1
|
Zhou M, Pei B, Cai P, Yi C, Akanyibah FA, Lyu C, Mao F. Human umbilical cord mesenchymal stem cell-derived exosomes repair IBD by activating the SIRT1-FXR pathway in macrophages. Stem Cell Res Ther 2025; 16:233. [PMID: 40346712 PMCID: PMC12065267 DOI: 10.1186/s13287-025-04365-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD), a chronic immune disorder, has increasing global incidence and poor treatment outcome. Abnormal macrophage function is implicated in the pathophysiology of IBD. In this study, we investigated the mechanism by which human umbilical cord mesenchymal stem cell-derived exosomes (hucMSC-Ex) inhibit inflammation in IBD mouse and macrophage inflammation models. METHODS We established a dextran sodium sulfate (DSS)-induce BALB/c mice model of IBD and treated with hucMSC-Ex via tail vein to evaluate their repair effect on IBD mice. An in vitro macrophage inflammation model was established using lipopolysaccharide (LPS) and Nigericin (Nig) by stimulating mouse macrophage RAW264.7 and human myeloid leukemia mononuclear (THP-1) cells to assess the repair effect of hucMSC-Ex on macrophage inflammation. EX 527, an effective inhibitor of silent information regulator of transcription 1 (SIRT1), was employed in both the in vivo and in vitro models to explore the effect of hucMSC-Ex on the SIRT1-FXR (farnesoid X receptor) pathway in macrophages during the attenuation of inflammation. RESULTS HucMSC-Ex effectively inhibited inflammation in both the in vivo and in vitro models by up-regulating the expressions of SIRT1 and FXR, which reduced the acetylation level of FXR and inhibited the activation of NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome. The addition of EX 527 further proved that hucMSC-Ex can reduce the acetylation of FXR by activating the SIRT1-FXR pathway, and the decrease of FXR acetylation was directly related to the inhibition of the activity of the NLRP3 inflammasome. CONCLUSION HucMSC-Ex alleviates IBD by reducing the acetylation level of FXR through activating the SIRT1-FXR pathway in macrophages and directly negatively regulating the activation of NLRP3 inflammasomes, thus inhibiting the occurrence of the inflammatory process.
Collapse
Affiliation(s)
- Mengjiao Zhou
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No.8 Dianli Road, Zhenjiang, Jiangsu, 212002, P. R. China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212013, P. R. China
| | - Bing Pei
- Department of Clinical Laboratory, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, Jiangsu, 223800, P. R. China
| | - Peipei Cai
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No.8 Dianli Road, Zhenjiang, Jiangsu, 212002, P. R. China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang College, Zhenjiang, Jiangsu, 212028, P. R. China
| | - Francis Atim Akanyibah
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No.8 Dianli Road, Zhenjiang, Jiangsu, 212002, P. R. China
| | - Changkun Lyu
- School of Medical Technology, Shangqiu Medical College Shangqiu, Shangqiu, Henan, 476100, P. R. China
| | - Fei Mao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No.8 Dianli Road, Zhenjiang, Jiangsu, 212002, P. R. China.
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212013, P. R. China.
| |
Collapse
|
2
|
Froom ZSCS, Callaghan NI, Davenport Huyer L. Cellular crosstalk in fibrosis: insights into macrophage and fibroblast dynamics. J Biol Chem 2025:110203. [PMID: 40334985 DOI: 10.1016/j.jbc.2025.110203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Pathological fibrosis, the excessive deposition of extracellular matrix and tissue stiffening that causes progressive organ dysfunction, underlies diverse chronic diseases. The fibrotic microenvironment is driven by the dynamic microenvironmental interaction between various cell types; macrophages and fibroblasts play central roles in fibrotic disease initiation, maintenance, and progression. Macrophage functional plasticity to microenvironmental stimuli modulates fibroblast functionality by releasing pro-inflammatory cytokines, growth factors, and matrix remodeling enzymes that promote fibroblast proliferation, activation, and differentiation into myofibroblasts. Activated fibroblasts and myofibroblasts serve as the fibrotic effector cells, secreting extracellular matrix components and initiating microenvironmental contracture. Fibroblasts also modulate macrophage function through the release of their own pro-inflammatory cytokines and growth factors, creating bidirectional crosstalk that reinforces the chronic fibrotic cycle. The intricate interplay between macrophages and fibroblasts, including their secretomes and signaling interactions, leads to tissue damage and pathological loss of tissue function. In this review, we examine macrophage-fibroblast reciprocal dynamic interactions in pathological fibrotic conditions. We discuss the specific lineages and functionality of macrophages and fibroblasts implicated in fibrotic progression, with focus on their signal transduction pathways and secretory signalling that enables their pro-fibrotic behaviour. We then finish with a set of recommendations for future experimentation with the goal of developing a set of potential targets for anti-fibrotic therapeutic candidates. Understanding the cellular interactions between macrophages and fibroblasts provides valuable insights into potential therapeutic strategies to mitigate fibrotic disease progression.
