51
|
Kang N, Kim J, Kwon M, Son Y, Eo SK, Baryawno N, Kim BS, Yoon S, Oh SO, Lee D, Kim K. Blockade of mTORC1 via Rapamycin Suppresses 27-Hydroxycholestrol-Induced Inflammatory Responses. Int J Mol Sci 2024; 25:10381. [PMID: 39408711 PMCID: PMC11477202 DOI: 10.3390/ijms251910381] [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: 08/21/2024] [Revised: 09/15/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Atherosclerosis is characterized by the deposition and accumulation of extracellular cholesterol and inflammatory cells in the arterial blood vessel walls, and 27-hydroxycholesterol (27OHChol) is the most abundant cholesterol metabolite. 27OHChol is an oxysterol that induces immune responses, including immune cell activation and chemokine secretion, although the underlying mechanisms are not fully understood. In this study, we investigated the roles of the mechanistic target of rapamycin (mTOR) in 27HChol-induced inflammation using rapamycin. Treating monocytic cells with rapamycin effectively reduced the expression of CCL2 and CD14, which was involved with the increased immune response by 27OHChol. Rapamycin also suppressed the phosphorylation of S6 and 4EBP1, which are downstream of mTORC1. Additionally, it also alleviates the increase in differentiation markers into macrophage. These results suggest that 27OHChol induces inflammation by activating the mTORC1 signaling pathway, and rapamycin may be useful for the treatment of atherosclerosis-related inflammation involving 27OHchol.
Collapse
Affiliation(s)
- Nakyung Kang
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| | - Jaesung Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| | - Munju Kwon
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Yonghae Son
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| | - Seong-Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea;
| | - Ninib Baryawno
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institute, 17177 Stockholm, Sweden;
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| |
Collapse
|
52
|
Liu D, Wu W, Wang T, Zhan G, Zhang Y, Gao J, Gong Q. Lithocarpus polystachyus Rehd. ameliorates cerebral ischemia/reperfusion injury through inhibiting PI3K/AKT/NF-κB pathway and regulating NLRP3-mediated pyroptosis. Front Pharmacol 2024; 15:1365642. [PMID: 39380903 PMCID: PMC11458458 DOI: 10.3389/fphar.2024.1365642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
Introduction Ischemic stroke (IS) is a serious threat to human life and health, and cerebral ischemia/reperfusion injury (CIRI) exacerbates IS by enhancing neuroinflammation and oxidative stress. Sweet tea (ST) comprises several bioactive components, such as phlorizin, trilobatin, and phloretin, with diverse pharmacological activities. However, it remains uncertain whether ST can confer protection against CIRI. In this study, we aimed to investigate the impact and potential underlying mechanism of ST in the context of CIRI. Methods CIRI model were established in male sprague dawley (SD) rats. The neurobehavioral assessment, the volume of cerebral infarction and the morphology of neurons were measured to complete the preliminary pharmacodynamic study. The therapeutic targets and pathways of ST on IS were obtained by protein-protein interaction, molecular docking and Metascape database. The predicted results were further verified in vivo. Results Our results revealed that ST treatment significantly ameliorated brain damage in rats subjected to CIRI by mitigating mitochondrial oxidative stress and neuroinflammation. Additionally, we identified the PI3K/AKT/NF-κB pathway and the NLRP3-mediated pyroptosis axis as crucial processes, with molecular docking suggested direct interactions between the main compounds of ST and NLRP3. Conclusion ST safeguards against CIRI-induced neuronal loss, neuroinflammation and oxidative stress through the inhibition of the PI3K/AKT/NF-κB pathway and the regulation of NLRP3-mediated pyroptosis.
Collapse
Affiliation(s)
- Daifang Liu
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wendan Wu
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Tingting Wang
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Guiyu Zhan
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Yuandong Zhang
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Jianmei Gao
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Qihai Gong
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| |
Collapse
|
53
|
Allsup BL, Gharpure S, Bryson BD. Proximity labeling defines the phagosome lumen proteome of murine and primary human macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611277. [PMID: 39282337 PMCID: PMC11398489 DOI: 10.1101/2024.09.04.611277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Proteomic analyses of the phagosome has significantly improved our understanding of the proteins which contribute to critical phagosome functions such as apoptotic cell clearance and microbial killing. However, previous methods of isolating phagosomes for proteomic analysis have relied on cell fractionation with some intrinsic limitations. Here, we present an alternative and modular proximity-labeling based strategy for mass spectrometry proteomic analysis of the phagosome lumen, termed PhagoID. We optimize proximity labeling in the phagosome and apply PhagoID to immortalized murine macrophages as well as primary human macrophages. Analysis of proteins detected by PhagoID in murine macrophages demonstrate that PhagoID corroborates previous proteomic studies, but also nominates novel proteins with unexpected residence at the phagosome for further study. A direct comparison between the proteins detected by PhagoID between mouse and human macrophages further reveals that human macrophage phagosomes have an increased abundance of proteins involved in the oxidative burst and antigen presentation. Our study develops and benchmarks a new approach to measure the protein composition of the phagosome and validates a subset of these findings, ultimately using PhagoID to grant further insight into the core constituent proteins and species differences at the phagosome lumen.
Collapse
Affiliation(s)
- Benjamin L Allsup
- Department of Biological Engineering, MIT, Cambridge, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| | - Supriya Gharpure
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| | - Bryan D Bryson
- Department of Biological Engineering, MIT, Cambridge, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| |
Collapse
|
54
|
Song Y, Liu P, Qi X, Shi XL, Wang YS, Guo D, Luo H, Du ZJ, Wang MY. Helicobacter pylori infection delays neutrophil apoptosis and exacerbates inflammatory response. Future Microbiol 2024; 19:1145-1156. [PMID: 39056165 PMCID: PMC11529197 DOI: 10.1080/17460913.2024.2360798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/24/2024] [Indexed: 07/28/2024] Open
Abstract
Aim: Understanding molecular mechanisms of Helicobacter pylori (H. pylori)-induced inflammation is important for developing new therapeutic strategies for gastrointestinal diseases.Materials & methods: We designed an H. pylori-neutrophil infection model and explored the effects of H. pylori infection on neutrophils.Results: H. pylori infected neutrophils showed a low level of apoptosis. H. pylori stimulation activated the NACHT/LRR/PYD domain-containing protein 3 (NLRP3)-gasdermin-D (GSDMD) pathway for interleukin (IL)-1β secretion. However, IL-1β secretion was not completely dependent on GSDMD, as inhibition of autophagy significantly reduced IL-1β release, and autophagy-related molecules were significantly upregulated in H. pylori-infected neutrophils.Conclusion: Therefore, H. pylori infection inhibits neutrophils apoptosis and induces IL-1β secretion through autophagy. These findings may be utilized to formulate therapeutic strategies against H. pylori mediated chronic gastritis.
Collapse
Affiliation(s)
- Yu Song
- Marine College, Shandong University, Weihai, Shandong, 264209, PR China
- Department of Central Lab, Weihai Municipal Hospital, Shandong University, Weihai, Shandong, 264200, PR China
| | - Peng Liu
- Department of Central Lab, Weihai Municipal Hospital, Shandong University, Weihai, Shandong, 264200, PR China
| | - Xi Qi
- Department of Central Lab, Weihai Municipal Hospital, Shandong University, Weihai, Shandong, 264200, PR China
- School of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning, 116044, PR China
| | - Xiao-Lin Shi
- Department of Central Lab, Weihai Municipal Hospital, Shandong University, Weihai, Shandong, 264200, PR China
- School of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning, 116044, PR China
| | - Yu-Shan Wang
- Department of Central Lab, Weihai Municipal Hospital, Shandong University, Weihai, Shandong, 264200, PR China
- School of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning, 116044, PR China
| | - Dong Guo
- Department of Central Lab, Weihai Municipal Hospital, Shandong University, Weihai, Shandong, 264200, PR China
| | - Hong Luo
- School of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning, 116044, PR China
| | - Zong-Jun Du
- Marine College, Shandong University, Weihai, Shandong, 264209, PR China
| | - Ming-Yi Wang
- Marine College, Shandong University, Weihai, Shandong, 264209, PR China
- Department of Central Lab, Weihai Municipal Hospital, Shandong University, Weihai, Shandong, 264200, PR China
| |
Collapse
|
55
|
Encarnacion-Garcia MR, De la Torre-Baez R, Hernandez-Cueto MA, Velázquez-Villegas LA, Candelario-Martinez A, Sánchez-Argáez AB, Horta-López PH, Montoya-García A, Jaimes-Ortega GA, Lopez-Bailon L, Piedra-Quintero Z, Carrasco-Torres G, De Ita M, Figueroa-Corona MDP, Muñoz-Medina JE, Sánchez-Uribe M, Ortiz-Fernández A, Meraz-Ríos MA, Silva-Olivares A, Betanzos A, Baay-Guzman GJ, Navarro-Garcia F, Villa-Treviño S, Garcia-Sierra F, Cisneros B, Schnoor M, Ortíz-Navarrete VF, Villegas-Sepúlveda N, Valle-Rios R, Medina-Contreras O, Noriega LG, Nava P. IFN-γ stimulates Paneth cell secretion through necroptosis mTORC1 dependent. Eur J Immunol 2024; 54:e2350716. [PMID: 38837757 DOI: 10.1002/eji.202350716] [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: 08/14/2023] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024]
Abstract
Immune mediators affect multiple biological functions of intestinal epithelial cells (IECs) and, like Paneth and Paneth-like cells, play an important role in intestinal epithelial homeostasis. IFN-γ a prototypical proinflammatory cytokine disrupts intestinal epithelial homeostasis. However, the mechanism underlying the process remains unknown. In this study, using in vivo and in vitro models we demonstrate that IFN-γ is spontaneously secreted in the small intestine. Furthermore, we observed that this cytokine stimulates mitochondrial activity, ROS production, and Paneth and Paneth-like cell secretion. Paneth and Paneth-like secretion downstream of IFN-γ, as identified here, is mTORC1 and necroptosis-dependent. Thus, our findings revealed that the pleiotropic function of IFN-γ also includes the regulation of Paneth cell function in the homeostatic gut.
Collapse
Affiliation(s)
- Maria R Encarnacion-Garcia
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Raúl De la Torre-Baez
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - María A Hernandez-Cueto
- Clinical Laboratory of Infectology, National Hospital "La Raza" Medical Center, IMSS, Mexico City, Mexico
| | - Laura A Velázquez-Villegas
- Physiology of Nutrition Department, The National Institute of Health Sciences and Nutrition "Salvador Zubirán", Mexico City, Mexico
| | - Aurora Candelario-Martinez
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Ana Beatriz Sánchez-Argáez
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Perla H Horta-López
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Armando Montoya-García
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Gustavo Alberto Jaimes-Ortega
- Immunology and Proteomics Research Laboratory, Children's Hospital of Mexico "Federico Gómez" (HIMFG), Mexico City, Mexico
- Experimental Biology Postgraduate Program, Department of Biological and Health Sciences, Metropolitan Autonomous University (UAM), Mexico City, Mexico
| | - Luis Lopez-Bailon
- Immunology Department and Immunology Postgraduate Program, National School of Biological Sciences of the National Polytechnic Institute (ENCB-IPN), Mexico City, Mexico
| | - Zayda Piedra-Quintero
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Gabriela Carrasco-Torres
- CICATA Unidad Morelos, Instituto Politécnico Nacional, Boulevard de la Tecnología, 1036 Z-1, P 2/2, Atlacholoaya, 62790, México
| | - Marlon De Ita
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
- Medical Research Unit in Human Genetics, UMAE Children's Hospital, National Medical Center "Siglo XXI", IMSS, Ciudad de México, 06720, Mexico
| | - María Del Pilar Figueroa-Corona
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - José Esteban Muñoz-Medina
- Clinical Laboratory of Infectology, National Hospital "La Raza" Medical Center, IMSS, Mexico City, Mexico
| | - Magdalena Sánchez-Uribe
- Pathological Anatomy, Specialized hospital "Dr. Antonio Fraga Mouret", National Hospital "La Raza" Medical Center, IMSS, Ciudad de México, México
| | - Arturo Ortiz-Fernández
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Marco Antonio Meraz-Ríos
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Angélica Silva-Olivares
- Departament of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Abigail Betanzos
- Departament of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | | | - Fernando Navarro-Garcia
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Saúl Villa-Treviño
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Francisco Garcia-Sierra
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Bulmaro Cisneros
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Michael Schnoor
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Vianney F Ortíz-Navarrete
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Nicolás Villegas-Sepúlveda
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Ricardo Valle-Rios
- Immunology and Proteomics Research Laboratory, Children's Hospital of Mexico "Federico Gómez" (HIMFG), Mexico City, Mexico
- University Research Unit, Research Division, Faculty of Medicine, National Autonomous University of Mexico-Children's Hospital of Mexico "Federico Gomez" (UNAM-HIMFG), Mexico City, Mexico
| | - Oscar Medina-Contreras
- Epidemiology, Endocrinology & Nutrition Research Unit, Children's Hospital of Mexico "Federico Gomez", Mexico City, Mexico
| | - Lilia G Noriega
- Physiology of Nutrition Department, The National Institute of Health Sciences and Nutrition "Salvador Zubirán", Mexico City, Mexico
| | - Porfirio Nava
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| |
Collapse
|
56
|
Gupta G, Afzal M, Moglad E, Ali H, Singh TG, Kumbhar P, Disouza J, Almujri SS, Kazmi I, Alzarea SI, Hemalatha KP, Goh BH, Singh SK, Dua K. Non-coding RNAs as key regulators of Gasdermin-D mediated pyroptosis in cancer therapy. Pathol Res Pract 2024; 261:155490. [PMID: 39126977 DOI: 10.1016/j.prp.2024.155490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024]
Abstract
Pyroptosis is an inflammatory programed cell death process that plays a crucial role in cancer therapeutic, while Gasdermin-D is a critical effector protein for pyroptosis execution. This review discusses the intricate interactions between Gasdermin-D and some non-coding RNAs (lncRNA, miRNA, siRNA) and their potential application in the regulation of pyroptosis as an anticancer therapy. Correspondingly, these ncRNAs significantly implicate in Gasdermin-D expression and function regarding the pyroptosis pathway. Functioning as competing endogenous RNAs (ceRNAs), these ncRNAs might regulate Gasdermin-D at the molecular level, underlying fatal cell death caused by cancer and tumor propagation. Therefore, these interactions appeal to therapeutics, offering new avenues for cancer treatment. It address this research gap by discussing the possible roles of ncRNAs as mediators of gasdermin-D regulation. It suggest therapeutic strategies based on the current research findings to ensure the interchange between the ideal pyroptosis and cancer cell death.
