1
|
Zhuang H, Lei W, Wu Q, Zhao S, Zhao Y, Zhang S, Zhao N, Sun J, Liu Y. Overexpressed CD73 attenuates GSDMD-mediated astrocyte pyroptosis induced by cerebral ischemia-reperfusion injury through the A2B/NF-κB pathway. Exp Neurol 2025; 386:115152. [PMID: 39832662 DOI: 10.1016/j.expneurol.2025.115152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Ischemic stroke, resulting from the blockage or narrowing of cerebral vessels, causes brain tissue damage due to ischemia and hypoxia. Although reperfusion therapy is essential to restore blood flow, it may also result in reperfusion injury, causing secondary damage through mechanisms like oxidative stress, inflammation, and excitotoxicity. These effects significantly impact astrocytes, neurons, and endothelial cells, aggravating brain injury and disrupting the blood-brain barrier. CD73, an ectoenzyme that regulates adenosine production through ATP hydrolysis, plays a critical role in purinergic signaling and neuroprotection. During ischemic stroke, CD73 expression is dynamically regulated in response to ischemia and inflammation. It catalyzes the conversion of AMP to adenosine, which activates adenosine receptors to exert neuroprotective effects. Targeting the CD73-adenosine pathway presents a potential therapeutic strategy for mitigating ischemic stroke damage. Pyroptosis, a highly inflammatory form of programmed cell death mediated by inflammasomes like NLRP3 and caspases, plays a significant role in cerebral ischemia-reperfusion injury. Astrocytes, the most abundant CNS cells, contribute to both neuroprotection and injury, with pyroptosis exacerbating inflammation and brain damage. Regulating astrocyte pyroptosis is a promising therapeutic target. Our study investigates CD73's role in regulating astrocyte pyroptosis during ischemia-reperfusion injury. Using CD73 knockout mice and overexpression models, along with in vitro oxygen-glucose deprivation/reperfusion experiments, we found that CD73 overexpression reduces GSDMD-mediated astrocyte pyroptosis via the A2B/NF-κB pathway. These findings offer a novel approach to reducing neuroinflammation, protecting astrocytes, and improving outcomes in ischemic stroke.
Collapse
Affiliation(s)
- Hao Zhuang
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214000, China; Wuxi Medical Center of Nanjing Medical University, Wuxi 214000, China
| | - Wen Lei
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214000, China; Wuxi Medical Center of Nanjing Medical University, Wuxi 214000, China
| | - Qiang Wu
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214000, China; Wuxi Medical Center of Nanjing Medical University, Wuxi 214000, China
| | - Songyun Zhao
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214000, China; Wuxi Medical Center of Nanjing Medical University, Wuxi 214000, China
| | - Yunxuan Zhao
- Department of Endocrinology, Nanjing Hospital of Traditional Chinese Medicine, Nanjing 210001, China
| | - Shizhe Zhang
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Bengbu Medical University, Hefei, Anhui 230001, China
| | - Ning Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Jun Sun
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214000, China; Wuxi Medical Center of Nanjing Medical University, Wuxi 214000, China.
| | - Yuankun Liu
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214000, China; Wuxi Medical Center of Nanjing Medical University, Wuxi 214000, China.
| |
Collapse
|
2
|
Xu X, Huang Z, Huang Z, Lv X, Jiang D, Huang Z, Han B, Lin G, Liu G, Li S, Fan J, Lv X. Butyrate attenuates intestinal inflammation in Crohn's disease by suppressing pyroptosis of intestinal epithelial cells via the cGSA-STING-NLRP3 axis. Int Immunopharmacol 2024; 143:113305. [PMID: 39426229 DOI: 10.1016/j.intimp.2024.113305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/21/2024] [Accepted: 09/29/2024] [Indexed: 10/21/2024]
Abstract
Butyrate can strengthen the intestinal epithelial barrier. However, the mechanisms by which butyrate affects intestinal epithelial cells (IECs) pyroptosis in Crohn's disease (CD) remain unclear. In this study, we collected colonic biopsy samples from CD patients and healthy controls to assess pyroptosis levels. Our findings indicated elevated expression of pyroptosis markers in CD patients, alongside distinct morphological evidence of pyroptosis in IECs. We further investigated the effects of tributyrin on pyroptosis and the cGAS-STING pathway in a trinitrobenzene sulfonic acid-induced colitis rat model. Tributyrin significantly mitigated intestinal inflammation, reduced pathological progression, and inhibited pyroptosis and cGAS-STING pathway activation in the colitis rat model. Similarly, in an in vitro model of IECs pyroptosis, sodium butyrate inhibited pyroptosis and cGAS-STING pathway activation in HT-29 cells. Co-treatment with a cGAS-STING pathway activator and butyrate demonstrated that the activator reversed the inhibitory effects of butyrate on pyroptosis and cGAS-STING pathway activation in both the colitis rat model and HT-29 cells. Mechanistically, the cGAS-STING pathway was found to interact with NLRP3. Taken together, butyrate may mitigate intestinal inflammation in CD by suppressing cGAS-STING-NLRP3 axis-mediated IECs pyroptosis. These findings offer new insights into potential therapeutic strategies for managing CD.
Collapse
Affiliation(s)
- Xiaofang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhou Huang
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhixi Huang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaodan Lv
- Department of Clinical Experimental Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Dan Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ziqian Huang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Bing Han
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guangfu Lin
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Gengfeng Liu
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shiquan Li
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Junhua Fan
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaoping Lv
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
3
|
Schiffelers LDJ, Tesfamariam YM, Jenster LM, Diehl S, Binder SC, Normann S, Mayr J, Pritzl S, Hagelauer E, Kopp A, Alon A, Geyer M, Ploegh HL, Schmidt FI. Antagonistic nanobodies implicate mechanism of GSDMD pore formation and potential therapeutic application. Nat Commun 2024; 15:8266. [PMID: 39327452 PMCID: PMC11427689 DOI: 10.1038/s41467-024-52110-1] [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: 03/12/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024] Open
Abstract
Inflammasome activation results in the cleavage of gasdermin D (GSDMD) by pro-inflammatory caspases. The N-terminal domains (GSDMDNT) oligomerize and assemble pores penetrating the target membrane. As methods to study pore formation in living cells are insufficient, the order of conformational changes, oligomerization, and membrane insertion remained unclear. We have raised nanobodies (VHHs) against human GSDMD and find that cytosolic expression of VHHGSDMD-1 and VHHGSDMD-2 prevents oligomerization of GSDMDNT and pyroptosis. The nanobody-stabilized GSDMDNT monomers partition into the plasma membrane, suggesting that membrane insertion precedes oligomerization. Inhibition of GSDMD pore formation switches cell death from pyroptosis to apoptosis, likely driven by the enhanced caspase-1 activity required to activate caspase-3. Recombinant antagonistic nanobodies added to the extracellular space prevent pyroptosis and exhibit unexpected therapeutic potential. They may thus be suitable to treat the ever-growing list of diseases caused by activation of (non-) canonical inflammasomes.
Collapse
Affiliation(s)
- Lisa D J Schiffelers
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Yonas M Tesfamariam
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lea-Marie Jenster
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Stefan Diehl
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Sophie C Binder
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Sabine Normann
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Jonathan Mayr
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Steffen Pritzl
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Elena Hagelauer
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Anja Kopp
- Institute of Structural Biology, Medical Faculty, University of Bonn, Bonn, Germany
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Assaf Alon
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Matthias Geyer
- Institute of Structural Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Hidde L Ploegh
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Florian I Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany.