Collapse
Affiliation(s)
- Zachary S C S Froom
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Neal I Callaghan
- Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Locke Davenport Huyer
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada; Nova Scotia Health, Halifax, NS B3S 0H6, Canada.
| |
Collapse
|
3
|
Aparicio-Trejo OE, Hernández-Cruz EY, Reyes-Fermín LM, Ceja-Galicia ZA, Pedraza-Chaverri J. The role of redox signaling in mitochondria and endoplasmic reticulum regulation in kidney diseases. Arch Toxicol 2025; 99:1865-1891. [PMID: 40214774 DOI: 10.1007/s00204-025-04041-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/20/2025] [Indexed: 05/18/2025]
Abstract
Kidney diseases are among the fastest worldwide growing pathologies. This growth together with their high mortality rate emphasizes the importance of generating vital information about the mechanism involved in their pathophysiology to determine possible therapeutic targets. Recently, mitochondrial damage and their implication in the reactive oxygen spices (ROS) signaling and redox homeostasis have emerged as a hub point in the pathologic mechanism involved in renal pathologies. ROS in low levels are necessary to maintain cell processes as well as the mitochondria homeostasis and its association with other organelles, especially the with the endoplasmic reticulum (ER). However, the information about how redox signaling interacts and interferes with other cellular processes and the mechanism involved has not been fully integrated. Furthermore, in higher concentrations, these ROS promotes pathologic pathways linked to renal disease progression like, mitochondrial biogenesis reduction, ER stress, calcium overload, inflammation, cell death and fibrosis. Therefore, the aim of this review is to describe the molecular mechanisms involved in the redox signaling influence on mitochondrial and ER homeostasis, focusing on lipid metabolism and ß-oxidation, mitochondrial biogenesis, inflammations, ER stress and calcium homeostasis, as well as the effects of these alteration in the genesis and development of renal disease, with emphasis in acute kidney injury (AKI) and chronic kidney disease (CKD).
Collapse
Affiliation(s)
- Omar Emiliano Aparicio-Trejo
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico
| | | | - Laura María Reyes-Fermín
- Laboratory F-315, Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City, Mexico
| | | | - José Pedraza-Chaverri
- Laboratory F-315, Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City, Mexico.
| |
Collapse
|
4
|
Wu Y, Zhang C, Jin H, Zheng R, Li T, Jin F, Li Y, Gao X, Xu H, Wei Z, Yang J. Comparative analysis of short-term and long-term LL-37-induced rosacea-like mouse models: Histopathological features and inflammatory immune responses. Animal Model Exp Med 2025. [PMID: 40296272 DOI: 10.1002/ame2.70020] [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: 08/21/2024] [Accepted: 03/15/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND It is well recognized that developing new animal models, refining the existing mouse models, and thoroughly characterizing their features are essential for gaining a deeper understanding of rosacea pathogenesis and for advancing therapeutic strategies in this direction. Accordingly, we aimed to characterize the pathological features of a long-term LL-37-induced mouse model of rosacea and to compare the disease manifestations and pathophysiological characteristics between short-term and long-term LL-37-induced models. A key focus was to investigate differential gene expression and the underlying mechanisms of immune system dysregulation in these models. METHODS We comparatively assessed skin lesion manifestations, the extent of inflammatory infiltration, sebaceous gland alterations, fibrosis, and angiogenesis in both models. Assessments were performed using photographic documentation, hematoxylin-eosin (HE) staining, Van Gieson's (VG) staining, immunohistochemistry, and Western blotting. Furthermore, we employed RNA sequencing to analyze differential gene expression in mouse skin. The RNA sequencing data were validated using immunofluorescence staining and Western blotting, with a specific focus on gene variations and mechanisms related to immune system dysregulation. RESULTS Mice subjected to long-term LL-37 induction developed rosacea-like pathological features, including angiogenesis, thickened skin tissue, and sebaceous gland hypertrophy. In the short-term LL-37-induced model, immune dysregulation primarily involved the innate immune response. However, long-term LL-37 induction resulted in significant activation of both innate and adaptive immune responses. CONCLUSION The long-term LL-37-induced mouse model offers a valuable animal model for the detailed investigation of the pathological mechanisms driving moderate-to-severe rosacea with prolonged disease duration. Importantly, this model provides a significant experimental foundation for exploring the potential role of immune system dysregulation in rosacea pathogenesis.
