and thus markedly enhance the efficacy of antibiotics, as demonstrated in animal experiments.
Collapse
Affiliation(s)
- Yuting Feng
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Shuyi Wang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Xiaoye Liu
- Beijing Traditional Chinese Veterinary Engineering Center and Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, 102206, Beijing, China
| | - Yiming Han
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Hongwei Xu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Xiaocen Duan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Wenyue Xie
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Zhuoling Tian
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Zuoying Yuan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Zhuo Wan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Liang Xu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Siying Qin
- School of Life Sciences, Peking University, 100871, Beijing, China
| | - Kangmin He
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China.
| |
Collapse
|
43
|
Zhu S, Xu K, Li S, Yu X, Liu Y, Zhang Q, Zeng L, Xu K, Fu C. Assessment of intestinal status in MPL W515L mutant myeloproliferative neoplasms mice model. Int Immunopharmacol 2023; 125:111091. [PMID: 37883814 DOI: 10.1016/j.intimp.2023.111091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/27/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023]
Abstract
The MPLW515L mutation is a prevalent genetic mutation in patients with myeloproliferative neoplasms (MPN), and utilizing this mutation in mice model can provide important insights into the disease. However, the relationship between intestinal homeostasis and MPN mice model remains elusive. In this study, we utilized a retroviral vector to transfect hematopoietic stem cells with the MPLW515L mutation, creating mutated MPN mice model to investigate their intestinal status. Our results revealed that the MPLW515L in MPN mice model aggravated inflammation in the intestines, decreased the levels of tight junction proteins and receptors for bacteria metabolites. Additionally, there was increased activation of the caspase1/IL-1β signaling pathway and a significant reduction in phos-p38 levels in the intestinal tissue in MPN mice. The MPLW515L mutation also led to up-expression of anti-microbial genes in the intestinal tract. Though the mutation had no impact on the alpha diversity and dominant bacterial taxa, it did influence the rare bacterial taxa/sub-communities and consequently impacted intestinal homeostasis. Our findings demonstrate the significance of MPLW515L mice model for studying MPN disease and highlight the mutation's influence on intestinal homeostasis, including inflammation, activation of the IL-1β signaling pathway, and the composition of gut microbial communities.
Collapse
Affiliation(s)
- Shengyun Zhu
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Hematology, Affliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Key Laboratory of Bone Marrow Stem Cells, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kairen Xu
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shuyao Li
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiangru Yu
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yahui Liu
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qigang Zhang
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lingyu Zeng
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Hematology, Affliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Key Laboratory of Bone Marrow Stem Cells, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kailin Xu
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Hematology, Affliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Key Laboratory of Bone Marrow Stem Cells, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Chunling Fu
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Hematology, Affliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Key Laboratory of Bone Marrow Stem Cells, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
44
|
Liu Z, Chen Y, Mei Y, Yan M, Liang H. Gasdermin D-Mediated Pyroptosis in Diabetic Cardiomyopathy: Molecular Mechanisms and Pharmacological Implications. Molecules 2023; 28:7813. [PMID: 38067543 PMCID: PMC10708146 DOI: 10.3390/molecules28237813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/21/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is a pathophysiological condition triggered by diabetes mellitus (DM), which can lead to heart failure (HF). One of the most important cellular processes associated with DCM is the death of cardiomyocytes. Gasdermin D (GSDMD) plays a key role in mediating pyroptosis, a type of programmed cell death closely associated with inflammasome activation. Recent studies have revealed that pyroptosis is induced during hyperglycemia, which is crucial to the development of DCM. Although the effects of pyroptosis on DCM have been discussed, the relationship between DCM and GSDMD is not fully clarified. Recent studies gave us the impetus for clarifying the meaning of GSDMD in DCM. The purpose of this review is to summarize new and emerging insights, mainly discussing the structures of GSDMD and the mechanism of pore formation, activation pathways, molecular mechanisms of GSDMD-mediated pyroptosis, and the therapeutic potential of GSDMD in DCM. The implications of this review will pave the way for a new therapeutic target in DCM.