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Core Facility Nanobodies, Medical Faculty, University of Bonn, Bonn, Germany.
| |
Collapse
|
4
|
Yaglikara E, Boluk O, Bayindir Y, Bilginer Y, Tasar MA, Ozen S, Sag E. The Potential Role of Cell-Death Mechanisms in the Pathogenesis of Familial Mediterranean Fever Attacks: Granzyme A and Beyond. Diagnostics (Basel) 2024; 14:2031. [PMID: 39335710 PMCID: PMC11431255 DOI: 10.3390/diagnostics14182031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/02/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND FMF is the most common autoinflammatory disease. The activation of the pyrin inflammasome is the mainstay of the pathogenesis, which might lead to a specific cell-death mechanism, pyroptosis. Pyroptosis is a programmed inflammatory cell death mediated by gasdermin proteins, featuring cell swelling, membrane rupture, and release of inflammatory contents Aim: In this study we aimed to analyze the cell-death mechanisms in the pathogenesis of FMF attacks. METHODS Twenty-five FMF patients were included, and PFAPA patients (n = 10) and healthy controls (HC, n = 10) served as controls. We collected plasma samples from FMF and PFAPA patients during the attack and the attack-free period. We measured the soluble plasma levels of sFas, sFasL, granzyme A, granzyme B, perforin, granulysin, IL-2, IL-4, IL-10, IL-6, IL-17A, TNF-α, and IFN-γ by commercial pre-defined cytometric bead array kits. RESULTS There was no significant difference between groups in terms of sex and age between FMF patients and HCs, but PFAPA patients were younger than other groups due to the nature of the disease. We then analyzed the components of apoptosis and pyroptosis. The levels of sFasL (p = 0.035) and granzyme A (p = 0.038) in FMF patients were significantly increased during the attack period and decreased to levels comparable to HCs during the attack-free period. This increase was not seen in the PFAPA patients, with comparable levels with the HC group both during attack period and attack-free period. During the attack period of FMF patients, granzyme B (p = 0.145) and perforin (p = 0.203) levels were also increased; however, the differences were not statistically significant. The levels of sFasL, granzyme A, granzyme B, and perforin were closely correlated with each other during the attack period of FMF patients. CONCLUSIONS Our study on death pathways during an FMF attack, suggests an upregulation in both pyroptosis through the granzyme-gasdermin pathway and apoptosis with the increased FasL and perforin levels, which was different from PFAPA patients. These findings might shed light on the reason for the nature of self-limited attacks, but further studies are needed to prove this hypothesis.
Collapse
Affiliation(s)
- Ece Yaglikara
- Department of Pediatrics, Ankara Training and Research Hospital, 06230 Ankara, Turkey
| | - Oguz Boluk
- Department of Pediatrics, Ankara Training and Research Hospital, 06230 Ankara, Turkey
| | - Yagmur Bayindir
- Department of Pediatric Rheumatology, Hacettepe University, 06230 Ankara, Turkey
| | - Yelda Bilginer
- Department of Pediatric Rheumatology, Hacettepe University, 06230 Ankara, Turkey
| | - Medine Aysin Tasar
- Department of Pediatrics, Ankara Training and Research Hospital, 06230 Ankara, Turkey
| | - Seza Ozen
- Department of Pediatric Rheumatology, Hacettepe University, 06230 Ankara, Turkey
- Pediatric Rheumatology Unit, Translational Medicine Laboratories, Hacettepe University, 06230 Ankara, Turkey
| | - Erdal Sag
- Department of Pediatric Rheumatology, Hacettepe University, 06230 Ankara, Turkey
- Pediatric Rheumatology Unit, Translational Medicine Laboratories, Hacettepe University, 06230 Ankara, Turkey
| |
Collapse
|
5
|
Pan Z, Xu K, Huang G, Hu H, Yang H, Shen H, Qiu K, Wang C, Xu T, Yu X, Fang J, Wang J, Lin Y, Dai J, Zhong Y, Song H, Zhu S, Wang S, Zhou Z, Sun C, Tang Z, Liao S, Yang G, You Z, Dai X, Mao Z. Pyroptotic-Spatiotemporally Selective Delivery of siRNA against Pyroptosis and Autoimmune Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407115. [PMID: 39081086 DOI: 10.1002/adma.202407115] [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: 05/19/2024] [Revised: 07/09/2024] [Indexed: 10/04/2024]
Abstract
Small-interfering RNAs (siRNAs) offer promising prospects for treating pyroptosis-related autoimmune diseases. However, poor stability and off-target effects during in vivo transportation hinder their practical clinical applications. Precision delivery and adaptive release of siRNAs into inflamed tissues and immune cells could unleash their full therapeutic potential. This study establishes a pyroptotic-spatiotemporally selective siRNA delivery system (PMRC@siGSDME) that selectively targets inflammatory tissues, responds to pyroptosis, and exhibits remarkable therapeutic efficacy against various autoimmune diseases. Novel hybrid nanovesicles (NVs) are designed as a combination of pyroptotic macrophage membranes (PMs) and R8-cardiolipin-containing nanovesicles (RC-NVs). Evidence provides that PM-derived proteins involved in cell-cell interactions and membrane trafficking may contribute to the specificity of NVs to inflammatory tissue. In addition, cardiolipin anchored in the hybrid NVs increases its affinity for activated gasdermin E (GSDME) and achieves pyroptosis-adaptive release of siGSDME for the spatiotemporally selective suppression of immune responses. More importantly, PMRC@siGSDME displays significant anti-inflammatory and therapeutic effects in multiple mouse autoimmune disease models, including arthritis and inflammatory bowel disease (IBD). Collectively, an innovative siRNA delivery strategy precisely tailored for pyroptotic cells has been developed, paving the way for new treatments for autoimmune inflammatory diseases with minimal side effects and wide clinical applicability.