Collapse
Affiliation(s)
- Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | | | - Popat Kumbhar
- Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala Dist, Kolhapur, Maharashtra 416113, India
| | - John Disouza
- Bombay Institute of Research and Pharmacy, Dombivli, Mumbai, Maharashtra 421203, India
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Aseer 61421, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf 72341, Saudi Arabia
| | - K P Hemalatha
- Sree Siddaganga College of Pharmacy, Tumkur, Karnataka, India
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway, Malaysia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| |
Collapse
|
57
|
Zhou B, Jiang ZH, Dai MR, Ai YL, Xiao L, Zhong CQ, Wu LZ, Chen QT, Chen HZ, Wu Q. Full-length GSDME mediates pyroptosis independent from cleavage. Nat Cell Biol 2024; 26:1545-1557. [PMID: 38997456 DOI: 10.1038/s41556-024-01463-2] [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: 12/21/2023] [Accepted: 06/19/2024] [Indexed: 07/14/2024]
Abstract
Gasdermin (GSDM) family proteins, known as the executors of pyroptosis, undergo protease-mediated cleavage before inducing pyroptosis. We here discovered a form of pyroptosis mediated by full-length (FL) GSDME without proteolytic cleavage. Intense ultraviolet-C irradiation-triggered DNA damage activates nuclear PARP1, leading to extensive formation of poly(ADP-ribose) (PAR) polymers. These PAR polymers are released to the cytoplasm, where they activate PARP5 to facilitate GSDME PARylation, resulting in a conformational change in GSDME that relieves autoinhibition. Moreover, ultraviolet-C irradiation promotes cytochrome c-catalysed cardiolipin peroxidation to elevate lipid reactive oxygen species, which is then sensed by PARylated GSDME, leading to oxidative oligomerization and plasma membrane targeting of FL-GSDME for perforation, eventually inducing pyroptosis. Reagents that concurrently stimulate PARylation and oxidation of FL-GSDME, synergistically promoting pyroptotic cell death. Overall, the present findings elucidate an unreported mechanism underlying the cleavage-independent function of GSDME in executing cell death, further enriching the paradigms and understanding of FL-GSDME-mediated pyroptosis.
Collapse
Affiliation(s)
- Bo Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Zhi-Hong Jiang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Meng-Ran Dai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yuan-Li Ai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Li Xiao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Chuan-Qi Zhong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Liu-Zheng Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Qi-Tao Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Hang-Zi Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China.
| | - Qiao Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
58
|
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
|
59
|
Ren Y, Wang Y, Chen C, Yan X, Chao M, Li Y, Yu D, Huang Y, Hou X, Gao F, Jiang G, Guan M. Solid-state electron-mediated z-scheme heterostructured semiconductor nanomaterials induce dual programmed cell death for melanoma therapy. J Nanobiotechnology 2024; 22:526. [PMID: 39217372 PMCID: PMC11365183 DOI: 10.1186/s12951-024-02770-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
The programmed cell death (PCD) pathway removes functionally insignificant, infection-prone, or potentially tumorigenic cells, underscoring its important role in maintaining the stability of the internal environment and warding off cancer and a host of other diseases. PCD includes various forms, such as apoptosis, copper death, iron death, and cellular pyroptosis. However, emerging solid-state electron-mediated Z-scheme heterostructured semiconductor nanomaterials with high electron-hole (e-h+) separation as a new method for inducing PCD have not been well studied. We synthesize the Bi2S3-Bi2O3-Au-PEG nanorods (BB-A-P NRs) Z-scheme heterostructured semiconductor has a higher redox capacity and biocompatibility. Firstly, the BB-A-P NRs are excited by near-infrared (NIR) light, which mimics the action of catalase by supplying oxygen (O2) and converting it to a single-linear state of oxygen (1O2) via e-h+ transfer. Secondly, they react with hydrogen peroxide (H2O2) and water (H2O) in tumor to produce hydroxyl radicals (•OH), inducing apoptosis. Intriguingly, the Caspase-1/Gasdermin D (GSDMD)-dependent conventional pyroptosis pathway induced cellular pyroptosis activated by apoptosis and reactive oxygen species (ROS) which causes the intense release of damage associated molecular patterns (DAMPs), leading to the inflammatory death of tumor cells. This, in turn, activates the immunological environment to achieve immunogenic cell death (ICD). BB-A-P enables computed tomography imaging, which allows for visualization of the treatment. BB-A-P activated dual PCD can be viewed as an effective mode of cell death that coordinates the intracellular environment, and the various pathways are interrelated and mutually reinforcing which shows promising therapeutic effects and provides a new strategy for eliminating anoxic tumors.
Collapse
Affiliation(s)
- Yiping Ren
- Department of Laboratory Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yun Wang
- Department of Dermatology, The Second People's Hospital of Huai'an, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, China
| | - Cheng Chen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Xiang Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Minghao Chao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Yuting Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Dehong Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Yuqi Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Xiaoyang Hou
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Fenglei Gao
- Department of Laboratory Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China.
| | - Guan Jiang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.
| | - Ming Guan
- Department of Laboratory Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
60
|
Valenstein ML, Lalgudi PV, Gu X, Kedir JF, Taylor MS, Chivukula RR, Sabatini DM. Rag-Ragulator is the central organizer of the physical architecture of the mTORC1 nutrient-sensing pathway. Proc Natl Acad Sci U S A 2024; 121:e2322755121. [PMID: 39163330 PMCID: PMC11363303 DOI: 10.1073/pnas.2322755121] [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: 12/24/2023] [Accepted: 07/12/2024] [Indexed: 08/22/2024] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) pathway regulates cell growth and metabolism in response to many environmental cues, including nutrients. Amino acids signal to mTORC1 by modulating the guanine nucleotide loading states of the heterodimeric Rag GTPases, which bind and recruit mTORC1 to the lysosomal surface, its site of activation. The Rag GTPases are tethered to the lysosome by the Ragulator complex and regulated by the GATOR1, GATOR2, and KICSTOR multiprotein complexes that localize to the lysosomal surface through an unknown mechanism(s). Here, we show that mTORC1 is completely insensitive to amino acids in cells lacking the Rag GTPases or the Ragulator component p18. Moreover, not only are the Rag GTPases and Ragulator required for amino acids to regulate mTORC1, they are also essential for the lysosomal recruitment of the GATOR1, GATOR2, and KICSTOR complexes, which stably associate and traffic to the lysosome as the "GATOR" supercomplex. The nucleotide state of RagA/B controls the lysosomal association of GATOR, in a fashion competitively antagonized by the N terminus of the amino acid transporter SLC38A9. Targeting of Ragulator to the surface of mitochondria is sufficient to relocalize the Rags and GATOR to this organelle, but not to enable the nutrient-regulated recruitment of mTORC1 to mitochondria. Thus, our results reveal that the Rag-Ragulator complex is the central organizer of the physical architecture of the mTORC1 nutrient-sensing pathway and underscore that mTORC1 activation requires signal transduction on the lysosomal surface.
Collapse
Affiliation(s)
- Max L. Valenstein
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
- Harvard Medical School, Boston, MA02115
| | - Pranav V. Lalgudi
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Xin Gu
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
- Harvard Medical School, Boston, MA02115
| | - Jibril F. Kedir
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
- Harvard Medical School, Boston, MA02115
| | - Martin S. Taylor
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Massachusetts General Hospital, Boston, MA02114
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI02903
- Brown Center on the Biology of Aging, Brown University, Providence, RI02903
- Legorreta Cancer Center, Brown University, Providence, RI02903
| | - Raghu R. Chivukula
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Medical School, Boston, MA02115
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA02114
- Department of Surgery, Massachusetts General Hospital, Boston, MA02114
- Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA02142
| | - David M. Sabatini
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague166 10, Czech Republic
| |
Collapse
|
61
|
Kong J, Zhang Y, Ju X, Wang B, Diao X, Li J, Qi G, Jin Y. Electrostimulation Evokes Caspase-3-Activated Fast Cancer Cell Pyroptosis and Its Nuclear Stress Response Pathways. Anal Chem 2024; 96:13438-13446. [PMID: 39129352 DOI: 10.1021/acs.analchem.4c01206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Pyroptosis of programmed cell death has been recognized as a more effective way to inhibit the occurrence and development of tumors than the better-studied apoptosis. However, it is still challenging to quickly and effectively trigger pyroptosis of cancer cells for high-efficacy cancer treatment. Here, we report on the first use of mild constant-potential electrostimulation (cp-ES) to quickly trigger cancer cell pyroptosis with a probability up to ∼91.4% and significantly shortened time (within 1 h), ∼3-6 times faster than typical drug stimulation to induce pyroptosis. We find that the ES-induced cancer cell pyroptosis is through the activated caspase-3 (pathway) cleavage of gasdermin E (GSDME) to form an N-terminal fragment (GSDME-N) and observe nuclear shrinkage and reduction of the number of nucleoli as well as down-/up-regulated expression of two important nucleoproteins of nucleolin and nucleophosmin (NPM1). The study enriches the basic understanding of pyroptosis and provides a new avenue for potential effective treatment of cancer.
Collapse
Affiliation(s)
- Jiao Kong
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Ying Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xingkai Ju
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Bo Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xingkang Diao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Jing Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Guohua Qi
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, P. R. China
| | - Yongdong Jin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, P. R. China
| |
Collapse
|
62
|
Brokatzky D, Gomes MC, Robertin S, Albino C, Miles SL, Mostowy S. Septins promote macrophage pyroptosis by regulating gasdermin D cleavage and ninjurin-1-mediated plasma membrane rupture. Cell Chem Biol 2024; 31:1518-1528.e6. [PMID: 39106869 DOI: 10.1016/j.chembiol.2024.07.003] [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: 12/13/2023] [Revised: 05/20/2024] [Accepted: 07/11/2024] [Indexed: 08/09/2024]
Abstract
The septin cytoskeleton is primarily known for roles in cell division and host defense against bacterial infection. Despite recent insights, the full breadth of roles for septins in host defense is poorly understood. In macrophages, Shigella induces pyroptosis, a pro-inflammatory form of cell death dependent upon gasdermin D (GSDMD) pores at the plasma membrane and cell surface protein ninjurin-1 (NINJ1) for membrane rupture. Here, we discover that septins promote macrophage pyroptosis induced by lipopolysaccharide (LPS)/nigericin and Shigella infection, but do not affect cytokine expression or release. We observe that septin filaments assemble at the plasma membrane, and cleavage of GSDMD is impaired in septin-depleted cells. We found that septins regulate mitochondrial dynamics and the expression of NINJ1. Using a Shigella-zebrafish infection model, we show that septin-mediated pyroptosis is an in vivo mechanism of infection control. The discovery of septins as a mediator of pyroptosis may inspire innovative anti-bacterial and anti-inflammatory treatments.