Collapse
Affiliation(s)
- Zhou Liu
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Z.L.); (Y.C.); (Y.M.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou 510006, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Yifan Chen
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Z.L.); (Y.C.); (Y.M.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou 510006, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Yu Mei
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Z.L.); (Y.C.); (Y.M.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou 510006, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Meiling Yan
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Z.L.); (Y.C.); (Y.M.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou 510006, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| | - Haihai Liang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Z.L.); (Y.C.); (Y.M.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangzhou 510006, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China
| |
Collapse
|
45
|
Fattinger SA, Maurer L, Geiser P, Bernard EM, Enz U, Ganguillet S, Gül E, Kroon S, Demarco B, Mack V, Furter M, Barthel M, Pelczar P, Shao F, Broz P, Sellin ME, Hardt WD. Gasdermin D is the only Gasdermin that provides protection against acute Salmonella gut infection in mice. Proc Natl Acad Sci U S A 2023; 120:e2315503120. [PMID: 37988464 PMCID: PMC10691232 DOI: 10.1073/pnas.2315503120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/05/2023] [Indexed: 11/23/2023] Open
Abstract
Gasdermins (GSDMs) share a common functional domain structure and are best known for their capacity to form membrane pores. These pores are hallmarks of a specific form of cell death called pyroptosis and mediate the secretion of pro-inflammatory cytokines such as interleukin 1β (IL1β) and interleukin 18 (IL18). Thereby, Gasdermins have been implicated in various immune responses against cancer and infectious diseases such as acute Salmonella Typhimurium (S.Tm) gut infection. However, to date, we lack a comprehensive functional assessment of the different Gasdermins (GSDMA-E) during S.Tm infection in vivo. Here, we used epithelium-specific ablation, bone marrow chimeras, and mouse lines lacking individual Gasdermins, combinations of Gasdermins or even all Gasdermins (GSDMA1-3C1-4DE) at once and performed littermate-controlled oral S.Tm infections in streptomycin-pretreated mice to investigate the impact of all murine Gasdermins. While GSDMA, C, and E appear dispensable, we show that GSDMD i) restricts S.Tm loads in the gut tissue and systemic organs, ii) controls gut inflammation kinetics, and iii) prevents epithelium disruption by 72 h of the infection. Full protection requires GSDMD expression by both bone-marrow-derived lamina propria cells and intestinal epithelial cells (IECs). In vivo experiments as well as 3D-, 2D-, and chimeric enteroid infections further show that infected IEC extrusion proceeds also without GSDMD, but that GSDMD controls the permeabilization and morphology of the extruding IECs, affects extrusion kinetics, and promotes overall mucosal barrier capacity. As such, this work identifies a unique multipronged role of GSDMD among the Gasdermins for mucosal tissue defense against a common enteric pathogen.
Collapse
Affiliation(s)
- Stefan A. Fattinger
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich8093, Switzerland
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala75123, Sweden
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
| | - Luca Maurer
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich8093, Switzerland
| | - Petra Geiser
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala75123, Sweden
| | - Elliott M. Bernard
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Ursina Enz
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich8093, Switzerland
| | - Suwannee Ganguillet
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich8093, Switzerland
| | - Ersin Gül
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich8093, Switzerland
| | - Sanne Kroon
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich8093, Switzerland
| | - Benjamin Demarco
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Vanessa Mack
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Markus Furter
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich8093, Switzerland
| | - Manja Barthel
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich8093, Switzerland
| | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, Basel4002, Switzerland
| | - Feng Shao