Collapse
Affiliation(s)
- Zongyou Pan
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kaiwang Xu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Guanrui Huang
- Department of Orthopedic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Haoran Hu
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong, 999077, China
| | - Huang Yang
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Haotian Shen
- Department of Orthopedic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Kaijie Qiu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Canlong Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Tengjing Xu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Xinning Yu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Jinhua Fang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Jiajie Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Yunting Lin
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Jiacheng Dai
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Yuting Zhong
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Hongyun Song
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Sunan Zhu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Siheng Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Zhuxing Zhou
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Chuyue Sun
- Department of Otorhinolaryngology, Head and Neck Surgery, Taihe Hospital of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Fuyang, 236000, China
| | - Zhaopeng Tang
- Department of Orthopedic Surgery, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, 730050, China
| | - Shiyao Liao
- Center for Plastic & Reconstructive Surgery, Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, 310014, China
| | - Guang Yang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Zhiyuan You
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xuesong Dai
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Zhengwei Mao
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
6
|
Cheng CK, Yi M, Wang L, Huang Y. Role of gasdermin D in inflammatory diseases: from mechanism to therapeutics. Front Immunol 2024; 15:1456244. [PMID: 39253076 PMCID: PMC11381298 DOI: 10.3389/fimmu.2024.1456244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
Inflammatory diseases compromise a clinically common and diverse group of conditions, causing detrimental effects on body functions. Gasdermins (GSDM) are pore-forming proteins, playing pivotal roles in modulating inflammation. Belonging to the GSDM family, gasdermin D (GSDMD) actively mediates the pathogenesis of inflammatory diseases by mechanistically regulating different forms of cell death, particularly pyroptosis, and cytokine release, in an inflammasome-dependent manner. Aberrant activation of GSDMD in different types of cells, such as immune cells, cardiovascular cells, pancreatic cells and hepatocytes, critically contributes to the persistent inflammation in different tissues and organs. The contributory role of GSDMD has been implicated in diabetes mellitus, liver diseases, cardiovascular diseases, neurodegenerative diseases, and inflammatory bowel disease (IBD). Clinically, alterations in GSDMD levels are potentially indicative to the occurrence and severity of diseases. GSDMD inhibition might represent an attractive therapeutic direction to counteract the progression of inflammatory diseases, whereas a number of GSDMD inhibitors have been shown to restrain GSDMD-mediated pyroptosis through different mechanisms. This review discusses the current understanding and future perspectives on the role of GSDMD in the development of inflammatory diseases, as well as the clinical insights of GSDMD alterations, and therapeutic potential of GSDMD inhibitors against inflammatory diseases. Further investigation on the comprehensive role of GSDM shall deepen our understanding towards inflammation, opening up more diagnostic and therapeutic opportunities against inflammatory diseases.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Min Yi
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
7
|
Xu K, Yang H, Fang J, Qiu K, Shen H, Huang G, Zheng Q, Wang C, Xu T, Yu X, Wang J, Lin Y, Dai J, Zhong Y, Song H, Zhu S, Wang S, Zhou Z, Yang G, Mao Z, Pan Z, Dai X. Self-adaptive pyroptosis-responsive nanoliposomes block pyroptosis in autoimmune inflammatory diseases. Bioact Mater 2024; 36:272-286. [PMID: 38496034 PMCID: PMC10940868 DOI: 10.1016/j.bioactmat.2024.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/03/2024] [Accepted: 02/19/2024] [Indexed: 03/19/2024] Open
Abstract
Nanoliposomes have a broad range of applications in the treatment of autoimmune inflammatory diseases because of their ability to considerably enhance drug transport. For their clinical application, nanoliposomes must be able to realize on-demand release of drugs at disease sites to maximize drug-delivery efficacy and minimize side effects. Therefore, responsive drug-release strategies for inflammation treatment have been explored; however, no specific design has been realized for a responsive drug-delivery system based on pyroptosis-related inflammation. Herein, we report a pioneering strategy for self-adaptive pyroptosis-responsive liposomes (R8-cardiolipin-containing nanoliposomes encapsulating dimethyl fumarate, RC-NL@DMF) that precisely release encapsulated anti-pyroptotic drugs into pyroptotic cells. The activated key pyroptotic protein, the N-terminal domain of gasdermin E, selectively integrates with the cardiolipin of liposomes, thus forming pores for controlled drug release, pyroptosis, and inflammation inhibition. Therefore, RC-NL@DMF exhibited effective therapeutic efficacies to alleviate autoimmune inflammatory damages in zymosan-induced arthritis mice and dextran sulfate sodium-induced inflammatory bowel disease mice. Our novel approach holds great promise for self-adaptive pyroptosis-responsive on-demand drug delivery, suppressing pyroptosis and treating autoimmune inflammatory diseases.
Collapse
Affiliation(s)
- Kaiwang Xu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Huang Yang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jinghua Fang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Kaijie Qiu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | | | | | - Qiangqiang Zheng
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Canlong Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Tengjing Xu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Xinning Yu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Jiajie Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Yunting Lin
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Jiacheng Dai
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Yuting Zhong
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Hongyun Song
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Sunan Zhu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Siheng Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Zhuxing Zhou
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Guang Yang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zongyou Pan
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Xuesong Dai
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| |
Collapse
|
8
|
Rudraprasad D, Gandhi J, Joseph J. Comparative extracellular vesicles proteomics unravels host-pathogen interactions: New insights in bacterial and fungal endophthalmitis in murine models. THE MICROBE 2024; 3:100074. [DOI: 10.1016/j.microb.2024.100074] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Wang Y, Li Y, Yang Y, Swift M, Zhang Z, Wu S, Sun Y, Yang K. In situ vaccination caused by diverse irradiation-driven cell death programs. Theranostics 2024; 14:1147-1167. [PMID: 38323315 PMCID: PMC10845208 DOI: 10.7150/thno.86004] [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: 05/10/2023] [Accepted: 11/20/2023] [Indexed: 02/08/2024] Open
Abstract
Interest surrounding the effect of irradiation on immune activation has exponentially grown within the last decade. This includes work regarding mechanisms of the abscopal effect and the success achieved by combination of radiotherapy and immunotherapy. It is hypothesized that irradiation triggers the immune system to eliminate tumors by inducing tumor cells immunogenic cell death (ICD) in tumor cells. Activation of the ICD pathways can be exploited as an in situ vaccine. In this review, we provide fundamental knowledge of various forms of ICD caused by irradiation, describe the relationship between various cell death pathways and the immune activation effect driven by irradiation, and focus on the therapeutic value of exploiting these cell death programs in the context of irradiation. Furthermore, we summarize the immunomodulatory effect of different cell death programs on combinative radiotherapy and immunotherapy. In brief, differences in cell death programs significantly impact the irradiation-induced immune activation effect. Evaluating the transition between them will provide clues to develop new strategies for radiotherapy and its combination with immunotherapy.
Collapse
Affiliation(s)
- Yijun Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430048, China
| | - Yan Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430048, China
| | - Yuxin Yang
- University of Southern California, Department of Biochemistry and Molecular Medicine
| | - Michelle Swift
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Zhenyu Zhang
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, California 90095-1772, USA
| | - Shuhui Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yajie Sun
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430048, China
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430048, China
| |
Collapse
|
10
|
Li S, Wang A, Wu Y, He S, Shuai W, Zhao M, Zhu Y, Hu X, Luo Y, Wang G. Targeted therapy for non-small-cell lung cancer: New insights into regulated cell death combined with immunotherapy. Immunol Rev 2024; 321:300-334. [PMID: 37688394 DOI: 10.1111/imr.13274] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
Non-small-cell lung cancer (NSCLC), which has a high rate of metastatic spread and drug resistance, is the most common subtype of lung cancer. Therefore, NSCLC patients have a very poor prognosis and a very low chance of survival. Human cancers are closely linked to regulated cell death (RCD), such as apoptosis, autophagy, ferroptosis, pyroptosis, and necroptosis. Currently, small-molecule compounds targeting various types of RCD have shown potential as anticancer treatments. Moreover, RCD appears to be a specific part of the antitumor immune response; hence, the combination of RCD and immunotherapy might increase the inhibitory effect of therapy on tumor growth. In this review, we summarize small-molecule compounds used for the treatment of NSCLC by focusing on RCD and pharmacological systems. In addition, we describe the current research status of an immunotherapy combined with an RCD-based regimen for NSCLC, providing new ideas for targeting RCD pathways in combination with immunotherapy for patients with NSCLC in the future.