Collapse
Affiliation(s)
- Dominik Brokatzky
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, Keppel Street, London WC1E 7HT, UK.
| | - Margarida C Gomes
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, Keppel Street, London WC1E 7HT, UK
| | - Stevens Robertin
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, Keppel Street, London WC1E 7HT, UK
| | - Carolina Albino
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, Keppel Street, London WC1E 7HT, UK
| | - Sydney L Miles
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, Keppel Street, London WC1E 7HT, UK
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, Keppel Street, London WC1E 7HT, UK.
| |
Collapse
|
63
|
Ren H, Wu Z, Tan J, Tao H, Zou W, Cao Z, Wen B, Cai Z, Du J, Deng Z. Co-delivery Nano System of MS-275 and V-9302 Induces Pyroptosis and Enhances Anti-Tumor Immunity Against Uveal Melanoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404375. [PMID: 38889339 PMCID: PMC11336933 DOI: 10.1002/advs.202404375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/10/2024] [Indexed: 06/20/2024]
Abstract
In the treatment of uveal melanoma (UVM), histone deacetylase inhibitors (HDACi) have emerged as a promising epigenetic therapy. However, their clinical efficacy is hindered by the suboptimal pharmacokinetics and the strong self-rescue of tumor cells. To overcome these limitations, reactive oxygen species (ROS)-responsive nanoparticles (NPs) are designed that encapsulate HDACi MS-275 and the glutamine metabolism inhibitor V-9302. Upon reaching the tumor microenvironment, these NPs can disintegrate, thereby releasing MS-275 to increase the level of ROS and V-9302 to reduce the production of glutathione (GSH) related to self-rescue. These synergistic effects lead to a lethal ROS storm and induce cell pyroptosis. When combined with programmed cell death protein 1 monoclonal antibodies (α-PD-1), these NPs facilitate immune cell infiltration, improving anti-tumor immunity, converting "immune-cold" tumors into "immune-hot" tumors, and enhancing immune memory in mice. The findings present a nano-delivery strategy for the co-delivery of epigenetic therapeutics and metabolic inhibitors, which induces pyroptosis in tumors cells and improves the effectiveness of chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Hong Ren
- Department of OphthalmologyThe Third Xiangya HospitalCentral South UniversityChangshaHunan410013China
| | - Zhenkai Wu
- Department of OphthalmologyChangde HospitalXiangya School of MedicineCentral South UniversityChangdeHunan415000China
- Department of OphthalmologyThe first people's hospital of Changde cityChangdeHunan415000China
| | - Jia Tan
- Eye Center of Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Hunan Key Laboratory of Ophthalmology and National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Hui Tao
- Department of OphthalmologyThe Third Xiangya HospitalCentral South UniversityChangshaHunan410013China
| | - Wangyuan Zou
- Department of AnesthesiologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Zheng Cao
- Department of Chemical and Biomolecular EngineeringUniversity of CaliforniaLos AngelesCA90066USA
| | - Binyu Wen
- Department of OphthalmologyThe Third Xiangya HospitalCentral South UniversityChangshaHunan410013China
| | - Ziyi Cai
- Department of OphthalmologyThe Third Xiangya HospitalCentral South UniversityChangshaHunan410013China
| | - Jiaqi Du
- Department of OphthalmologyThe Third Xiangya HospitalCentral South UniversityChangshaHunan410013China
| | - Zhihong Deng
- Department of OphthalmologyThe Third Xiangya HospitalCentral South UniversityChangshaHunan410013China
| |
Collapse
|
64
|
Yu J, Fu Y, Gao J, Zhang Q, Zhang N, Zhang Z, Jiang X, Chen C, Wen Z. Cathepsin C from extracellular histone-induced M1 alveolar macrophages promotes NETosis during lung ischemia-reperfusion injury. Redox Biol 2024; 74:103231. [PMID: 38861835 PMCID: PMC11209641 DOI: 10.1016/j.redox.2024.103231] [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: 04/10/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024] Open
Abstract
Primary graft dysfunction (PGD) is a severe form of acute lung injury resulting from lung ischemia/reperfusion injury (I/R) in lung transplantation (LTx), associated with elevated post-transplant morbidity and mortality rates. Neutrophils infiltrating during reperfusion are identified as pivotal contributors to lung I/R injury by releasing excessive neutrophil extracellular traps (NETs) via NETosis. While alveolar macrophages (AMs) are involved in regulating neutrophil chemotaxis and infiltration, their role in NETosis during lung I/R remains inadequately elucidated. Extracellular histones constitute the main structure of NETs and can activate AMs. In this study, we confirmed the significant involvement of extracellular histone-induced M1 phenotype of AMs (M1-AMs) in driving NETosis during lung I/R. Using secretome analysis, public protein databases, and transwell co-culture models of AMs and neutrophils, we identified Cathepsin C (CTSC) derived from AMs as a major mediator in NETosis. Further elucidating the molecular mechanisms, we found that CTSC induced NETosis through a pathway dependent on NADPH oxidase-mediated production of reactive oxygen species (ROS). CTSC could significantly activate p38 MAPK, resulting in the phosphorylation of the NADPH oxidase subunit p47phox, thereby facilitating the trafficking of cytoplasmic subunits to the cell membrane and activating NADPH oxidase. Moreover, CTSC up-regulated and activated its substrate membrane proteinase 3 (mPR3), resulting in an increased release of NETosis-related inflammatory factors. Inhibiting CTSC revealed great potential in mitigating NETosis-related injury during lung I/R. These findings suggests that CTSC from AMs may be a crucial factor in mediating NETosis during lung I/R, and targeting CTSC inhition may represent a novel intervention for PGD in LTx.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Yu Fu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiameng Gao
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingqing Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Nan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuemei Jiang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
65
|
Ni K, Meng L. Mechanism of PANoptosis in metabolic dysfunction-associated steatotic liver disease. Clin Res Hepatol Gastroenterol 2024; 48:102381. [PMID: 38821484 DOI: 10.1016/j.clinre.2024.102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/16/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
In recent years, the incidence of metabolic dysfunction-associated steatotic liver disease (MASLD) has been steadily rising, emerging as a major chronic liver disease of global concern. The course of MASLD is varied, spanning from MASLD to metabolic dysfunction associated steatohepatitis (MASH). MASH is an important contributor to cirrhosis, which may subsequently lead to hepatocellular carcinoma. It has been found that PANoptosis, an emerging inflammatory programmed cell death (PCD), is involved in the pathogenesis of MASLD and facilitates the development of NASH, eventually resulting in inflammatory fibrosis and hepatocyte death. This paper reviews the latest research progress on PANoptosis and MASLD to understand the mechanism of MASLD and provide new directions for future treatment and drug development.
Collapse
Affiliation(s)
- Keying Ni
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medical), Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou, China
| | - Lina Meng
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medical), Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
66
|
Glorieux C, Liu S, Trachootham D, Huang P. Targeting ROS in cancer: rationale and strategies. Nat Rev Drug Discov 2024; 23:583-606. [PMID: 38982305 DOI: 10.1038/s41573-024-00979-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 07/11/2024]
Abstract
Reactive oxygen species (ROS) in biological systems are transient but essential molecules that are generated and eliminated by a complex set of delicately balanced molecular machineries. Disruption of redox homeostasis has been associated with various human diseases, especially cancer, in which increased ROS levels are thought to have a major role in tumour development and progression. As such, modulation of cellular redox status by targeting ROS and their regulatory machineries is considered a promising therapeutic strategy for cancer treatment. Recently, there has been major progress in this field, including the discovery of novel redox signalling pathways that affect the metabolism of tumour cells as well as immune cells in the tumour microenvironment, and the intriguing ROS regulation of biomolecular phase separation. Progress has also been made in exploring redox regulation in cancer stem cells, the role of ROS in determining cell fate and new anticancer agents that target ROS. This Review discusses these research developments and their implications for cancer therapy and drug discovery, as well as emerging concepts, paradoxes and future perspectives.
Collapse
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shihua Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | | | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Metabolic Innovation Center, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
67
|
Zhivaki D, Kennedy SN, Park J, Boriello F, Devant P, Cao A, Bahleda KM, Murphy S, McCabe C, Evavold CL, Chapman KL, Zanoni I, Ashenberg O, Xavier RJ, Kagan JC. Correction of age-associated defects in dendritic cells enables CD4 + T cells to eradicate tumors. Cell 2024; 187:3888-3903.e18. [PMID: 38870946 PMCID: PMC11283364 DOI: 10.1016/j.cell.2024.05.026] [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: 11/02/2022] [Revised: 04/02/2024] [Accepted: 05/13/2024] [Indexed: 06/15/2024]
Abstract
Defective host defenses later in life are associated with changes in immune cell activities, suggesting that age-specific considerations are needed in immunotherapy approaches. In this study, we found that PD-1 and CTLA4-based cancer immunotherapies are unable to eradicate tumors in elderly mice. This defect in anti-tumor activity correlated with two known age-associated immune defects: diminished abundance of systemic naive CD8+ T cells and weak migratory activities of dendritic cells (DCs). We identified a vaccine adjuvant, referred to as a DC hyperactivator, which corrects DC migratory defects in the elderly. Vaccines containing tumor antigens and DC hyperactivators induced T helper type 1 (TH1) CD4+ T cells with cytolytic activity that drive anti-tumor immunity in elderly mice. When administered early in life, DC hyperactivators were the only adjuvant identified that elicited anti-tumor CD4+ T cells that persisted into old age. These results raise the possibility of correcting age-associated immune defects through DC manipulation.
Collapse
Affiliation(s)
- Dania Zhivaki
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Stephanie N Kennedy
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Josh Park
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Francesco Boriello
- Harvard Medical School and Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Pascal Devant
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Anh Cao
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Kristin M Bahleda
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Shane Murphy
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Cristin McCabe
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Charles L Evavold
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Kate L Chapman
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Ivan Zanoni
- Harvard Medical School and Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Orr Ashenberg
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ramnik J Xavier
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan C Kagan
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
68
|
Yu J, Fu Y, Zhang N, Gao J, Zhang Z, Jiang X, Chen C, Wen Z. Extracellular histones promote TWIK2-dependent potassium efflux and associated NLRP3 activation in alveolar macrophages during sepsis-induced lung injury. Inflamm Res 2024; 73:1137-1155. [PMID: 38733398 DOI: 10.1007/s00011-024-01888-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 03/15/2024] [Accepted: 04/16/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND AND AIM Sepsis-induced acute lung injury (ALI) is a complex and life-threatening condition lacking specific and efficient clinical treatments. Extracellular histones, identified as a novel type of damage-associated molecular patterns, have been implicated in the inflammatory process of ALI. However, further elucidation is needed regarding the precise mechanism through which extracellular histones induce inflammation. The aim of this study was to investigate whether extracellular histones can activate NLRP3 inflammasome-mediated inflammation in alveolar macrophages (AMs) by affecting TWIK2-dependent potassium efflux. METHODS AND RESULTS We conducted experiments using cecal ligation and puncture (CLP) C57BL/6 mice and extracellular histone-stimulated LPS-primed MH-S cells. The results demonstrated a significant increase in the levels of extracellular histones in the plasma and bronchoalveolar lavage fluid (BALF) of CLP mice. Furthermore, neutralizing extracellular histone mitigated lung injury and inflammation in CLP-induced ALI mice. In vitro studies confirmed that extracellular histones upregulated the expression of NLRP3 inflammasome activation-related proteins in MH-S cells, and this effect was dependent on increased potassium efflux mediated by the TWIK2 channel on the plasma membrane. Moreover, extracellular histones directly triggered a substantial influx of calcium, leading to increased Rab11 activity and facilitating the trafficking and location of TWIK2 to the plasma membrane. CONCLUSION These findings underscore the critical role of extracellular histone-induced upregulation of TWIK2 expression on the plasma membrane of alveolar macrophages (AMs). This upregulation leads to potassium efflux and subsequent activation of the NLRP3 inflammasome, ultimately exacerbating lung inflammation and injury during sepsis.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu Fu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Nan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiameng Gao
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuemei Jiang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
69
|
Yang J, Chen N, Zhao P, Yang X, Li Y, Fu Z, Yan Y, Dong N, Li S, Yao R, Du X, Yao Y. DIMINISHED EXPRESSION OF GLS IN CD4 + T CELLS SERVES AS A PROGNOSTIC INDICATOR ASSOCIATED WITH CUPROPTOSIS IN SEPTIC PATIENTS. Shock 2024; 62:51-62. [PMID: 38662604 DOI: 10.1097/shk.0000000000002370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
ABSTRACT Objectives: Sepsis is defined as a life-threatening disease associated with a dysfunctional host immune response. Stratified identification of critically ill patients might significantly improve the survival rate. The present study sought to probe molecular markers associated with cuproptosis in septic patients to aid in stratification and improve prognosis. Methods: We studied expression of cuproptosis-related genes (CRGs) using peripheral blood samples from septic patients. Further classification was made by examining levels of expression of these potential CRGs in patients. Coexpression networks were constructed using the Weighted Gene Coexpression Network Analysis (WGCNA) method to identify crucial prognostic CRGs. Additionally, we utilized immune cell infiltration analysis to further examine the immune status of septic patients with different subtypes and its association with the CRGs. scRNA-seq data were also analyzed to verify expression of key CRGs among specific immune cells. Finally, immunoblotting, flow cytometry, immunofluorescence, and CFSE analysis were used to investigate possible regulatory mechanisms. Results: We classified septic patients based on CRG expression levels and found significant differences in prognosis and gene expression patterns. Three key CRGs that may influence the prognosis of septic patients were identified. A decrease in GLS expression was subsequently verified in Jurkat cells, accompanied by a reduction in O-GlcNAc levels, and chelation of copper by tetrathiomolybdate could not rescue the reduction in GLS and O-GLcNAc levels. Moreover, immoderate chelation of copper was detrimental to mitochondrial function, cell viability, and cell proliferation, as well as the immune status of the host. Conclusion: We have identified novel molecular markers associated with cuproptosis, which could potentially function as diagnostic indicators for septic patients. The reversible nature of the observed alterations in FDX1 and LIAS was demonstrated through copper chelation, whereas the correlation between copper and the observed changes in GLS requires further investigation.