- National Institute of Biological Sciences, Beijing102206, China
| | - Petr Broz
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Mikael E. Sellin
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala75123, Sweden
| | - Wolf-Dietrich Hardt
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich8093, Switzerland
| |
Collapse
|
46
|
Wang W, He Z. Gasdermins in sepsis. Front Immunol 2023; 14:1203687. [PMID: 38022612 PMCID: PMC10655013 DOI: 10.3389/fimmu.2023.1203687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis is a hyper-heterogeneous syndrome in which the systemic inflammatory response persists throughout the course of the disease and the inflammatory and immune responses are dynamically altered at different pathogenic stages. Gasdermins (GSDMs) proteins are pore-forming executors in the membrane, subsequently mediating the release of pro-inflammatory mediators and inflammatory cell death. With the increasing research on GSDMs proteins and sepsis, it is believed that GSDMs protein are one of the most promising therapeutic targets in sepsis in the future. A more comprehensive and in-depth understanding of the functions of GSDMs proteins in sepsis is important to alleviate the multi-organ dysfunction and reduce sepsis-induced mortality. In this review, we focus on the function of GSDMs proteins, the molecular mechanism of GSDMs involved in sepsis, and the regulatory mechanism of GSDMs-mediated signaling pathways, aiming to provide novel ideas and therapeutic strategies for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Wenhua Wang
- Department of Intensive Care Unit, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhihui He
- Department of Intensive Care Unit, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, Hunan, China
| |
Collapse
|
47
|
Pang J, Vince JE. The role of caspase-8 in inflammatory signalling and pyroptotic cell death. Semin Immunol 2023; 70:101832. [PMID: 37625331 DOI: 10.1016/j.smim.2023.101832] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/20/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
The programmed cell death machinery exhibits surprising flexibility, capable of crosstalk and non-apoptotic roles. Much of this complexity arises from the diverse functions of caspase-8, a cysteine-aspartic acid protease typically associated with activating caspase-3 and - 7 to induce apoptosis. However, recent research has revealed that caspase-8 also plays a role in regulating the lytic gasdermin cell death machinery, contributing to pyroptosis and immune responses in contexts such as infection, autoinflammation, and T-cell signalling. In mice, loss of caspase-8 results in embryonic lethality from unrestrained necroptotic killing, while in humans caspase-8 deficiency can lead to an autoimmune lymphoproliferative syndrome, immunodeficiency, inflammatory bowel disease or, when it can't cleave its substrate RIPK1, early onset periodic fevers. This review focuses on non-canonical caspase-8 signalling that drives immune responses, including its regulation of inflammatory gene transcription, activation within inflammasome complexes, and roles in pyroptotic cell death. Ultimately, a deeper understanding of caspase-8 function will aid in determining whether, and when, targeting caspase-8 pathways could be therapeutically beneficial in human diseases.
Collapse
Affiliation(s)
- Jiyi Pang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - James E Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
48
|
Zhou B, Abbott DW. Chemical modulation of gasdermin D activity: Therapeutic implications and consequences. Semin Immunol 2023; 70:101845. [PMID: 37806032 PMCID: PMC10841450 DOI: 10.1016/j.smim.2023.101845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 08/01/2023] [Accepted: 09/25/2023] [Indexed: 10/10/2023]
Abstract
The gasdermin family of proteins are central effectors of the inflammatory, lytic cell death modality known as pyroptosis. Characterized in 2015, the most well-studied member gasdermin D can be proteolyzed, typically by caspases, to generate an active pore-forming N-terminal domain. At least well-studied three pharmacological inhibitors (necrosulfonamide, disulfiram, dimethyl fumarate) since 2018 have been shown to affect gasdermin D activity either through modulation of processing or interference with pore formation. A multitude of murine in vivo studies have since followed. Here, we discuss the current state of research surrounding these three inhibitors, caveats to their use, and a set of guiding principles that researchers should consider when pursuing further studies of gasdermin D inhibition.