Collapse
Affiliation(s)
- Shutong Li
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Aoxue Wang
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yongya Wu
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Shengyuan He
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Wen Shuai
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Min Zhao
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yumeng Zhu
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Xiuying Hu
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yubin Luo
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Guan Wang
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Yadav R, Momin A, Godugu C. DNase based therapeutic approaches for the treatment of NETosis related inflammatory diseases. Int Immunopharmacol 2023; 124:110846. [PMID: 37634446 DOI: 10.1016/j.intimp.2023.110846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/19/2023] [Accepted: 08/20/2023] [Indexed: 08/29/2023]
Abstract
Neutrophils are the primary host innate immune cells defending against pathogens. One proposed mechanism by which neutrophils limit pathogen transmission is NETosis, which includes releasing the nuclear content into the cytosol by forming pores in the plasma membrane. The extrusion of cellular deoxyribonucleic acid (DNA) results in neutrophil extracellular traps (NETs) composed of nuclear DNA associated with histones and granule proteins. NETosis is driven by the enzyme PAD-4 (Peptidylarginine deiminase-4), which converts arginine into citrulline, leading to decondensation of chromatin, separation of DNA, and eventual extrusion. DNase is responsible for the breakdown of NETs. On the one hand, the release of DNase may interfere with the antibacterial effects of NETs; further, DNase may protect tissues from self-destruction caused by the increased release of NET under septic conditions. NETs in physiological quantities are expected to have a role in anti-infectious innate immune responses. In contrast, abnormally high concentrations of NETs in the body that are not adequately cleared by DNases can damage tissues and cause inflammation. Through several novel approaches, it is now possible to avoid the adverse effects caused by the continued release of NETs into the extracellular environment. In this review we have highlighted the basic mechanisms of NETosis, its significance in the pathogenesis of various inflammatory disorders, and the role of DNase enzyme with a focus on the possible function of nanotechnology in its management.
Collapse
Affiliation(s)
- Rachana Yadav
- Department of Biological Sciences (Regulatory Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Alfiya Momin
- Department of Biological Sciences (Regulatory Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Chandraiah Godugu
- Department of Biological Sciences (Regulatory Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India.
| |
Collapse
|
12
|
Cai Y, Chen X, Lu T, Yu Z, Hu S, Liu J, Zhou X, Wang X. Single-cell transcriptome analysis profiles the expression features of TMEM173 in BM cells of high-risk B-cell acute lymphoblastic leukemia. BMC Cancer 2023; 23:372. [PMID: 37095455 PMCID: PMC10123968 DOI: 10.1186/s12885-023-10830-5] [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: 04/19/2022] [Accepted: 04/08/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND As an essential regulator of type I interferon (IFN) response, TMEM173 participates in immune regulation and cell death induction. In recent studies, activation of TMEM173 has been regarded as a promising strategy for cancer immunotherapy. However, transcriptomic features of TMEM173 in B-cell acute lymphoblastic leukemia (B-ALL) remain elusive. METHODS Quantitative real-time PCR (qRT-PCR) and western blotting (WB) were applied to determine the mRNA and protein levels of TMEM173 in peripheral blood mononuclear cells (PBMCs). TMEM173 mutation status was assessed by Sanger sequencing. Single-cell RNA sequencing (scRNA-seq) analysis was performed to explore the expression of TMEM173 in different types of bone marrow (BM) cells. RESULTS The mRNA and protein levels of TMEM173 were increased in PBMCs from B-ALL patients. Besides, frameshift mutation was presented in TMEM173 sequences of 2 B-ALL patients. ScRNA-seq analysis identified the specific transcriptome profiles of TMEM173 in the BM of high-risk B-ALL patients. Specifically, expression levels of TMEM173 in granulocytes, progenitor cells, mast cells, and plasmacytoid dendritic cells (pDCs) were higher than that in B cells, T cells, natural killer (NK) cells, and dendritic cells (DCs). Subset analysis further revealed that TMEM173 and pyroptosis effector gasdermin D (GSDMD) restrained in precursor-B (pre-B) cells with proliferative features, which expressed nuclear factor kappa-B (NF-κB), CD19, and Bruton's tyrosine kinase (BTK) during the progression of B-ALL. In addition, TMEM173 was associated with the functional activation of NK cells and DCs in B-ALL. CONCLUSIONS Our findings provide insights into the transcriptomic features of TMEM173 in the BM of high-risk B-ALL patients. Targeted activation of TMEM173 in specific cells might provide new therapeutic strategies for B-ALL patients.
Collapse
Affiliation(s)
- Yiqing Cai
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Xiaomin Chen
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Tiange Lu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Zhuoya Yu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Shunfeng Hu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Jiarui Liu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong, 250021, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong, 250021, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
13
|
Hildebrandt X, Ibrahim M, Peltzer N. Cell death and inflammation during obesity: "Know my methods, WAT(son)". Cell Death Differ 2023; 30:279-292. [PMID: 36175539 PMCID: PMC9520110 DOI: 10.1038/s41418-022-01062-4] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/08/2022] Open
Abstract
Obesity is a state of low-grade chronic inflammation that causes multiple metabolic diseases. During obesity, signalling via cytokines of the TNF family mediate cell death and inflammation within the adipose tissue, eventually resulting in lipid spill-over, glucotoxicity and insulin resistance. These events ultimately lead to ectopic lipid deposition, glucose intolerance and other metabolic complications with life-threatening consequences. Here we review the literature on how inflammatory responses affect metabolic processes such as energy homeostasis and insulin signalling. This review mainly focuses on the role of cell death in the adipose tissue as a key player in metabolic inflammation.
Collapse
Affiliation(s)
- Ximena Hildebrandt
- University of Cologne, Faculty of Medicine, Centre for Molecular Medicine Cologne (CMMC); Department of Translational Genomics and; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Mohamed Ibrahim
- University of Cologne, Faculty of Medicine, Centre for Molecular Medicine Cologne (CMMC); Department of Translational Genomics and; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Nieves Peltzer
- University of Cologne, Faculty of Medicine, Centre for Molecular Medicine Cologne (CMMC); Department of Translational Genomics and; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany.
| |
Collapse
|
14
|
Liu Y, Zhang Y, Liu Q, Li T, Wang W, Li H, Yang F, Gao W, Li Z, Bai X, Wang Y. Inhibition of DDX3X ameliorated CD4 + T cells pyroptosis and improves survival in septic mice. Mol Immunol 2023; 154:54-60. [PMID: 36603305 DOI: 10.1016/j.molimm.2022.12.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/18/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023]
Abstract
Over-expression of DDX3X mRNA is associated with T cell loss in septic patients. This study aimed to investigate the molecular mechanism of DDX3X on T cell reduction in sepsis. The sepsis model was established using lipopolysaccharide stimulation in vitro and cecal ligation and puncture (CLP) surgery in vivo. Results showed that the expression of DDX3X was significantly upregulated in CD4+ T cells in sepsis. RK-33, the inhibitor of DDX3X, was found to dramatically increase CD4+ T cell counts and prolong the survival rate of mice with sepsis. The results also showed that the expression of caspase-1/GSDMD in CD4+ T cells was significantly increased in vitro and in vivo, and RK-33 can substantially reduce CD4+ T cell pyroptosis through inhibiting NLRP3/caspase-1/GSDMD. Globally, our results suggest that DDX3X is involved in the loss of CD4+ T cells partly through activating the pyroptotic pathway during sepsis, which may provide potential targets for therapeutic interventions in this highly lethal disease.