Collapse
Affiliation(s)
| | - Ning Chen
- Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | | | | | | | | | - Yang Yan
- Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ning Dong
- Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Songyan Li
- Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, China
| | | | | | - Yongming Yao
- Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
70
|
Wang J, Wu Z, Zhu M, Zhao Y, Xie J. ROS induced pyroptosis in inflammatory disease and cancer. Front Immunol 2024; 15:1378990. [PMID: 39011036 PMCID: PMC11246884 DOI: 10.3389/fimmu.2024.1378990] [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: 02/06/2024] [Accepted: 06/18/2024] [Indexed: 07/17/2024] Open
Abstract
Pyroptosis, a form of caspase-1-dependent cell death, also known as inflammation-dependent death, plays a crucial role in diseases such as stroke, heart disease, or tumors. Since its elucidation, pyroptosis has attracted widespread attention from various sectors. Reactive oxygen species (ROS) can regulate numerous cellular signaling pathways. Through further research on ROS and pyroptosis, the level of ROS has been revealed to be pivotal for the occurrence of pyroptosis, establishing a close relationship between the two. This review primarily focuses on the molecular mechanisms of ROS and pyroptosis in tumors and inflammatory diseases, exploring key proteins that may serve as drug targets linking ROS and pyroptosis and emerging fields targeting pyroptosis. Additionally, the potential future development of compounds and proteins that influence ROS-regulated cell pyroptosis is anticipated, aiming to provide insights for the development of anti-tumor and anti-inflammatory drugs.
Collapse
Affiliation(s)
- Jingsong Wang
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, China
| | - Ziyong Wu
- Department of Pharmacy, Ezhou Central Hospital, Ezhou, Hubei, China
| | - Min Zhu
- Department of Pharmacy, Xuchang Central Hospital, Xuchang, Henan, China
| | - Yang Zhao
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, China
| | - Jingwen Xie
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, China
- Department of Health, Chongqing Industry & Trade Polytechnic, Chongqing, China
| |
Collapse
|
71
|
Liao C, Xu F, Yu Z, Ding K, Jia Y. The Novel Role of the NLRP3 Inflammasome in Mycotoxin-Induced Toxicological Mechanisms. Vet Sci 2024; 11:291. [PMID: 39057975 PMCID: PMC11281663 DOI: 10.3390/vetsci11070291] [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: 05/30/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Mycotoxins are secondary metabolites produced by several fungi and moulds that exert toxicological effects on animals including immunotoxicity, genotoxicity, hepatotoxicity, teratogenicity, and neurotoxicity. However, the toxicological mechanisms of mycotoxins are complex and unclear. The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome is a multimeric cytosolic protein complex composed of the NLRP3 sensor, ASC adapter protein, and caspase-1 effector. Activation of the NLRP3 inflammasome plays a crucial role in innate immune defence and homeostatic maintenance. Recent studies have revealed that NLRP3 inflammasome activation is linked to tissue damage and inflammation induced by mycotoxin exposure. Thus, this review summarises the latest advancements in research on the roles of NLRP3 inflammasome activation in the pathogenesis of mycotoxin exposure. The effects of exposure to multiple mycotoxins, including deoxynivalenol, aflatoxin B1, zearalenone, T-2 toxin, ochratoxin A, and fumonisim B1, on pyroptosis-related factors and inflammation-related factors in vitro and in vivo and the pharmacological inhibition of specific and nonspecific NLRP3 inhibitors are summarized and examined. This comprehensive review contributes to a better understanding of the role of the NLRP3 inflammasome in toxicity induced by mycotoxin exposure and provides novel insights for pharmacologically targeting NLRP3 as a novel anti-inflammatory agent against mycotoxin exposure.
Collapse
Affiliation(s)
- Chengshui Liao
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China; (C.L.); (F.X.); (Z.Y.); (K.D.)
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471023, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Fengru Xu
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China; (C.L.); (F.X.); (Z.Y.); (K.D.)
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471023, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Zuhua Yu
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China; (C.L.); (F.X.); (Z.Y.); (K.D.)
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471023, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Ke Ding
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China; (C.L.); (F.X.); (Z.Y.); (K.D.)
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471023, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Yanyan Jia
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China; (C.L.); (F.X.); (Z.Y.); (K.D.)
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471023, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| |
Collapse
|
72
|
Tran N, Mills EL. Redox regulation of macrophages. Redox Biol 2024; 72:103123. [PMID: 38615489 PMCID: PMC11026845 DOI: 10.1016/j.redox.2024.103123] [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: 01/22/2024] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 04/16/2024] Open
Abstract
Redox signaling, a mode of signal transduction that involves the transfer of electrons from a nucleophilic to electrophilic molecule, has emerged as an essential regulator of inflammatory macrophages. Redox reactions are driven by reactive oxygen/nitrogen species (ROS and RNS) and redox-sensitive metabolites such as fumarate and itaconate, which can post-translationally modify specific cysteine residues in target proteins. In the past decade our understanding of how ROS, RNS, and redox-sensitive metabolites control macrophage function has expanded dramatically. In this review, we discuss the latest evidence of how ROS, RNS, and metabolites regulate macrophage function and how this is dysregulated with disease. We highlight the key tools to assess redox signaling and important questions that remain.
Collapse
Affiliation(s)
- Nhien Tran
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Evanna L Mills
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
73
|
Qi W, Jin L, Huang S, Aikebaier A, Xue S, Wang Q, Chen Q, Lu Y, Ding C. Modulating synovial macrophage pyroptosis and mitophagy interactions to mitigate osteoarthritis progression using functionalized nanoparticles. Acta Biomater 2024; 181:425-439. [PMID: 38729544 DOI: 10.1016/j.actbio.2024.05.014] [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: 02/04/2024] [Revised: 04/29/2024] [Accepted: 05/05/2024] [Indexed: 05/12/2024]
Abstract
Synovial macrophages play an important role in the progression of osteoarthritis (OA). In this study, we noted that synovial macrophages can activate pyroptosis in a gasdermin d-dependent manner and produce reactive oxygen species (ROS), aberrantly activating the mammalian target of rapamycin complex 1 (mTORC1) pathway and matrix metalloproteinase-9 (MMP9) expression in synovial tissue samples collected from both patients with OA and collagen-induced osteoarthritis (CIOA) mouse model. To overcome this, we constructed rapamycin- (RAPA, a mTORC1 inhibitor) loaded mesoporous Prussian blue nanoparticles (MPB NPs, for catalyzing ROS) and modified the NPs with MMP9-targeted peptides (favor macrophage targeting) to develop RAPA@MPB-MMP9 NPs. The inherent enzyme-like activity and RAPA released from RAPA@MPB-MMP9 NPs synergistically impeded the pyroptosis of macrophages and the activation of the mTORC1 pathway. In particular, the NPs decreased pyroptosis-mediated ROS generation, thereby inhibiting cGAS-STING signaling pathway activation caused by the release of mitochondrial DNA. Moreover, the NPs promoted macrophage mitophagy to restore mitochondrial stability, alleviate pyroptosis-related inflammatory responses, and decrease senescent synoviocytes. After the as-prepared NPs were intra-articularly injected into the CIOA mouse model, they efficiently attenuated synovial macrophage pyroptosis and cartilage degradation. In conclusion, our study findings provide a novel therapeutic strategy for OA that modulates the pyroptosis and mitophagy of synovial macrophage by utilizing functionalized NPs. STATEMENT OF SIGNIFICANCE: Osteoarthritis (OA) presents a significant global challenge owing to its complex pathogenesis and finite treatment options. Synovial macrophages have emerged as key players in the progression of OA, managing inflammation and tissue destruction. In this study, we discovered a novel therapeutic strategy in which the pyroptosis and mitophagy of synovial macrophages are targeted to mitigate OA pathology. For this, we designed and prepared rapamycin-loaded mesoporous Prussian blue nanoparticles (RAPA@MPB-MMP9 NPs) to specifically target synovial macrophages and modulate their inflammatory responses. These NPs could efficiently suppress macrophage pyroptosis, diminish reactive oxygen species production, and promote mitophagy, thereby alleviating inflammation and protecting cartilage integrity. Our study findings not only clarify the intricate mechanisms underlying OA pathogenesis but also present a promising therapeutic approach for effectively managing OA by targeting dysregulation in synovial macrophages.
Collapse
Affiliation(s)
- Weizhong Qi
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Li Jin
- Rheumatology and Clinical Immunology, ZhuJiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Shiqian Huang
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Alafate Aikebaier
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Song Xue
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - QianYi Wang
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Qiyue Chen
- Stomatological Hospital, Southern Medical University, Guangzhou, 510282, China.
| | - Yao Lu
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China; Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China.
| | - Changhai Ding
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China; Menzies Institute for Medical Research, University of Tasmania, 7000, Hobart, Tasmania, Australia.
| |
Collapse
|
74
|
Rios FJ, de Ciuceis C, Georgiopoulos G, Lazaridis A, Nosalski R, Pavlidis G, Tual-Chalot S, Agabiti-Rosei C, Camargo LL, Dąbrowska E, Quarti-Trevano F, Hellmann M, Masi S, Lopreiato M, Mavraganis G, Mengozzi A, Montezano AC, Stavropoulos K, Winklewski PJ, Wolf J, Costantino S, Doumas M, Gkaliagkousi E, Grassi G, Guzik TJ, Ikonomidis I, Narkiewicz K, Paneni F, Rizzoni D, Stamatelopoulos K, Stellos K, Taddei S, Touyz RM, Virdis A. Mechanisms of Vascular Inflammation and Potential Therapeutic Targets: A Position Paper From the ESH Working Group on Small Arteries. Hypertension 2024; 81:1218-1232. [PMID: 38511317 DOI: 10.1161/hypertensionaha.123.22483] [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: 03/22/2024]
Abstract
Inflammatory responses in small vessels play an important role in the development of cardiovascular diseases, including hypertension, stroke, and small vessel disease. This involves various complex molecular processes including oxidative stress, inflammasome activation, immune-mediated responses, and protein misfolding, which together contribute to microvascular damage. In addition, epigenetic factors, including DNA methylation, histone modifications, and microRNAs influence vascular inflammation and injury. These phenomena may be acquired during the aging process or due to environmental factors. Activation of proinflammatory signaling pathways and molecular events induce low-grade and chronic inflammation with consequent cardiovascular damage. Identifying mechanism-specific targets might provide opportunities in the development of novel therapeutic approaches. Monoclonal antibodies targeting inflammatory cytokines and epigenetic drugs, show promise in reducing microvascular inflammation and associated cardiovascular diseases. In this article, we provide a comprehensive discussion of the complex mechanisms underlying microvascular inflammation and offer insights into innovative therapeutic strategies that may ameliorate vascular injury in cardiovascular disease.
Collapse
Affiliation(s)
- Francisco J Rios
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Carolina de Ciuceis
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
| | - Georgios Georgiopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Antonios Lazaridis
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Greece (A.L., E.G.)
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute, University of Edinburgh, United Kingdom (R.N., T.J.G.)
- Department of Internal Medicine, Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland (R.N., T.J.G.)
| | - George Pavlidis
- Medical School (G.P., I.I.), National and Kapodistrian University of Athens
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2-Cardiology Department, Attikon Hospital, Athens, Greece (G.P., I.I.)
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, United Kingdom (S.T.-C., K. Stellos)
| | - Claudia Agabiti-Rosei
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
| | - Livia L Camargo
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Edyta Dąbrowska
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Fosca Quarti-Trevano
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (F.Q.-T., G.G.)
| | - Marcin Hellmann
- Cardiac Diagnostics (M.H.), Medical University of Gdansk, Poland
| | - Stefano Masi
- Institute of Cardiovascular Science, University College London, United Kingdom (S.M.)
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Mariarosaria Lopreiato
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Georgios Mavraganis
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland (A.M., F.P.)
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa (A.M.)
| | - Augusto C Montezano
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Konstantinos Stavropoulos
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, Greece (K. Stavropoulos)
| | - Pawel J Winklewski
- Departments of Human Physiology (P.J.W.), Medical University of Gdansk, Poland
| | - Jacek Wolf
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Sarah Costantino
- University Heart Center (S.C., F.P.), University Hospital Zurich, Switzerland
| | - Michael Doumas
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Eugenia Gkaliagkousi
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Greece (A.L., E.G.)
| | - Guido Grassi
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (F.Q.-T., G.G.)
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute, University of Edinburgh, United Kingdom (R.N., T.J.G.)
- Department of Internal Medicine, Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland (R.N., T.J.G.)
| | - Ignatios Ikonomidis
- Medical School (G.P., I.I.), National and Kapodistrian University of Athens
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2-Cardiology Department, Attikon Hospital, Athens, Greece (G.P., I.I.)
| | - Krzysztof Narkiewicz
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland (A.M., F.P.)