Collapse
Affiliation(s)
- Bowen Zhou
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Derek W Abbott
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
49
|
Wu D, Zhu J, Yang F, Li R, Liu L, Liu D, Liu C, Qu X, Liu H, Ji M, Qin X, Hua L, Xiang Y. CTNNAL1 deficiency suppresses CFTR expression in HDM-induced asthma mouse model through ROCK1-CAL signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1618-1629. [PMID: 37715489 PMCID: PMC10579809 DOI: 10.3724/abbs.2023152] [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: 11/18/2022] [Accepted: 04/28/2023] [Indexed: 09/17/2023] Open
Abstract
The downregulation of adhesion molecule catenin alpha-like 1 (CTNNAL1) in airway epithelial cells of asthma patients and house dust mite (HDM)-induced asthma animal models was illustrated in our previous study. It is assumed to contribute to airway inflammation and mucus hypersecretion. In this work, we further explore the underlying mechanism of CTNNAL1 in asthma. CTNNAL1-silenced female mice exhibit a decreased level of cystic fibrosis transmembrane conductance regulator (CFTR), a cAMP-activated and ATP-gated Cl - channel that correlates with mucus hypersecretion. Our previous study demonstrated that ROCK1 expression decreases but ROCK2 expression increases in the lungs of a CTNNAL1-silenced mouse model. Inhibition of ROCK1 leads to a reduction in CFTR expression in CTNNAL1-overexpressing and CTNNAL1-silenced human bronchial epithelial (HBE) cells. It has been reported that ROCK1 is a downstream target of RhoA and that activation of RhoA increases CFTR expression after CTNNAL1 deficiency in vitro and in vivo. The above results indicate that CTNNAL1 regulates CFTR expression through the ROCK1 pathway. In addition, the expression of CFTR-associated ligand (CAL) is increased after CTNNAL1 silencing, and immunoprecipitation results confirm the interaction between ROCK1 and CAL. Inhibition of CAL does not influence ROCK1 expression but increases CFTR expression in CTNNAL1-silenced HBE cells. These data suggest that CTNNAL1 deficiency decreases CFTR expression in the HDM-induced asthma mouse model through the ROCK1-CAL signaling pathway.
Collapse
Affiliation(s)
- Di Wu
- School of MedicineFoshan UniversityFoshan528000China
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Jiahui Zhu
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Fang Yang
- School of MedicineFoshan UniversityFoshan528000China
| | - Riwang Li
- School of MedicineFoshan UniversityFoshan528000China
| | - Lexin Liu
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Dahai Liu
- School of MedicineFoshan UniversityFoshan528000China
| | - Chi Liu
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Xiangping Qu
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Huijun Liu
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Ming Ji
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Xiaoqun Qin
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Lan Hua
- the Second Xiangya Hospital of Central South UniversityChangsha410011China
| | - Yang Xiang
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| |
Collapse
|
50
|
Traughber CA, Timinski K, Prince A, Bhandari N, Neupane K, Khan MR, Opoku E, Opoku E, Brubaker G, Nageshwar K, Ertugral EG, Naggareddy P, Kothapalli CR, Smith JD, Gulshan K. Disulfiram reduces atherosclerosis and enhances efferocytosis, autophagy, and atheroprotective gut microbiota in hyperlipidemic mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.17.562757. [PMID: 37905037 PMCID: PMC10614849 DOI: 10.1101/2023.10.17.562757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Pyroptosis executor Gasdermin (GsdmD) promotes atherosclerosis in mice and humans. Disulfiram (DSF) was recently shown to potently inhibit GsdmD, but the in-vivo efficacy and mechanism of DSF's anti-atherosclerotic activity is yet to be explored. We used human/mouse macrophages and a hyperlipidemic mouse model of atherosclerosis to determine DSF anti-atherosclerotic efficacy and mechanism. DSF-fed hyperlipidemic apoE -/- mice showed significantly reduced IL-1β release upon in-vivo Nlrp3 inflammasome assembly and showed smaller atherosclerotic lesions (∼27% and 29% reduction in males and females, respectively). The necrotic core area was also smaller (∼50% and 46% reduction in DSF-fed males and females, respectively). DSF induced autophagy in macrophages, hepatocytes/liver, and in atherosclerotic plaques. DSF modulated other atheroprotective pathways such as efferocytosis, phagocytosis, and gut microbiota. DSF-treated macrophages showed enhanced phagocytosis/efferocytosis, with a mechanism being a marked increase in cell-surface expression of efferocytic receptor MerTK. Atomic-force microscopy analysis revealed altered biophysical membrane properties of DSF treated macrophages, showing increased ordered-state of the plasma membrane and increased adhesion strength. Furthermore, the 16sRNA sequencing of DSF-fed hyperlipidemic mice showed highly significant enrichment in atheroprotective gut microbiota Akkermansia and a reduction in atherogenic Romboutsia species. Taken together, our data shows that DSF can simultaneously modulate multiple atheroprotective pathways, and thus may serve as novel adjuvant therapeutic to treat atherosclerosis.
Collapse
|