Collapse
Affiliation(s)
- Yukun Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yongsheng Zhang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Qinxin Liu
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Tianyu Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Wei Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Hui Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Fan Yang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Wei Gao
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Yuchang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
15
|
Schoettler N, Dissanayake E, Craven MW, Yee JS, Eliason J, Schauberger EM, Lemanske RF, Ober C, Gern JE. New Insights Relating Gasdermin B to the Onset of Childhood Asthma. Am J Respir Cell Mol Biol 2022; 67:430-437. [PMID: 35580164 PMCID: PMC9564923 DOI: 10.1165/rcmb.2022-0043ps] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/17/2022] [Indexed: 02/06/2023] Open
Abstract
Chromosome 17q12-q21 is the most replicated genetic locus for childhood-onset asthma. Polymorphisms in this locus containing ∼10 genes interact with a variety of environmental exposures in the home and outdoors to modify asthma risk. However, the functional basis for these associations and their linkages to the environment have remained enigmatic. Within this extended region, regulation of GSDMB (gasdermin B) expression in airway epithelial cells has emerged as the primary mechanism underlying the 17q12-q21 genome-wide association study signal. Asthma-associated SNPs influence the abundance of GSDMB transcripts as well as the functional properties of GSDMB protein in airway epithelial cells. GSDMB is a member of the gasdermin family of proteins, which regulate pyroptosis and inflammatory responses to microbial infections. The aims of this review are to synthesize recent studies on the relationship of 17q12-q21 SNPs to childhood asthma and the evidence pointing to GSDMB gene expression or protein function as the underlying mechanism and to explore the potential functions of GSDMB that may influence the risk of developing asthma during childhood.
Collapse
Affiliation(s)
| | | | - Mark W. Craven
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Jeremiah S. Yee
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Joshua Eliason
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | | | | | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, Illinois; and
| | | |
Collapse
|
16
|
Abstract
Background Pyroptosis has been attracting much attention recently. We have briefly compared its differences and similarities with other programmed deaths and the process of its study. With further exploration of the caspase family, including caspase-1/3/4/5/8/11, new insights into the molecular pathways of action of pyroptosis have been gained. It is also closely related to the development of many cancers, which at the same time provides us with new ideas for the treatment of cancer. Scope of Review We describe what is known regarding the impact of pyroptosis on anticancer immunity and give insight into the potential of harnessing pyroptosis as a tool and applying it to novel or existing anticancer strategies. Major Conclusions Pyroptosis, a caspase-dependent cell death, causes pore formation, cell swelling, rupture of the plasma membrane, and release of all intracellular contents. The role of pyroptosis in cancer is an extremely complex issue. There is growing evidence that tumor pyroptosis has anti-tumor and pro-tumor roles. It should be discussed in different cancer periods according to the characteristics of cancer occurrence and development. In cancer treatment, pyroptosis provides us with some potential new targets. For the existing drugs, the study of pyroptosis also helps us make better use of existing drugs for anticancer treatment. Immunotherapy is a hot research direction in the field of cancer treatment.
Collapse
Affiliation(s)
- Chen Huang
- Department of Abdominal Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, China
| | - Jian Li
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Chenliang Zhang
- Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
17
|
Deoxynivalenol induces caspase-3/GSDME-dependent pyroptosis and inflammation in mouse liver and HepaRG cells. Arch Toxicol 2022; 96:3091-3112. [PMID: 35925383 DOI: 10.1007/s00204-022-03344-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 07/14/2022] [Indexed: 11/02/2022]
Abstract
Deoxynivalenol (DON), a frequent food and feed contaminant, poses a severe threat to human and livestock health. Some studies have demonstrated that DON could induce liver damage and cell death. However, novel cell death styles and detailed mechanisms to explain DON-induced liver inflammatory injury are still lacking. Here, we found both chronic and subacute oral administration of DON (3 mg/kg for 4 weeks and 4 mg/kg for 8 days) induced mouse liver inflammatory injury and activated caspase-3, PARP and gasdermin E (GSDME), which were inhibited by caspase-3 inhibitor Z-DEVD and Ac-DEVD. In vitro, HepaRG cells showed typical pyroptotic characteristics after 32 and 64 μM DON exposure for 24 h, including balloon-like bubbling emerging, release of lactate dehydrogenase (LDH), secretion of IL-1β and IL-6 and activation of caspase-3 and GSDME. Furthermore, knocking down GSDME and inhibiting caspases activity by Z-VAD and Z-DEVD dramatically blocked DON-induced pyroptotic characteristics, while over-expressed GSDME prompted that. These data demonstrate that caspase-3/GSDME pathway plays a key factor in DON-induced pyroptosis and inflammation in liver. Interestingly, knocking down GSDME could inhibit DON-induced pyroptosis but prompt DON-induced apoptosis, while opposite results were obtained when over-expressed GSDME, indicating the critical role of GSDME in DON-induced crosstalk between apoptosis and pyroptosis. Taken together, our data determine DON-induced caspase-3/GSDME-dependent pyroptosis in liver and its role in DON-induced liver inflammatory injury, which provide a novel mechanistic view into DON-induced hepatotoxicity and may offer a new target to reduce latent harm of DON to both humans and animals.
Collapse
|
18
|
Rana N, Privitera G, Kondolf HC, Bulek K, Lechuga S, De Salvo C, Corridoni D, Antanaviciute A, Maywald RL, Hurtado AM, Zhao J, Huang EH, Li X, Chan ER, Simmons A, Bamias G, Abbott DW, Heaney JD, Ivanov AI, Pizarro TT. GSDMB is increased in IBD and regulates epithelial restitution/repair independent of pyroptosis. Cell 2022; 185:283-298.e17. [PMID: 35021065 PMCID: PMC8879997 DOI: 10.1016/j.cell.2021.12.024] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/09/2021] [Accepted: 12/16/2021] [Indexed: 02/08/2023]
Abstract
Gasdermins are a family of structurally related proteins originally described for their role in pyroptosis. Gasdermin B (GSDMB) is currently the least studied, and while its association with genetic susceptibility to chronic mucosal inflammatory disorders is well established, little is known about its functional relevance during active disease states. Herein, we report increased GSDMB in inflammatory bowel disease, with single-cell analysis identifying epithelial specificity to inflamed colonocytes/crypt top colonocytes. Surprisingly, mechanistic experiments and transcriptome profiling reveal lack of inherent GSDMB-dependent pyroptosis in activated epithelial cells and organoids but instead point to increased proliferation and migration during in vitro wound closure, which arrests in GSDMB-deficient cells that display hyper-adhesiveness and enhanced formation of vinculin-based focal adhesions dependent on PDGF-A-mediated FAK phosphorylation. Importantly, carriage of disease-associated GSDMB SNPs confers functional defects, disrupting epithelial restitution/repair, which, altogether, establishes GSDMB as a critical factor for restoration of epithelial barrier function and the resolution of inflammation.