- University Heart Center (S.C., F.P.), University Hospital Zurich, Switzerland
- Department of Research and Education (F.P.), University Hospital Zurich, Switzerland
| | - Damiano Rizzoni
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
- Division of Medicine, Spedali Civili di Brescia, Italy (D.R.)
| | - Kimon Stamatelopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, United Kingdom (S.T.-C., K. Stellos)
- Department of Cardiovascular Research, European Center for Angioscience, Medical Faculty Mannheim (K. Stellos), Heidelberg University, Germany
- Department of Cardiology, University Hospital Mannheim (K. Stellos), Heidelberg University, Germany
- German Centre for Cardiovascular Research, Heidelberg/Mannheim Partner Site (K. Stellos)
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Rhian M Touyz
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| |
Collapse
|
75
|
Deng D, Zhao M, Liu H, Zhou S, Liu H, You L, Hao Y. Xijiao Dihuang decoction combined with Yinqiao powder promotes autophagy-dependent ROS decrease to inhibit ROS/NLRP3/pyroptosis regulation axis in influenza virus infection. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155446. [PMID: 38518643 DOI: 10.1016/j.phymed.2024.155446] [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: 07/16/2023] [Revised: 01/17/2024] [Accepted: 02/09/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND Influenza viral pneumonia is a common complication after influenza virus infection. Xijiao Dihuang Decoction combined with Yinqiao Powder (XDY) is effective on improving influenza viral pneumonia. PURPOSE This study further explores the anti-inflammatory mechanism of XDY in the treatment of influenza viral pneumonia. STUDY DESIGN The effects of XDY on inflammation, autophagy, NACHT-LRR-PYD-containing protein 3 (NLRP3) inflammasome and pyroptosis were assessed in the mice with influenza viral pneumonia. In addition, the mouse macrophage cell line (J774A.1) infected with influenza virus was adopted to decode the in vitro effects of XDY on autophagy, reactive oxygen species (ROS), NLRP3 inflammasome and pyroptosis. We analyzed the XDY-induced autophagy, especially the mitophagy-related ROS clearance, and the subsequent inhibition of ROS/NLRP3 inflammasome/pyroptosis signaling in the infected macrophages by different assays based on quantitative polymerase chain reaction, western blot, flow cytometry, immunofluorescence and enzyme-linked immunosorbent assay. RESULTS In vivo, XDY could effectively improve the lung inflammatory response in the mice with influenza virus pneumonia, due to an intact autophagy flux-promoting effect and the inhibiting roles on NLRP3 inflammasome and pyroptosis. Notably, in vitro, compared with the infected macrophages treated by the NLRP3 inflammasome agonist (Monosodium urate) or the mitochondrial-targeted antioxidant agent, the XDY-dependent treating could inhibit pyroptosis by negatively regulating the signaling axis of ROS/NLRP3 inflammasome/pyroptosis in the influenza virus-infected macrophages. More interestingly, XDY could promote an intact autophagy flux, inducing mitophagy eliminating the damaged mitochondria to reduce the intracellular ROS accumulation, and thus decrease the oxidative stress in the infected macrophages. Especially, the inhibitor of autophagy inition, 3-Methyladenine, could reverse the inhibitory effect of XDY on ROS-NLRP3 inflammasome-mediated pyroptosis, indicating an XDY-promoted mitophagy-dependent ROS scavenging. CONCLUSION XDY can promote an intact autophagy flux to eliminate damaged mitochondria, namely mitophagy, which reduces the intracellular ROS accumulation contributing to NLRP3 inflammasome activation, restricting pyroptosis and eventually alleviating the influenza virus-induced inflammatory lesions. The obtained results provide new insights into the mechanism of action of XDY in alleviating influenza virus pneumonia, especially the roles of XDY in anti-oxidation, anti-inflammation and anti-pyroptosis, with potential therapeutic targets for future application in integrative medicine.
Collapse
Affiliation(s)
- Di Deng
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, No. 11 North Three-ring East Road, Chao Yang District, Beijing 100029, China
| | - Mengfan Zhao
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, No. 11 North Three-ring East Road, Chao Yang District, Beijing 100029, China
| | - Huanwei Liu
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, No. 11 North Three-ring East Road, Chao Yang District, Beijing 100029, China
| | - Siyao Zhou
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, No. 11 North Three-ring East Road, Chao Yang District, Beijing 100029, China
| | - Hui Liu
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, No. 11 North Three-ring East Road, Chao Yang District, Beijing 100029, China
| | - Leiming You
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, No. 11 North Three-ring East Road, Chao Yang District, Beijing 100029, China.
| | - Yu Hao
- Department of Immunology and Microbiology, School of Life Sciences, Beijing University of Chinese Medicine, No. 11 North Three-ring East Road, Chao Yang District, Beijing 100029, China.
| |
Collapse
|
76
|
Du G, Healy LB, David L, Walker C, El-Baba TJ, Lutomski CA, Goh B, Gu B, Pi X, Devant P, Fontana P, Dong Y, Ma X, Miao R, Balasubramanian A, Puthenveetil R, Banerjee A, Luo HR, Kagan JC, Oh SF, Robinson CV, Lieberman J, Wu H. ROS-dependent S-palmitoylation activates cleaved and intact gasdermin D. Nature 2024; 630:437-446. [PMID: 38599239 PMCID: PMC11283288 DOI: 10.1038/s41586-024-07373-5] [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: 08/10/2023] [Accepted: 04/02/2024] [Indexed: 04/12/2024]
Abstract
Gasdermin D (GSDMD) is the common effector for cytokine secretion and pyroptosis downstream of inflammasome activation and was previously shown to form large transmembrane pores after cleavage by inflammatory caspases to generate the GSDMD N-terminal domain (GSDMD-NT)1-10. Here we report that GSDMD Cys191 is S-palmitoylated and that palmitoylation is required for pore formation. S-palmitoylation, which does not affect GSDMD cleavage, is augmented by mitochondria-generated reactive oxygen species (ROS). Cleavage-deficient GSDMD (D275A) is also palmitoylated after inflammasome stimulation or treatment with ROS activators and causes pyroptosis, although less efficiently than palmitoylated GSDMD-NT. Palmitoylated, but not unpalmitoylated, full-length GSDMD induces liposome leakage and forms a pore similar in structure to GSDMD-NT pores shown by cryogenic electron microscopy. ZDHHC5 and ZDHHC9 are the major palmitoyltransferases that mediate GSDMD palmitoylation, and their expression is upregulated by inflammasome activation and ROS. The other human gasdermins are also palmitoylated at their N termini. These data challenge the concept that cleavage is the only trigger for GSDMD activation. They suggest that reversible palmitoylation is a checkpoint for pore formation by both GSDMD-NT and intact GSDMD that functions as a general switch for the activation of this pore-forming family.
Collapse
Affiliation(s)
- Gang Du
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
| | - Liam B Healy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Liron David
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
- Seqirus, Cambridge, MA, USA.
| | - Caitlin Walker
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Life Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Tarick J El-Baba
- Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for NanoScience Discovery, University of Oxford, Oxford, UK
| | - Corinne A Lutomski
- Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for NanoScience Discovery, University of Oxford, Oxford, UK
| | - Byoungsook Goh
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Bowen Gu
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Xiong Pi
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Pascal Devant
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Pietro Fontana
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Ying Dong
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Xiyu Ma
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Rui Miao
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Arumugam Balasubramanian
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School, Boston, MA, USA
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Robbins Puthenveetil
- Section on Structural and Chemical Biology, Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Anirban Banerjee
- Section on Structural and Chemical Biology, Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Hongbo R Luo
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School, Boston, MA, USA
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Jonathan C Kagan
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Sungwhan F Oh
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Carol V Robinson
- Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for NanoScience Discovery, University of Oxford, Oxford, UK
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
77
|
Lamontagne F, Paz-Trejo C, Zamorano Cuervo N, Grandvaux N. Redox signaling in cell fate: Beyond damage. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119722. [PMID: 38615720 DOI: 10.1016/j.bbamcr.2024.119722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/20/2024] [Accepted: 04/02/2024] [Indexed: 04/16/2024]
Abstract
This review explores the nuanced role of reactive oxygen species (ROS) in cell fate, challenging the traditional view that equates ROS with cellular damage. Through significant technological advancements in detecting localized redox states and identifying oxidized cysteines, a paradigm shift has emerged: from ROS as merely damaging agents to crucial players in redox signaling. We delve into the intricacies of redox mechanisms, which, although confined, exert profound influences on cellular physiological responses. Our analysis extends to both the positive and negative impacts of these mechanisms on cell death processes, including uncontrolled and programmed pathways. By unraveling these complex interactions, we argue against the oversimplified notion of a 'stress response', advocating for a more nuanced understanding of redox signaling. This review underscores the importance of localized redox states in determining cell fate, highlighting the sophistication and subtlety of ROS functions beyond mere damage.
Collapse
Affiliation(s)
- Felix Lamontagne
- CRCHUM - Centre de Recherche du Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal H2X 0A9, Québec, Canada
| | - Cynthia Paz-Trejo
- CRCHUM - Centre de Recherche du Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal H2X 0A9, Québec, Canada; Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal H3C 3J7, Québec, Canada
| | - Natalia Zamorano Cuervo
- CRCHUM - Centre de Recherche du Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal H2X 0A9, Québec, Canada
| | - Nathalie Grandvaux
- CRCHUM - Centre de Recherche du Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal H2X 0A9, Québec, Canada; Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal H3C 3J7, Québec, Canada.
| |
Collapse
|
78
|
Xin Y, Gao C, Wang L, Liu Q, Lu Q. Lipopolysaccharide released from gut activates pyroptosis of macrophages via Caspase 11-Gasdermin D pathway in systemic lupus erythematosus. MedComm (Beijing) 2024; 5:e610. [PMID: 38881675 PMCID: PMC11176733 DOI: 10.1002/mco2.610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 06/18/2024] Open
Abstract
Noncanonical pyroptosis is triggered by Caspase 4/5/11, which cleaves Gasdermin D (GSDMD), leading to cell lysis. While GSDMD has been studied previously in systemic lupus erythematosus (SLE), the role of pyroptosis in SLE pathogenesis remains unclear and contentious, with limited understanding of Caspase 11-mediated pyroptosis in this condition. In this study, we explored the level of Caspase 11-mediated pyroptosis in SLE, identifying both the upstream pathways and the interaction between pyroptosis and adaptive immune responses. We observed increased Caspase 5/11 and GSDMD-dependent pyroptosis in the macrophages/monocytes of both lupus patients and mice. We identified serum lipopolysaccharide (LPS), released from the gut due to a compromised gut barrier, as the signal that triggers Caspase 11 activation in MRL/lpr mice. We further discovered that pyroptotic macrophages promote the differentiation of mature B cells independently of T cells. Additionally, inhibiting Caspase 11 and preventing LPS leakage proved effective in improving lupus symptoms in MRL/lpr mice. These findings suggest that elevated serum LPS, resulting from a damaged gut barrier, induces Caspase 11/GSDMD-mediated pyroptosis, which in turn promotes B cell differentiation and enhances autoimmune responses in SLE. Thus, targeting Caspase 11 could be a viable therapeutic strategy for SLE.
Collapse
Affiliation(s)
- Yue Xin
- Hospital for Skin Diseases Institute of Dermatology Chinese Academy of Medical Sciences and Peking Union Medical College Nanjing China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases Chinese Academy of Medical Sciences Nanjing China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs Chinese Academy of Medical Sciences Nanjing China
| | - Changxing Gao
- Hospital for Skin Diseases Institute of Dermatology Chinese Academy of Medical Sciences and Peking Union Medical College Nanjing China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases Chinese Academy of Medical Sciences Nanjing China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs Chinese Academy of Medical Sciences Nanjing China
| | - Lai Wang
- Hospital for Skin Diseases Institute of Dermatology Chinese Academy of Medical Sciences and Peking Union Medical College Nanjing China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases Chinese Academy of Medical Sciences Nanjing China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs Chinese Academy of Medical Sciences Nanjing China
| | - Qianmei Liu
- Hospital for Skin Diseases Institute of Dermatology Chinese Academy of Medical Sciences and Peking Union Medical College Nanjing China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases Chinese Academy of Medical Sciences Nanjing China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs Chinese Academy of Medical Sciences Nanjing China
| | - Qianjin Lu
- Hospital for Skin Diseases Institute of Dermatology Chinese Academy of Medical Sciences and Peking Union Medical College Nanjing China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases Chinese Academy of Medical Sciences Nanjing China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs Chinese Academy of Medical Sciences Nanjing China
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China Changsha China
| |
Collapse
|
79
|
Wang L, Li M, Lian G, Yang S, Wu Y, Cui J. USP18 Antagonizes Pyroptosis by Facilitating Selective Autophagic Degradation of Gasdermin D. RESEARCH (WASHINGTON, D.C.) 2024; 7:0380. [PMID: 38779488 PMCID: PMC11109516 DOI: 10.34133/research.0380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024]
Abstract
As a key executioner of pyroptosis, Gasdermin D (GSDMD) plays a crucial role in host defense and emerges as an essential therapeutic target in the treatment of inflammatory diseases. So far, the understanding of the mechanisms that regulate the protein level of GSDMD to prevent detrimental effects and maintain homeostasis is currently limited. Here, we unveil that ubiquitin-specific peptidase 18 (USP18) works as a negative regulator of pyroptosis by targeting GSDMD for degradation and preventing excessive innate immune responses. Mechanically, USP18 recruits E3 ubiquitin ligase mind bomb homolog 2 (MIB2) to catalyze ubiquitination on GSDMD at lysine (K) 168, which acts as a recognition signal for the selective autophagic degradation of GSDMD. We further confirm the alleviating effect of USP18 on LPS-triggered inflammation in vivo. Collectively, our study demonstrates the role of USP18 in regulating GSDMD-mediated pyroptosis and reveals a previously unknown mechanism by which GSDMD protein level is rigorously controlled by selective autophagy.
Collapse
Affiliation(s)
- Liqiu Wang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol,
School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mengqiu Li
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol,
School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guangyu Lian
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol,
School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuai Yang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol,
School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yaoxing Wu
- Department of Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol,
School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
80
|
Su R, Shao Y, Huang M, Liu D, Yu H, Qiu Y. Immunometabolism in cancer: basic mechanisms and new targeting strategy. Cell Death Discov 2024; 10:236. [PMID: 38755125 PMCID: PMC11099033 DOI: 10.1038/s41420-024-02006-2] [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: 07/31/2023] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/18/2024] Open
Abstract
Maturing immunometabolic research empowers immune regulation novel approaches. Progressive metabolic adaptation of tumor cells permits a thriving tumor microenvironment (TME) in which immune cells always lose the initial killing capacity, which remains an unsolved dilemma even with the development of immune checkpoint therapies. In recent years, many studies on tumor immunometabolism have been reported. The development of immunometabolism may facilitate anti-tumor immunotherapy from the recurrent crosstalk between metabolism and immunity. Here, we discuss clinical studies of the core signaling pathways of immunometabolism and their inhibitors or agonists, as well as the specific functions of these pathways in regulating immunity and metabolism, and discuss some of the identified immunometabolic checkpoints. Understanding the comprehensive advances in immunometabolism helps to revise the status quo of cancer treatment. An overview of the new landscape of immunometabolism. The PI3K pathway promotes anabolism and inhibits catabolism. The LKB1 pathway inhibits anabolism and promotes catabolism. Overactivation of PI3K/AKT/mTOR pathway and IDO, IL4I1, ACAT, Sirt2, and MTHFD2 promote immunosuppression of TME formation, as evidenced by increased Treg and decreased T-cell proliferation. The LKBI-AMPK pathway promotes the differentiation of naive T cells to effector T cells and memory T cells and promotes anti-tumor immunity in DCs.
Collapse
Affiliation(s)
- Ranran Su
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Yingying Shao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Manru Huang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Donghui Liu
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Haiyang Yu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China.
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
81
|
Liu R, Cao H, Zhang S, Cai M, Zou T, Wang G, Zhang D, Wang X, Xu J, Deng S, Li T, Xu D, Gu J. ZBP1-mediated apoptosis and inflammation exacerbate steatotic liver ischemia/reperfusion injury. J Clin Invest 2024; 134:e180451. [PMID: 38743492 PMCID: PMC11213514 DOI: 10.1172/jci180451] [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: 02/20/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Steatotic donor livers are becoming more and more common in liver transplantation. However, the current use of steatotic grafts is less acceptable than normal grafts due to their higher susceptibility to ischemia/reperfusion (I/R) injury. To investigate the mechanism underlying the susceptibility of steatotic liver to I/R injury, we detected cell death markers and inflammation in clinical donor livers and animal models. We found that caspase-8-mediated hepatic apoptosis is activated in steatotic liver I/R injury. However, ablation of caspase-8 only slightly mitigated steatotic liver I/R injury without affecting inflammation. We further demonstrated that RIPK1 kinase induces both caspase-8-mediated apoptosis and cell death-independent inflammation. Inhibition of RIPK1 kinase significantly protects against steatotic liver I/R injury by alleviating both hepatic apoptosis and inflammation. Additionally, we found that RIPK1 activation is induced by Z-DNA binding protein 1 (ZBP1) but not the canonical TNF-α pathway during steatotic liver I/R injury. Deletion of ZBP1 substantially decreases the steatotic liver I/R injury. Mechanistically, ZBP1 is amplified by palmitic acid-activated JNK pathway in steatotic livers. Upon I/R injury, excessive reactive oxygen species trigger ZBP1 activation by inducing its aggregation independent of the Z-nucleic acids sensing action in steatotic livers, leading to the kinase activation of RIPK1 and the subsequent aggravation of liver injury. Thus, ZBP1-mediated RIPK1-driven apoptosis and inflammation exacerbate steatotic liver I/R injury, which could be targeted to protect steatotic donor livers during transplantation.
Collapse
Affiliation(s)
- Ran Liu
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huan Cao
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhua Zhang
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mao Cai
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tianhao Zou
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guoliang Wang
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Di Zhang
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xueling Wang
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jianjun Xu
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shenghe Deng
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tongxi Li
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Daichao Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Jinyang Gu
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| |
Collapse
|
82
|
Kim HS, Halabi EA, Enbergs N, Kohler RH, Fei F, Garris CS, Weissleder R. A non-lipid nucleic acid delivery vector with dendritic cell tropism and stimulation. Theranostics 2024; 14:2934-2945. [PMID: 38773971 PMCID: PMC11103498 DOI: 10.7150/thno.95267] [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: 02/12/2024] [Accepted: 04/10/2024] [Indexed: 05/24/2024] Open
Abstract
Rationale: Nucleic acid constructs are commonly used for vaccination, immune stimulation, and gene therapy, but their use in cancer still remains limited. One of the reasons is that systemic delivery to tumor-associated antigen-presenting cells (dendritic cells and macrophages) is often inefficient, while off-target nucleic acid-sensing immune pathways can stimulate systemic immune responses. Conversely, certain carbohydrate nanoparticles with small molecule payloads have been shown to target these cells efficiently in the tumor microenvironment. Yet, nucleic acid incorporation into such carbohydrate-based nanoparticles has proven challenging. Methods: We developed a novel approach using cross-linked bis succinyl-cyclodextrin (b-s-CD) nanoparticles to efficiently deliver nucleic acids and small-molecule immune enhancer to phagocytic cells in tumor environments and lymph nodes. Our study involved incorporating these components into the nanoparticles and assessing their efficacy in activating antigen-presenting cells. Results: The multi-modality immune stimulators effectively activated antigen-presenting cells and promoted anti-tumor immunity in vivo. This was evidenced by enhanced delivery to phagocytic cells and subsequent immune response activation in tumor environments and lymph nodes. Conclusion: Here, we describe a new approach to incorporating both nucleic acids and small-molecule immune enhancers into cross-linked bis succinyl-cyclodextrin (b-s-CD) nanoparticles for efficient delivery to phagocytic cells in tumor environments and lymph nodes in vivo. These multi-modality immune stimulators can activate antigen-presenting cells and foster anti-tumor immunity. We argue that this strategy can potentially be used to enhance anti-tumor efficacy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ralph Weissleder
- ✉ Corresponding author: R. Weissleder, MD, PhD. Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114. 617-726-8226;
| |
Collapse
|
83
|
Yang J, Liang J, Huang C, Wu Z, Lei Y. Hyperactivation of succinate dehydrogenase promotes pyroptosis of macrophage via ROS-induced GSDMD oligomerization in acute liver failure. Mol Immunol 2024; 169:86-98. [PMID: 38552285 DOI: 10.1016/j.molimm.2024.02.004] [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: 08/28/2023] [Revised: 11/28/2023] [Accepted: 02/02/2024] [Indexed: 04/13/2024]
Abstract
Acute liver failure (ALF) is a life-threatening disease with high mortality. Given excessive inflammation is one of the major pathogenesis of ALF, candidates targeting inflammation could be beneficial in the condition. Now the effect of hyperactivated succinate dehydrogenase (SDH) on promoting inflammation in lipopolysaccharide (LPS)-treated macrophages has been studied. However, its role and mechanism in ALF is not well understood. Here intraperitoneal injection of D-galactosamine and LPS was conducted in male C57BL/6 J mice to induce the ALF model. Dimethyl malonate (DMM), which inhibited SDH activity, was injected intraperitoneally 30 min before ALF induction. Macrophage pyroptosis was induced by LPS plus adenosine triphosphate (ATP). Pyroptosis-related molecules and proteins including GSDMD oligomer were examined by ELISA and western blot techniques, respectively. ROS production was assessed by fluorescence staining. The study demonstrated SDH activity was increased in liver macrophages from ALF mice. Importantly, DMM administration inhibited ROS, IL-1β, and pyroptosis-associated proteins levels (NLRP3, cleaved caspase-1, GSDMD-N, and GSDMD oligomers) both in the ALF model and in macrophages stimulated with LPS plus ATP. In vitro, ROS promoted pyroptosis by facilitating GSDMD oligomerization. Additionally, when ROS levels were increased through the addition of H2O2 to the DMM group, the levels of GSDMD oligomers were reverted. In conclusion, SDH hyperactivation promotes macrophage pyroptosis by ROS-mediated GSDMD oligomerization, suggesting that targeting this pathway holds promise as a strategy for treating ALF and other inflammatory diseases.
Collapse
Affiliation(s)
- Jiao Yang
- Department of gastroenterology, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi 545000, China
| | - JingWen Liang
- Department of gastroenterology, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi 545000, China
| | - Cai Huang
- Department of gastroenterology, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi 545000, China
| | - ZaiCheng Wu
- Department of gastroenterology, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi 545000, China
| | - YanChang Lei
- Department of gastroenterology, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi 545000, China.
| |
Collapse
|
84
|
Zhang N, Zhang J, Yang Y, Shan H, Hou S, Fang H, Ma M, Chen Z, Tan L, Xu D. A palmitoylation-depalmitoylation relay spatiotemporally controls GSDMD activation in pyroptosis. Nat Cell Biol 2024; 26:757-769. [PMID: 38538834 DOI: 10.1038/s41556-024-01397-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/05/2024] [Indexed: 05/18/2024]
Abstract
Gasdermin D (GSDMD) is the executor of pyroptosis, which is important for host defence against pathogen infection. Following activation, caspase-mediated cleavage of GSDMD releases an amino-terminal fragment (GSDMD-NT), which oligomerizes and forms pores in the plasma membrane, leading to cell death and release of proinflammatory cytokines. The spatial and temporal regulation of this process in cells remains unclear. Here we identify GSDMD as a substrate for reversible S-palmitoylation on C192 during pyroptosis. The palmitoyl acyltransferase DHHC7 palmitoylates GSDMD to direct its cleavage by caspases. Subsequently, palmitoylation of GSDMD-NT promotes its translocation to the plasma membrane, where APT2 depalmitoylates GSDMD-NT to unmask the C192 residue and promote GSDMD-NT oligomerization. Perturbation of either palmitoylation or depalmitoylation suppresses pyroptosis, leading to increased survival of mice with lipopolysaccharide-induced lethal septic shock and increased sensitivity to bacterial infection. Our findings reveal a model through which a palmitoylation-depalmitoylation relay spatiotemporally controls GSDMD activation during pyroptosis.
Collapse
Affiliation(s)
- Na Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuanxin Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hengyue Shan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Shouqiao Hou
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hongwen Fang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Min Ma
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhongwen Chen
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Li Tan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Daichao Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- Shanghai Key Laboratory of Aging Studies, Shanghai, China.
| |
Collapse
|
85
|
Kuroshima T, Kawaguchi S, Okada M. Current Perspectives of Mitochondria in Sepsis-Induced Cardiomyopathy. Int J Mol Sci 2024; 25:4710. [PMID: 38731929 PMCID: PMC11083471 DOI: 10.3390/ijms25094710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Sepsis-induced cardiomyopathy (SICM) is one of the leading indicators for poor prognosis associated with sepsis. Despite its reversibility, prognosis varies widely among patients. Mitochondria play a key role in cellular energy production by generating adenosine triphosphate (ATP), which is vital for myocardial energy metabolism. Over recent years, mounting evidence suggests that severe sepsis not only triggers mitochondrial structural abnormalities such as apoptosis, incomplete autophagy, and mitophagy in cardiomyocytes but also compromises their function, leading to ATP depletion. This metabolic disruption is recognized as a significant contributor to SICM, yet effective treatment options remain elusive. Sepsis cannot be effectively treated with inotropic drugs in failing myocardium due to excessive inflammatory factors that blunt β-adrenergic receptors. This review will share the recent knowledge on myocardial cell death in sepsis and its molecular mechanisms, focusing on the role of mitochondria as an important metabolic regulator of SICM, and discuss the potential for developing therapies for sepsis-induced myocardial injury.
Collapse
Affiliation(s)
| | | | - Motoi Okada
- Department of Emergency Medicine, Asahikawa Medical University, Asahikawa 078-8510, Japan; (T.K.); (S.K.)
| |
Collapse
|
86
|
Kappelhoff S, Margheritis EG, Cosentino K. New insights into Gasdermin D pore formation. Biochem Soc Trans 2024; 52:681-692. [PMID: 38497302 DOI: 10.1042/bst20230549] [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: 12/20/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
Gasdermin D (GSDMD) is a pore-forming protein that perforates the plasma membrane (PM) during pyroptosis, a pro-inflammatory form of cell death, to induce the unconventional secretion of inflammatory cytokines and, ultimately, cell lysis. GSDMD is activated by protease-mediated cleavage of its active N-terminal domain from the autoinhibitory C-terminal domain. Inflammatory caspase-1, -4/5 are the main activators of GSDMD via either the canonical or non-canonical pathways of inflammasome activation, but under certain stimuli, caspase-8 and other proteases can also activate GSDMD. Activated GSDMD can oligomerize and assemble into various nanostructures of different sizes and shapes that perforate cellular membranes, suggesting plasticity in pore formation. Although the exact mechanism of pore formation has not yet been deciphered, cysteine residues are emerging as crucial modulators of the oligomerization process. GSDMD pores and thus the outcome of pyroptosis can be modulated by various regulatory mechanisms. These include availability of activated GSDMD at the PM, control of the number of GSDMD pores by PM repair mechanisms, modulation of the lipid environment and post-translational modifications. Here, we review the latest findings on the mechanisms that induce GSDMD to form membrane pores and how they can be tightly regulated for cell content release and cell fate modulation.