Collapse
Affiliation(s)
- Nitish Rana
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Departments of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Giuseppe Privitera
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Hannah C Kondolf
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Katarzyna Bulek
- Department of Inflammation & Immunity, Learner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Susana Lechuga
- Department of Inflammation & Immunity, Learner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Carlo De Salvo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Daniele Corridoni
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Agne Antanaviciute
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Rebecca L Maywald
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander M Hurtado
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Junjie Zhao
- Department of Inflammation & Immunity, Learner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Emina H Huang
- Departments of Cancer Biology and Colon & Rectal Surgery, Learner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Xiaoxia Li
- Department of Inflammation & Immunity, Learner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - E Ricky Chan
- Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Alison Simmons
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Giorgos Bamias
- Academic Department of Gastroenterology, Ethnikon & Kapodistriakon University of Athens, Laikon Hospital, Athens, Greece
| | - Derek W Abbott
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Andrei I Ivanov
- Department of Inflammation & Immunity, Learner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
19
|
Molecular and structural aspects of gasdermin family pores and insights into gasdermin-elicited programmed cell death. Biochem Soc Trans 2021; 49:2697-2710. [PMID: 34812891 PMCID: PMC8786298 DOI: 10.1042/bst20210672] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 01/04/2023]
Abstract
Pyroptosis is a highly inflammatory and lytic type of programmed cell death (PCD) commenced by inflammasomes, which sense perturbations in the cytosolic environment. Recently, several ground-breaking studies have linked a family of pore-forming proteins known as gasdermins (GSDMs) to pyroptosis. The human genome encodes six GSDM proteins which have a characteristic feature of forming pores in the plasma membrane resulting in the disruption of cellular homeostasis and subsequent induction of cell death. GSDMs have an N-terminal cytotoxic domain and an auto-inhibitory C-terminal domain linked together through a flexible hinge region whose proteolytic cleavage by various enzymes releases the N-terminal fragment that can insert itself into the inner leaflet of the plasma membrane by binding to acidic lipids leading to pore formation. Emerging studies have disclosed the involvement of GSDMs in various modalities of PCD highlighting their role in diverse cellular and pathological processes. Recently, the cryo-EM structures of the GSDMA3 and GSDMD pores were resolved which have provided valuable insights into the pore formation process of GSDMs. Here, we discuss the current knowledge regarding the role of GSDMs in PCD, structural and molecular aspects of autoinhibition, and pore formation mechanism followed by a summary of functional consequences of gasdermin-induced membrane permeabilization.
Collapse
|
20
|
Huang J, Fan P, Liu M, Weng C, Fan G, Zhang T, Duan X, Wu Y, Tang L, Yang G, Liu Y. Famotidine promotes inflammation by triggering cell pyroptosis in gastric cancer cells. BMC Pharmacol Toxicol 2021; 22:62. [PMID: 34686215 PMCID: PMC8539739 DOI: 10.1186/s40360-021-00533-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 10/15/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Cell pyroptosis has been characterized by cell swelling and pro-inflammatory factors release to aggravate inflammatory reaction., such as interlukin-1 beta (IL-1β) and interlukin18 (IL-18). However, the function of famotidine, an antagonist of histamine H2-receptor antagonists, in cell pyroptosis remained unknown. METHODS Real-time quantitative PCR (qPCR), western blotting (WB), LDH release assay and enzyme linked immunosorbent assay (Elisa) combined with inhibitor were performed to analyze the effect of famotidine on cell pyroptosis-related gene expression. RESULTS In this study, we found that famotidine (300 μm) treatment led to a phenomenon of cell pyroptosis as confirmed by LDH assay. Further results showed that famotidine triggered cell pyroptosis in gastric cancer cells by activation of NLPR3 inflammasomes including ASC, Caspase-1 and NLRP, leading to enhanced IL-18, not IL-1β, mature and secretion. What's more, the results also showed GSDME, not GSDMD, was increased in response to famotidine stimulation in BGC823 and AGS cells. Mechanically, phosphorylation of ERK1/2 was drastically enhanced in present with famotidine treatment, while inhibition of ERK1/2 activity by U0126 could reverse the promotion of famotidine in IL-18 secretion. CONCLUSION These findings revealed a novel role of famotidine in cell pyroptosis in patients with gastric cancer, a comprehensive consideration is needed in treatment of gastric cancer.
Collapse
Affiliation(s)
- Jin Huang
- Department of Medical Oncology, the First affiliated Hospital of University of Science and Technology of China(West), Heifei, Anhui, China
| | - Pingsheng Fan
- Department of Medical Oncology, the First affiliated Hospital of University of Science and Technology of China(West), Heifei, Anhui, China
| | - Miao Liu
- Department of Medical Oncology, the First affiliated Hospital of University of Science and Technology of China(West), Heifei, Anhui, China
| | - Chengtao Weng
- Department of Medical Oncology, the First affiliated Hospital of University of Science and Technology of China(West), Heifei, Anhui, China
| | - Gaofei Fan
- Department of Medical Oncology, the First affiliated Hospital of University of Science and Technology of China(West), Heifei, Anhui, China
| | - Tengyue Zhang
- Department of Medical Oncology, the First affiliated Hospital of University of Science and Technology of China(West), Heifei, Anhui, China
| | - Xiaohong Duan
- Department of Medical Oncology, the First affiliated Hospital of University of Science and Technology of China(West), Heifei, Anhui, China
| | - Yang Wu
- Department of Medical Oncology, the First affiliated Hospital of University of Science and Technology of China(West), Heifei, Anhui, China
| | - Lili Tang
- Department of Medical Oncology, the First affiliated Hospital of University of Science and Technology of China(West), Heifei, Anhui, China
| | - Guohong Yang
- Department of Medical Oncology, the First affiliated Hospital of University of Science and Technology of China(West), Heifei, Anhui, China
| | - Yabei Liu
- Department of Medical Oncology, the First affiliated Hospital of University of Science and Technology of China(West), Heifei, Anhui, China.
| |
Collapse
|
21
|
Bagayoko S, Leon-Icaza SA, Pinilla M, Hessel A, Santoni K, Péricat D, Bordignon PJ, Moreau F, Eren E, Boyancé A, Naser E, Lefèvre L, Berrone C, Iakobachvili N, Metais A, Rombouts Y, Lugo-Villarino G, Coste A, Attrée I, Frank DW, Clevers H, Peters PJ, Cougoule C, Planès R, Meunier E. Host phospholipid peroxidation fuels ExoU-dependent cell necrosis and supports Pseudomonas aeruginosa-driven pathology. PLoS Pathog 2021; 17:e1009927. [PMID: 34516571 PMCID: PMC8460005 DOI: 10.1371/journal.ppat.1009927] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 09/23/2021] [Accepted: 08/29/2021] [Indexed: 11/20/2022] Open
Abstract
Regulated cell necrosis supports immune and anti-infectious strategies of the body; however, dysregulation of these processes drives pathological organ damage. Pseudomonas aeruginosa expresses a phospholipase, ExoU that triggers pathological host cell necrosis through a poorly characterized pathway. Here, we investigated the molecular and cellular mechanisms of ExoU-mediated necrosis. We show that cellular peroxidised phospholipids enhance ExoU phospholipase activity, which drives necrosis of immune and non-immune cells. Conversely, both the endogenous lipid peroxidation regulator GPX4 and the pharmacological inhibition of lipid peroxidation delay ExoU-dependent cell necrosis and improve bacterial elimination in vitro and in vivo. Our findings also pertain to the ExoU-related phospholipase from the bacterial pathogen Burkholderia thailandensis, suggesting that exploitation of peroxidised phospholipids might be a conserved virulence mechanism among various microbial phospholipases. Overall, our results identify an original lipid peroxidation-based virulence mechanism as a strong contributor of microbial phospholipase-driven pathology.