Collapse
Affiliation(s)
- Shirin Kappelhoff
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Eleonora G Margheritis
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Katia Cosentino
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
87
|
Wright SS, Wang C, Ta A, Havira MS, Ruan J, Rathinam VA, Vanaja SK. A bacterial toxin co-opts caspase-3 to disable active gasdermin D and limit macrophage pyroptosis. Cell Rep 2024; 43:114004. [PMID: 38522070 PMCID: PMC11095105 DOI: 10.1016/j.celrep.2024.114004] [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: 11/07/2023] [Revised: 02/15/2024] [Accepted: 03/11/2024] [Indexed: 03/26/2024] Open
Abstract
During infections, host cells are exposed to pathogen-associated molecular patterns (PAMPs) and virulence factors that stimulate multiple signaling pathways that interact additively, synergistically, or antagonistically. The net effect of such higher-order interactions is a vital determinant of the outcome of host-pathogen interactions. Here, we demonstrate one such complex interplay between bacterial exotoxin- and PAMP-induced innate immune pathways. We show that two caspases activated during enterohemorrhagic Escherichia coli (EHEC) infection by lipopolysaccharide (LPS) and Shiga toxin (Stx) interact in a functionally antagonistic manner; cytosolic LPS-activated caspase-11 cleaves full-length gasdermin D (GSDMD), generating an active pore-forming N-terminal fragment (NT-GSDMD); subsequently, caspase-3 activated by EHEC Stx cleaves the caspase-11-generated NT-GSDMD to render it nonfunctional, thereby inhibiting pyroptosis and interleukin-1β maturation. Bacteria typically subvert inflammasomes by targeting upstream components such as NLR sensors or full-length GSDMD but not active NT-GSDMD. Thus, our findings uncover a distinct immune evasion strategy where a bacterial toxin disables active NT-GSDMD by co-opting caspase-3.
Collapse
Affiliation(s)
- Skylar S Wright
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Chengliang Wang
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Atri Ta
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | | | - Jianbin Ruan
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Vijay A Rathinam
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Sivapriya Kailasan Vanaja
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Avenue, Farmington, CT 06030, USA.
| |
Collapse
|
88
|
Huang S, Shang M, Guo L, Sun X, Xiao S, Shi D, Meng D, Zhao Y, Wang X, Liu R, Li J. Hydralazine loaded nanodroplets combined with ultrasound-targeted microbubble destruction to induce pyroptosis for tumor treatment. J Nanobiotechnology 2024; 22:193. [PMID: 38643134 PMCID: PMC11031971 DOI: 10.1186/s12951-024-02453-0] [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: 01/26/2024] [Accepted: 04/01/2024] [Indexed: 04/22/2024] Open
Abstract
Pyroptosis, a novel type of programmed cell death (PCD), which provides a feasible therapeutic option for the treatment of tumors. However, due to the hypermethylation of the promoter, the critical protein Gasdermin E (GSDME) is lacking in the majority of cancer cells, which cannot start the pyroptosis process and leads to dissatisfactory therapeutic effects. Additionally, the quick clearance, systemic side effects, and low concentration at the tumor site of conventional pyroptosis reagents restrict their use in clinical cancer therapy. Here, we described a combination therapy that induces tumor cell pyroptosis via the use of ultrasound-targeted microbubble destruction (UTMD) in combination with DNA demethylation. The combined application of UTMD and hydralazine-loaded nanodroplets (HYD-NDs) can lead to the rapid release of HYD (a demethylation drug), which can cause the up-regulation of GSDME expression, and produce reactive oxygen species (ROS) by UTMD to cleave up-regulated GSDME, thereby inducing pyroptosis. HYD-NDs combined with ultrasound (US) group had the strongest tumor inhibition effect, and the tumor inhibition rate was 87.15% (HYD-NDs group: 51.41 ± 3.61%, NDs + US group: 32.73%±7.72%), indicating that the strategy had a more significant synergistic anti-tumor effect. In addition, as a new drug delivery carrier, HYD-NDs have great biosafety, tumor targeting, and ultrasound imaging performance. According to the results, the combined therapy reasonably regulated the process of tumor cell pyroptosis, which offered a new strategy for optimizing the therapy of GSDME-silenced solid tumors.
Collapse
Affiliation(s)
- Shuting Huang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xiao Sun
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Shan Xiao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Dong Meng
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Yading Zhao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xiaoxuan Wang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Rui Liu
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
- Department of Ultrasound, Qilu Hospital (Qingdao) of Shandong University, Qingdao, Shandong, 266035, China.
| |
Collapse
|
89
|
La Rosa F, Varotto-Boccazzi I, Saresella M, Marventano I, Cattaneo GM, Hernis A, Piancone F, Otranto D, Epis S, Bandi C, Clerici M. The non-pathogenic protozoon Leishmania tarentolae interferes with the activation of NLRP3 inflammasome in human cells: new perspectives in the control of inflammation. Front Immunol 2024; 15:1298275. [PMID: 38707903 PMCID: PMC11066211 DOI: 10.3389/fimmu.2024.1298275] [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: 09/21/2023] [Accepted: 04/03/2024] [Indexed: 05/07/2024] Open
Abstract
Background Innate immune responses against infectious agents can act as triggers of inflammatory diseases. On the other hand, various pathogens have developed mechanisms for the evasion of the immune response, based on an inhibition of innate immunity and inflammatory responses. Inflammatory diseases could thus be controlled through the administration of pathogens or pathogen-derived molecules, capable of interfering with the mechanisms at the basis of inflammation. In this framework, the NLRP3 inflammasome is an important component in innate antimicrobial responses and a major player in the inflammatory disease. Parasites of the genus Leishmania are master manipulators of innate immune mechanisms, and different species have been shown to inhibit inflammasome formation. However, the exploitation of pathogenic Leishmania species as blockers of NLRP3-based inflammatory diseases poses safety concerns. Methods To circumvent safety issues associated with pathogenic parasites, we focused on Leishmania tarentolae, a species of Leishmania that is not infectious to humans. Because NLRP3 typically develops in macrophages, in response to the detection and engulfment microorganisms, we performed our experiments on a monocyte-macrophage cell line (THP-1), either wild type or knockout for ASC, a key component of NLRP3 formation, with determination of cytokines and other markers of inflammation. Results L. tarentolae was shown to possess the capability of dampening the formation of NLRP3 inflammasome and the consequent expression of pro-inflammatory molecules, with minor differences compared to effects of pathogenic Leishmania species. Conclusion The non-pathogenic L. tarentolae appears a promising pro-biotic microbe with anti-inflammatory properties or a source of immune modulating cellular fractions or molecules, capable of interfering with the formation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
| | - Ilaria Varotto-Boccazzi
- Department of Biosciences, University of Milan, Milan, Italy
- Pediatric Clinical Research Center 'Romeo ed Enrica Invernizzi', University of Milan, Milan, Italy
| | | | | | | | - Ambra Hernis
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | | | - Domenico Otranto
- Department of Veterinary Medicine, University of Bari, Valenzano, Italy
- Faculty of Veterinary Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Sara Epis
- Department of Biosciences, University of Milan, Milan, Italy
- Pediatric Clinical Research Center 'Romeo ed Enrica Invernizzi', University of Milan, Milan, Italy
| | - Claudio Bandi
- Department of Biosciences, University of Milan, Milan, Italy
- Pediatric Clinical Research Center 'Romeo ed Enrica Invernizzi', University of Milan, Milan, Italy
| | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
90
|
Yang Y, More S, De Smet F, De Vleeschouwer S, Agostinis P. Antioxidant network-based signatures cluster glioblastoma into distinct redox-resistant phenotypes. Front Immunol 2024; 15:1342977. [PMID: 38698847 PMCID: PMC11063242 DOI: 10.3389/fimmu.2024.1342977] [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: 11/22/2023] [Accepted: 04/03/2024] [Indexed: 05/05/2024] Open
Abstract
Introduction Aberrant reactive oxygen species (ROS) production is one of the hallmarks of cancer. During their growth and dissemination, cancer cells control redox signaling to support protumorigenic pathways. As a consequence, cancer cells become reliant on major antioxidant systems to maintain a balanced redox tone, while avoiding excessive oxidative stress and cell death. This concept appears especially relevant in the context of glioblastoma multiforme (GBM), the most aggressive form of brain tumor characterized by significant heterogeneity, which contributes to treatment resistance and tumor recurrence. From this viewpoint, this study aims to investigate whether gene regulatory networks can effectively capture the diverse redox states associated with the primary phenotypes of GBM. Methods In this study, we utilized publicly available GBM datasets along with proprietary bulk sequencing data. Employing computational analysis and bioinformatics tools, we stratified GBM based on their antioxidant capacities and evaluated the distinctive functionalities and prognostic values of distinct transcriptional networks in silico. Results We established three distinct transcriptional co-expression networks and signatures (termed clusters C1, C2, and C3) with distinct antioxidant potential in GBM cancer cells. Functional analysis of each cluster revealed that C1 exhibits strong antioxidant properties, C2 is marked with a discrepant inflammatory trait and C3 was identified as the cluster with the weakest antioxidant capacity. Intriguingly, C2 exhibited a strong correlation with the highly aggressive mesenchymal subtype of GBM. Furthermore, this cluster holds substantial prognostic importance: patients with higher gene set variation analysis (GSVA) scores of the C2 signature exhibited adverse outcomes in overall and progression-free survival. Conclusion In summary, we provide a set of transcriptional signatures that unveil the antioxidant potential of GBM, offering a promising prognostic application and a guide for therapeutic strategies in GBM therapy.
Collapse
Affiliation(s)
- Yihan Yang
- Research Group Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Laboratory of Cell Death Research & Therapy, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Vlaams Instituut voor Biotechnologie (VIB) Center for Cancer Biology Research, Leuven, Belgium
| | - Sanket More
- Laboratory of Cell Death Research & Therapy, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Vlaams Instituut voor Biotechnologie (VIB) Center for Cancer Biology Research, Leuven, Belgium
| | - Frederik De Smet
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Leuven Institute for Single-Cell Omics (LISCO), Leuven, Belgium
| | - Steven De Vleeschouwer
- Research Group Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, University Hospitals Leuven, Leuven, Belgium
- Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Laboratory of Cell Death Research & Therapy, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Vlaams Instituut voor Biotechnologie (VIB) Center for Cancer Biology Research, Leuven, Belgium
| |
Collapse
|
91
|
Song N, Mei S, Wang X, Hu G, Lu M. Focusing on mitochondria in the brain: from biology to therapeutics. Transl Neurodegener 2024; 13:23. [PMID: 38632601 PMCID: PMC11022390 DOI: 10.1186/s40035-024-00409-w] [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: 12/10/2023] [Accepted: 03/13/2024] [Indexed: 04/19/2024] Open
Abstract
Mitochondria have multiple functions such as supplying energy, regulating the redox status, and producing proteins encoded by an independent genome. They are closely related to the physiology and pathology of many organs and tissues, among which the brain is particularly prominent. The brain demands 20% of the resting metabolic rate and holds highly active mitochondrial activities. Considerable research shows that mitochondria are closely related to brain function, while mitochondrial defects induce or exacerbate pathology in the brain. In this review, we provide comprehensive research advances of mitochondrial biology involved in brain functions, as well as the mitochondria-dependent cellular events in brain physiology and pathology. Furthermore, various perspectives are explored to better identify the mitochondrial roles in neurological diseases and the neurophenotypes of mitochondrial diseases. Finally, mitochondrial therapies are discussed. Mitochondrial-targeting therapeutics are showing great potentials in the treatment of brain diseases.
Collapse
Affiliation(s)
- Nanshan Song
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shuyuan Mei
- The First Clinical Medical College, Nanjing Medical University, Nanjing, 211166, China
| | - Xiangxu Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Gang Hu
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
- Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China.
| |
Collapse
|
92
|
Balasubramanian A, Hsu AY, Ghimire L, Tahir M, Devant P, Fontana P, Du G, Liu X, Fabin D, Kambara H, Xie X, Liu F, Hasegawa T, Xu R, Yu H, Wei W, Chen M, Kolakowski S, Trauger S, Larsen MR, Wei W, Wu H, Kagan JC, Lieberman J, Luo HR. The palmitoylation of gasdermin D directs its membrane translocation and pore formation during pyroptosis. Sci Immunol 2024; 9:eadn1452. [PMID: 38530158 PMCID: PMC11367861 DOI: 10.1126/sciimmunol.adn1452] [Citation(s) in RCA: 59] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Abstract
Plasma membrane perforation elicited by caspase cleavage of the gasdermin D (GSDMD) N-terminal domain (GSDMD-NT) triggers pyroptosis. The mechanisms underlying GSDMD membrane translocation and pore formation are not fully understood. Here, using a proteomic approach, we identified fatty acid synthase (FASN) as a GSDMD-binding partner. S-palmitoylation of GSDMD at Cys191/Cys192 (human/mouse), catalyzed by palmitoyl acyltransferases ZDHHC5 and ZDHHC9 and facilitated by reactive oxygen species (ROS), directly mediated membrane translocation of GSDMD-NT but not full-length GSDMD (GSDMD-FL). Palmitoylation of GSDMD-FL could be induced before inflammasome activation by stimuli such as lipopolysaccharide (LPS), consequently serving as an essential molecular event in macrophage priming. Inhibition of GSDMD palmitoylation suppressed macrophage pyroptosis and IL-1β release, mitigated organ damage, and enhanced the survival of septic mice. Thus, GSDMD-NT palmitoylation is a key regulatory mechanism controlling GSDMD membrane localization and activation, which may offer an additional target for modulating immune activity in infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Arumugam Balasubramanian
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children’s Hospital; Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Alan Y. Hsu
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children’s Hospital; Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Laxman Ghimire
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children’s Hospital; Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Muhammad Tahir
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children’s Hospital; Enders Research Building, Room 811, Boston, MA, 02115, USA
- Biomedical Mass Spectrometry and Systems Biology, University of Southern Denmark; Odense, DK
| | - Pascal Devant
- Division of Gastroenterology, Boston Children’s Hospital and Harvard Medical School; 300 Longwood Avenue, Boston, MA 02115, USA
| | - Pietro Fontana
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School; Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Gang Du
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School; Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Xing Liu
- Department of Pediatrics, Harvard Medical School; Program in Cellular and Molecular Medicine; Boston Children’s Hospital, Boston, MA 02115, USA
| | - Dang Fabin
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Boston, MA, USA
| | - Hiroto Kambara
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children’s Hospital; Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Xuemei Xie
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children’s Hospital; Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Fei Liu
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children’s Hospital; Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Tomoya Hasegawa
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children’s Hospital; Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Rong Xu
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children’s Hospital; Enders Research Building, Room 811, Boston, MA, 02115, USA
| | - Hongbo Yu
- VA Boston Healthcare System, Department of Pathology and Laboratory Medicine; 1400 VFW Parkway, West Roxbury, MA 02132 USA
| | - Wenyi Wei
- Department of Pediatrics, Harvard Medical School; Program in Cellular and Molecular Medicine; Boston Children’s Hospital, Boston, MA 02115, USA
| | - Mei Chen
- Harvard Center for Mass Spectrometry, Harvard University; Boston, MA 02115, USA
| | - Steven Kolakowski
- Harvard Center for Mass Spectrometry, Harvard University; Boston, MA 02115, USA
| | - Sunia Trauger
- Harvard Center for Mass Spectrometry, Harvard University; Boston, MA 02115, USA
| | - Martin Røssel Larsen
- Biomedical Mass Spectrometry and Systems Biology, University of Southern Denmark; Odense, DK
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Boston, MA, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School; Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Jonathan C. Kagan
- Division of Gastroenterology, Boston Children’s Hospital and Harvard Medical School; 300 Longwood Avenue, Boston, MA 02115, USA
| | - Judy Lieberman
- Department of Pediatrics, Harvard Medical School; Program in Cellular and Molecular Medicine; Boston Children’s Hospital, Boston, MA 02115, USA
| | - Hongbo R. Luo
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School; Department of Laboratory Medicine, Boston Children’s Hospital; Enders Research Building, Room 811, Boston, MA, 02115, USA
| |
Collapse
|
93
|
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
|
94
|
Zhu D, Liang H, Du Z, Liu Q, Li G, Zhang W, Wu D, Zhou X, Song Y, Yang C. Altered Metabolism and Inflammation Driven by Post-translational Modifications in Intervertebral Disc Degeneration. RESEARCH (WASHINGTON, D.C.) 2024; 7:0350. [PMID: 38585329 PMCID: PMC10997488 DOI: 10.34133/research.0350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2024]
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent cause of low back pain and a leading contributor to disability. IVDD progression involves pathological shifts marked by low-grade inflammation, extracellular matrix remodeling, and metabolic disruptions characterized by heightened glycolytic pathways, mitochondrial dysfunction, and cellular senescence. Extensive posttranslational modifications of proteins within nucleus pulposus cells and chondrocytes play crucial roles in reshaping the intervertebral disc phenotype and orchestrating metabolism and inflammation in diverse contexts. This review focuses on the pivotal roles of phosphorylation, ubiquitination, acetylation, glycosylation, methylation, and lactylation in IVDD pathogenesis. It integrates the latest insights into various posttranslational modification-mediated metabolic and inflammatory signaling networks, laying the groundwork for targeted proteomics and metabolomics for IVDD treatment. The discussion also highlights unexplored territories, emphasizing the need for future research, particularly in understanding the role of lactylation in intervertebral disc health, an area currently shrouded in mystery.
Collapse
Affiliation(s)
- Dingchao Zhu
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Huaizhen Liang
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Zhi Du
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Qian Liu
- College of Life Sciences,
Wuhan University, Wuhan 430072, Hubei Province, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Weifeng Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Di Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Xingyu Zhou
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Yu Song
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
95
|
Enbergs N, Halabi EA, Goubet A, Schleyer K, Fredrich IR, Kohler RH, Garris CS, Pittet MJ, Weissleder R. Pharmacological Polarization of Tumor-Associated Macrophages Toward a CXCL9 Antitumor Phenotype. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309026. [PMID: 38342608 PMCID: PMC11022742 DOI: 10.1002/advs.202309026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/16/2024] [Indexed: 02/13/2024]
Abstract
Tumor-associated macrophages (TAM) are a diverse population of myeloid cells that are often abundant and immunosuppressive in human cancers. CXCL9Hi TAM has recently been described to have an antitumor phenotype and is linked to immune checkpoint response. Despite the emerging understanding of the unique antitumor TAM phenotype, there is a lack of TAM-specific therapeutics to exploit this new biological understanding. Here, the discovery and characterization of multiple small-molecule enhancers of chemokine ligand 9 (CXCL9) and their targeted delivery in a TAM-avid systemic nanoformulation is reported. With this strategy, it is efficient encapsulation and release of multiple drug loads that can efficiently induce CXCL9 expression in macrophages, both in vitro and in vivo in a mouse tumor model. These observations provide a window into the molecular features that define TAM-specific states, an insight a novel therapeutic anticancer approach is used to discover.
Collapse
Affiliation(s)
- Noah Enbergs
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Elias A. Halabi
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Anne‐Gaëlle Goubet
- Department of Pathology and ImmunologyUniversity of GenevaGeneva1211Switzerland
- AGORA Cancer CenterSwiss Cancer Center LemanLausanne1011Switzerland
| | - Kelton Schleyer
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Ina R. Fredrich
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Rainer H. Kohler
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Christopher S. Garris
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Mikaël J. Pittet
- Department of Pathology and ImmunologyUniversity of GenevaGeneva1211Switzerland
- AGORA Cancer CenterSwiss Cancer Center LemanLausanne1011Switzerland
- Ludwig Institute for Cancer ResearchLausanne1005Switzerland
| | - Ralph Weissleder
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
- Department of Systems BiologyHarvard Medical School200 Longwood AveBostonMA02115USA
| |
Collapse
|
96
|
Liu J, Kang R, Tang D. Lipopolysaccharide delivery systems in innate immunity. Trends Immunol 2024; 45:274-287. [PMID: 38494365 DOI: 10.1016/j.it.2024.02.003] [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: 02/16/2024] [Revised: 02/22/2024] [Accepted: 02/24/2024] [Indexed: 03/19/2024]
Abstract
Lipopolysaccharide (LPS), a key component of the outer membrane in Gram-negative bacteria (GNB), is widely recognized for its crucial role in mammalian innate immunity and its link to mortality in intensive care units. While its recognition via the Toll-like receptor (TLR)-4 receptor on cell membranes is well established, the activation of the cytosolic receptor caspase-11 by LPS is now known to lead to inflammasome activation and subsequent induction of pyroptosis. Nevertheless, a fundamental question persists regarding the mechanism by which LPS enters host cells. Recent investigations have identified at least four primary pathways that can facilitate this process: bacterial outer membrane vesicles (OMVs); the spike (S) protein of SARS-CoV-2; host-secreted proteins; and host extracellular vesicles (EVs). These delivery systems provide new avenues for therapeutic interventions against sepsis and infectious diseases.
Collapse
Affiliation(s)
- Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
97
|
Bhat AA, Riadi Y, Afzal M, Bansal P, Kaur H, Deorari M, Ali H, Shahwan M, Almalki WH, Kazmi I, Alzarea SI, Dureja H, Singh SK, Dua K, Gupta G. Exploring ncRNA-mediated pathways in sepsis-induced pyroptosis. Pathol Res Pract 2024; 256:155224. [PMID: 38452584 DOI: 10.1016/j.prp.2024.155224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/08/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024]
Abstract
Sepsis, a potentially fatal illness caused by an improper host response to infection, remains a serious problem in the world of healthcare. In recent years, the role of ncRNA has emerged as a pivotal aspect in the intricate landscape of cellular regulation. The exploration of ncRNA-mediated regulatory networks reveals their profound influence on key molecular pathways orchestrating pyroptotic responses during septic conditions. Through a comprehensive analysis of current literature, we navigate the diverse classes of ncRNAs, including miRNAs, lncRNAs, and circRNAs, elucidating their roles as both facilitators and inhibitors in the modulation of pyroptotic processes. Furthermore, we highlight the potential diagnostic and therapeutic implications of targeting these ncRNAs in the context of sepsis, aiming to cover the method for novel and effective strategies to mitigate the devastating consequences of septic pathogenesis. As we unravel the complexities of this regulatory axis, a deeper understanding of the intricate crosstalk between ncRNAs and pyroptosis emerges, offering promising avenues for advancing our approach to sepsis intervention. The intricate pathophysiology of sepsis is examined in this review, which explores the dynamic interaction between ncRNAs and pyroptosis, a highly regulated kind of programmed cell death.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Yassine Riadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh 247341, India; Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand 831001, India
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Moyad Shahwan
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 3467, United Arab Emirates; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman 3469, United Arab Emirates
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341, Saudi Arabia
| | - Hairsh Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo-NSW 2007, Australia
| | - Gaurav Gupta
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman 3469, United Arab Emirates; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India.
| |
Collapse
|
98
|
Wallace HL, Russell RS. Inflammatory Consequences: Hepatitis C Virus-Induced Inflammasome Activation and Pyroptosis. Viral Immunol 2024; 37:126-138. [PMID: 38593460 DOI: 10.1089/vim.2023.0138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024] Open
Abstract
Hepatitis C virus (HCV), despite the availability of effective direct-acting antivirals (DAAs) that clear the virus from >95% of individuals treated, continues to cause significant health care burden due to disease progression that can lead to fibrosis, cirrhosis, and/or hepatocellular carcinoma. The fact that some people who are treated with DAAs still go on to develop worsening liver disease warrants further study into the immunopathogenesis of HCV. Many viral infections, including HCV, have been associated with activation of the inflammasome/pyroptosis pathway. This inflammatory cell death pathway ultimately results in cell lysis and release of inflammatory cytokines, IL-18 and IL-1β. This review will report on studies that investigated HCV and inflammasome activation/pyroptosis. This includes clinical in vivo data showing elevated pyroptosis-associated cytokines in the blood of individuals living with HCV, studies of genetic associations of pyroptosis-related genes and development of liver disease, and in vitro studies aimed at understanding the mechanism of pyroptosis induced by HCV. Finally, we discuss major gaps in understanding and outstanding questions that remain in the field of HCV-induced pyroptosis.
Collapse
Affiliation(s)
- Hannah L Wallace
- Immunology and Infectious Diseases Group, Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St John's, Canada
| | - Rodney S Russell
- Immunology and Infectious Diseases Group, Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St John's, Canada
| |
Collapse
|
99
|
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
|
100
|
Win S, Than TA, Kaplowitz N. Mitochondrial P-JNK target, SAB (SH3BP5), in regulation of cell death. Front Cell Dev Biol 2024; 12:1359152. [PMID: 38559813 PMCID: PMC10978662 DOI: 10.3389/fcell.2024.1359152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
Cell death occurs in various circumstances, such as homeostasis, stress response, and defense, via specific pathways and mechanisms that are regulated by specific activator-induced signal transductions. Among them, Jun N-terminal kinases (JNKs) participate in various aspects, and the recent discovery of JNKs and mitochondrial protein SAB interaction in signal regulation of cell death completes our understanding of the mechanism of sustained activation of JNK (P-JNK), which leads to triggering of the machinery of cell death. This understanding will lead the investigators to discover the modulators facilitating or preventing cell death for therapeutic application in acute or chronic diseases and cancer. We discuss here the mechanism and modulators of the JNK-SAB-ROS activation loop, which is the core component of mitochondria-dependent cell death, specifically apoptosis and mitochondrial permeability transition (MPT)-driven necrosis, and which may also contribute to cell death mechanisms of ferroptosis and pyroptosis. The discussion here is based on the results and evidence discovered from liver disease models, but the JNK-SAB-ROS activation loop to sustain JNK activation is universally applicable to various disease models where mitochondria and reactive oxygen species contribute to the mechanism of disease.
Collapse
Affiliation(s)
- Sanda Win
- *Correspondence: Sanda Win, ; Neil Kaplowitz,
| | | | - Neil Kaplowitz
- Department of Medicine, Division of Gastroenterology and Liver Diseases, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|