Collapse
Affiliation(s)
- Salimata Bagayoko
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Stephen Adonai Leon-Icaza
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Miriam Pinilla
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Audrey Hessel
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Karin Santoni
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - David Péricat
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Pierre-Jean Bordignon
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Flavie Moreau
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
- Level 3 Biosafety Animal Core facility, Anexplo platform, Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Elif Eren
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Aurélien Boyancé
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Emmanuelle Naser
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
- Cytometry & Imaging Core facility, Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Lise Lefèvre
- RESTORE institute, University of Toulouse, CNRS, Toulouse, France
| | - Céline Berrone
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
- Level 3 Biosafety Animal Core facility, Anexplo platform, Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Nino Iakobachvili
- Division of Nanoscopy, Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, The Netherlands
| | - Arnaud Metais
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Yoann Rombouts
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Geanncarlo Lugo-Villarino
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Agnès Coste
- RESTORE institute, University of Toulouse, CNRS, Toulouse, France
| | - Ina Attrée
- Univ. Grenoble Alpes, CNRS, CEA, IBS, Bacterial Pathogenesis and Cellular Responses, Grenoble, France
| | - Dara W. Frank
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, Netherlands
| | - Peter J. Peters
- Division of Nanoscopy, Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, The Netherlands
| | - Céline Cougoule
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Rémi Planès
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Etienne Meunier
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| |
Collapse
|
22
|
Senkevich TG, Yutin N, Wolf YI, Koonin EV, Moss B. Ancient Gene Capture and Recent Gene Loss Shape the Evolution of Orthopoxvirus-Host Interaction Genes. mBio 2021; 12:e0149521. [PMID: 34253028 PMCID: PMC8406176 DOI: 10.1128/mbio.01495-21] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 01/27/2023] Open
Abstract
The survival of viruses depends on their ability to resist host defenses and, of all animal virus families, the poxviruses have the most antidefense genes. Orthopoxviruses (ORPV), a genus within the subfamily Chordopoxvirinae, infect diverse mammals and include one of the most devastating human pathogens, the now eradicated smallpox virus. ORPV encode ∼200 genes, of which roughly half are directly involved in virus genome replication and expression as well as virion morphogenesis. The remaining ∼100 "accessory" genes are responsible for virus-host interactions, particularly counter-defense of innate immunity. Complete sequences are currently available for several hundred ORPV genomes isolated from a variety of mammalian hosts, providing a rich resource for comparative genomics and reconstruction of ORPV evolution. To identify the provenance and evolutionary trends of the ORPV accessory genes, we constructed clusters including the orthologs of these genes from all chordopoxviruses. Most of the accessory genes were captured in three major waves early in chordopoxvirus evolution, prior to the divergence of ORPV and the sister genus Centapoxvirus from their common ancestor. The capture of these genes from the host was followed by extensive gene duplication, yielding several paralogous gene families. In addition, nine genes were gained during the evolution of ORPV themselves. In contrast, nearly every accessory gene was lost, some on multiple, independent occasions in numerous lineages of ORPV, so that no ORPV retains them all. A variety of functional interactions could be inferred from examination of pairs of ORPV accessory genes that were either often or rarely lost concurrently. IMPORTANCE Orthopoxviruses (ORPV) include smallpox (variola) virus, one of the most devastating human pathogens, and vaccinia virus, comprising the vaccine used for smallpox eradication. Among roughly 200 ORPV genes, about half are essential for genome replication and expression as well as virion morphogenesis, whereas the remaining half consists of accessory genes counteracting the host immune response. We reannotated the accessory genes of ORPV, predicting the functions of uncharacterized genes, and reconstructed the history of their gain and loss during the evolution of ORPV. Most of the accessory genes were acquired in three major waves antedating the origin of ORPV from chordopoxviruses. The evolution of ORPV themselves was dominated by gene loss, with numerous genes lost at the base of each major group of ORPV. Examination of pairs of ORPV accessory genes that were either often or rarely lost concurrently during ORPV evolution allows prediction of different types of functional interactions.
Collapse
Affiliation(s)
- Tatiana G. Senkevich
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Instutes of Health, Bethesda, Maryland, USA
| | - Natalya Yutin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Yuri I. Wolf
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Eugene V. Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Instutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
23
|
Loveless R, Bloomquist R, Teng Y. Pyroptosis at the forefront of anticancer immunity. J Exp Clin Cancer Res 2021; 40:264. [PMID: 34429144 PMCID: PMC8383365 DOI: 10.1186/s13046-021-02065-8] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/07/2021] [Indexed: 02/07/2023] Open
Abstract
Tumor resistance to apoptosis and the immunosuppressive tumor microenvironment are two major contributors to poor therapeutic responses during cancer intervention. Pyroptosis, a lytic and inflammatory programmed cell death pathway distinct from apoptosis, has subsequently sparked notable interest among cancer researchers for its potential to be clinically harnessed and to address these problems. Recent evidence indicates that pyroptosis induction in tumor cells leads to a robust inflammatory response and marked tumor regression. Underlying its antitumor effect, pyroptosis is mediated by pore-forming gasdermin proteins that facilitate immune cell activation and infiltration through their release of pro-inflammatory cytokines and immunogenic material following cell rupture. Considering its inflammatory nature, however, aberrant pyroptosis may also be implicated in the formation of a tumor supportive microenvironment, as evidenced by the upregulation of gasdermin proteins in certain cancers. In this review, the molecular pathways leading to pyroptosis are introduced, followed by an overview of the seemingly entangled links between pyroptosis and cancer. We describe what is known regarding the impact of pyroptosis on anticancer immunity and give insight into the potential of harnessing pyroptosis as a tool and applying it to novel or existing anticancer strategies.
Collapse
Affiliation(s)
- Reid Loveless
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Ryan Bloomquist
- Department of Restorative Sciences, Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, 201 Dowman Dr, Atlanta, GA, 30322, USA.
| |
Collapse
|
24
|
Li Y, Wen C, Zhong J, Ling J, Jiang Q. Enterococcus faecalis OG1RF induces apoptosis in MG63 cells via caspase-3/-8/-9 without activation of caspase-1/GSDMD. Oral Dis 2021; 28:2026-2035. [PMID: 34370363 DOI: 10.1111/odi.13996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/05/2021] [Accepted: 08/01/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Regulated cell death is key in the pathogenesis of persistent apical periodontitis. Here, we investigated the mechanisms of regulated cell death in osteoblast-like MG63 cells infected with Enterococcus faecalis OG1RF. MATERIALS AND METHODS MG63 cells were infected with live E. faecalis OG1RF at the indicated multiplicity of infection for the indicated infection time. We evaluated the cells by flow cytometry, terminal deoxynucleotidyl transferase dUTP nick end labelling assay and lactate dehydrogenase release analysis; measured the activity of caspase-1/-3/-8/-9 and the release of interleukin-1β; and determined the expression of apoptosis-associated proteins and gasdermin D by apoptosis antibody array and Western blotting. RESULTS Enterococcus faecalis OG1RF reduced the mitochondrial membrane potential of the infected cells, increased the percentage of apoptotic and terminal deoxynucleotidyl transferase dUTP nick end labelling-positive cells, and enhanced lactate dehydrogenase release. The expression of caspase-3 and survivin and the activity of caspase-3/-8/-9 were upregulated, while the expression of death receptor 6 was downregulated. The activity of caspase-1/gasdermin D and the release of interleukin-1β remained unaltered. CONCLUSION Enterococcus faecalis OG1RF induced both intrinsic and extrinsic MG63 cell apoptosis via caspase-3/-8/-9 activation but did not activate the pyroptotic pathway regulated by caspase-1/gasdermin D.
Collapse
Affiliation(s)
- Yang Li
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Cheng Wen
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Jialin Zhong
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Junqi Ling
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China.,Guangdong Province Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Qianzhou Jiang
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| |
Collapse
|
25
|
Xie J, Fan L, Xiong L, Chen P, Wang H, Chen H, Zhao J, Xu Z, Geng L, Xu W, Gong S. Rabeprazole inhibits inflammatory reaction by inhibition of cell pyroptosis in gastric epithelial cells. BMC Pharmacol Toxicol 2021; 22:44. [PMID: 34266494 PMCID: PMC8283986 DOI: 10.1186/s40360-021-00509-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 07/07/2021] [Indexed: 01/04/2023] Open
Abstract
Background Helicobacter pylori (H. pylori) is a common pathogen in development of peptic ulcers with pyroptosis. Rabeprazole, a critical component of standard triple therapy, has been widely used as the first-line regimen for H. pylori infectious treatment. The aim of this study to explore the function of Rabeprazole on cell pyroptosis in vitro. Methods The clinical sample from patients diagnosed with or without H. pylori-infection were collected to analyze by Immunohistochemistry (IHC). Real-time quantitative PCR (qPCR), western blot (WB) and enzyme linked immunosorbent assay (Elisa) were performed to analyze the effect of Rabeprazole on cell pyroptosis, including LDH, IL-1β and IL-18. Results In this study, we showed that Rabeprazole regulated a phenomenon of cell pyroptosis as confirmed by lactate dehydrogenase (LDH) assay. Further results showed that Rabeprazole inhibited cell pyroptosis in gastric epithelial cells by alleviating GSDMD-executed pyroptosis, leading to decrease IL-1β and IL-18 mature and secretion, which is attributed to NLRP3 inflammasome activation inhibition. Further analysis showed that ASC, NLRP3 and Caspase-1, was significantly repressed in response to Rabeprazole stimulation, resulting in decreasing cleaved-caspase-1 expression. Most important, NLRP3 and GSDMD is significantly increased in gastric tissue of patients with H. pylori infection. Conclusion These findings revealed a critical role of Rabeprazole in cell pyroptosis in patients with H. pylori infection, suggesting that targeting cell pyroptosis is an alternative strategy in improving H. pylori treatment.
Collapse
Affiliation(s)
- Jing Xie
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China.,Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Long Fan
- Department of Pharmacy. Zhuhai Maternal and Child Health Hospital, Zhuhai, China
| | - Liya Xiong
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Peiyu Chen
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Hongli Wang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Huan Chen
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Junhong Zhao
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Zhaohui Xu
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Lanlan Geng
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wanfu Xu
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China. .,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| | - Sitang Gong
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China. .,Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China. .,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
26
|
Wang Y, Zhou X, Zou K, Chen G, Huang L, Yang F, Pan W, Xu H, Xu Z, Chen H, Chen J, Gong S, Zhou X, Xu W, Zhao J. Monocarboxylate Transporter 4 Triggered Cell Pyroptosis to Aggravate Intestinal Inflammation in Inflammatory Bowel Disease. Front Immunol 2021; 12:644862. [PMID: 34093533 PMCID: PMC8170300 DOI: 10.3389/fimmu.2021.644862] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/26/2021] [Indexed: 01/17/2023] Open
Abstract
NLRP3 inflammasome has emerged as a crucial regulator of inflammatory bowel disease (IBD) characterized by a chronic inflammatory disease of the gastrointestinal tract. The expression of MCT4 is significantly increased in intestinal mucosal tissue of IBD, which has been identified to regulate intestinal barrier function. However, the function of MCT4 in cell pyroptosis remained unknown. In this study, we have established a stable cell line with MCT4 overexpression in HT-29 and CaCO2 cells, respectively. Functional analysis revealed that ectopic expression of MCT4 in CaCO2 cells contributed to cell pyroptosis as evidenced by LDH assay, which is largely attributed to Caspase-1-mediated canonical pyroptosis, but not Caspase-4 and Caspase-5, leading to cleave pro-IL-1β and IL-18 into mature form and release mediated by cleaved GSDMD. Mechanically, MCT4 overexpression in HT-29 and CaCO2 cell triggered the phosphorylation of ERK1/2 and NF-κB p65, while inhibition of MCT4 by MCT inhibitor α-Cyano-4-hydroxycinnamic acid (α-CHCA) in HT-29 and CaCO2 cells led to a significant downregulation of ERK1/2 and NF-κB activity. What’s more, blockade of ERK1/2-NF-κB pathway could reverse the promotion effect of MCT4 on IL-1β expression. Importantly, both MCT4 and Caspase-1, GSDMD were significantly increased in patients with IBD, and a positive clinical correlation between MCT4 and Caspase-1 expression was observed (p < 0.001). Taken together, these findings suggested that MCT4 promoted Caspase-1-mediated canonical cell pyroptosis to aggravate intestinal inflammation in intestinal epithelial cells (IECs) through the ERK1/2-NF-κB pathway.
Collapse
Affiliation(s)
- Yaodong Wang
- Department of Gastroenterology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, China
| | - Xiaorong Zhou
- Department of Respiratory and Critical Care, The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, China
| | - Kejian Zou
- Department of General Surgery, Hainan General Hospital, Haikou, China
| | - Guanhua Chen
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ling Huang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Fangying Yang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wenxu Pan
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Hongwei Xu
- Department of Gastroenterology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, China
| | - Zhaohui Xu
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huan Chen
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jiayu Chen
- Department of Neonatal Intensive Care Unit, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Sitang Gong
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xuan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanfu Xu
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Junhong Zhao
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
27
|
Morimoto N, Kono T, Sakai M, Hikima JI. Inflammasomes in Teleosts: Structures and Mechanisms That Induce Pyroptosis during Bacterial Infection. Int J Mol Sci 2021; 22:4389. [PMID: 33922312 PMCID: PMC8122782 DOI: 10.3390/ijms22094389] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Pattern recognition receptors (PRRs) play a crucial role in inducing inflammatory responses; they recognize pathogen-associated molecular patterns, damage-associated molecular patterns, and environmental factors. Nucleotide-binding oligomerization domain-leucine-rich repeat-containing receptors (NLRs) are part of the PRR family; they form a large multiple-protein complex called the inflammasome in the cytosol. In mammals, the inflammasome consists of an NLR, used as a sensor molecule, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) as an adaptor protein, and pro-caspase1 (Casp1). Inflammasome activation induces Casp1 activation, promoting the maturation of proinflammatory cytokines, such as interleukin (IL)-1β and IL-18, and the induction of inflammatory cell death called pyroptosis via gasdermin D cleavage in mammals. Inflammasome activation and pyroptosis in mammals play important roles in protecting the host from pathogen infection. Recently, numerous inflammasome-related genes in teleosts have been identified, and their conservation and/or differentiation between their expression in mammals and teleosts have also been elucidated. In this review, we summarize the current knowledge of the molecular structure and machinery of the inflammasomes and the ASC-spec to induce pyroptosis; moreover, we explore the protective role of the inflammasome against pathogenic infection in teleosts.
Collapse
Affiliation(s)
- Natsuki Morimoto
- Interdisciplinary Graduate School of Agriculture and Engineering, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan;
| | - Tomoya Kono
- Department of Biochemistry and Applied Bioscience, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan; (T.K.); (M.S.)
| | - Masahiro Sakai
- Department of Biochemistry and Applied Bioscience, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan; (T.K.); (M.S.)
| | - Jun-ichi Hikima
- Department of Biochemistry and Applied Bioscience, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan; (T.K.); (M.S.)
| |
Collapse
|