Collapse
Affiliation(s)
- Yiling Wu
- Department of Dermatology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Chuanxi Zhang
- Department of Dermatology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Hui Jin
- Department of Dermatology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Ruiping Zheng
- Department of Dermatology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Tian Li
- School of Public and Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Fuyu Jin
- School of Public and Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Yaqian Li
- School of Public and Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Xuemin Gao
- School of Public and Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Hong Xu
- School of Public and Health, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, China
| | - Zhongqiu Wei
- Department of Pathology, Hebei Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, China
| | - Jie Yang
- Department of Dermatology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| |
Collapse
|
5
|
Chen X, Zhu Y, Xia L, Su S, Fan S, Lu Y, Chen Q, Wei Y, Huang Q, Liu X, Peng X. Glutamine limits NLRP3 inflammasome activation and pyroptosis in macrophages by sustaining the IRG1/itaconate axis. FEBS J 2025. [PMID: 40296302 DOI: 10.1111/febs.70119] [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: 09/02/2024] [Revised: 01/18/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
Aberrant activation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome increases the release of mature pro-inflammatory cytokines interleukin (IL)-1β and IL-18, and enhances pyroptosis; thereby necessitating tight regulation of the NLRP3 inflammasome. Dysfunctional glutamine metabolism contributes to the pathogenesis of multiple inflammatory disorders, and the precise mechanism remains to be elucidated. Here, we provide evidence that glutamine deprivation enhances NLRP3 inflammasome activation in macrophages. Indeed, the absence of exogenous glutamine specifically enhanced NLRP3 inflammasome assembly, thereby accelerating pyroptosis and promoting the maturation of IL-1β and IL-18. Inhibition of glutaminolysis exhibited a similar effect to glutamine deprivation, whereas this effect was reversed by α-ketoglutarate (α-KG), a tricarboxylic acid (TCA)-cycle intermediate that can be replenished by glutamine supply. We further observed reduced generation of endogenous itaconate by glutamine deprivation and verified that both exogenous supplementation of itaconate derivative and increased endogenous itaconate production by overexpressing immune-responsive gene 1 [IRG1; also known as aconitate decarboxylase 1 (ACOD1)] could replace glutamine to inhibit the NLRP3 inflammasome. Mechanistically, glutamine deprivation decreased the source of substrate and inhibited transcription factor EB (TFEB)-dependent transcriptional upregulation of IRG1, thereby impairing the IRG1/itaconate axis that suppresses the NLRP3 inflammasome. Furthermore, glutamine deficiency was detected in a murine sepsis model, whereas extrinsic glutamine supplementation conferred protection against intestinal inflammation and tissue damage in septic mice. Taken together, our findings provide a novel insight into the link between glutamine metabolism and NLRP3 inflammasome activation, highlighting the target of glutamine metabolism, which holds as a potential therapeutic strategy for inflammatory diseases.
Collapse
Affiliation(s)
- Xiaoli Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuanfeng Zhu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lin Xia
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Sen Su
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shijun Fan
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yongling Lu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qian Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan Wei
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qianying Huang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xin Liu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xi Peng
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
6
|
Carnazzo V, Rigante D, Restante G, Basile V, Pocino K, Basile U. The entrenchment of NLRP3 inflammasomes in autoimmune disease-related inflammation. Autoimmun Rev 2025; 24:103815. [PMID: 40233890 DOI: 10.1016/j.autrev.2025.103815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/24/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025]
Abstract
Autoinflammation and autoimmunity are almost "opposite" phenomena characterized by chronic activation of the immune system, 'innate' in the first and 'adaptive' in the second, leading to inflammation of several tissues with specific protean effectors of tissue damage. The mechanism of involvement of multiprotein complexes called 'inflammasomes' within autoimmune pictures, differently from autoinflammatory conditions, is yet undeciphered. In this review we provide a comprehensive overview on NLRP3 inflammasome contribution into the pathogenesis of some autoimmune diseases. In response to autoantibodies against nucleic acids or tissue-specific antigens the NLRP3 inflammasome is activated within dendritic cells and macrophages of patients with systemic lupus erythematosus. Crucial is NLRP3 inflammasome to amplify tissue inflammation with interleukin-1 overexpression and matrix metalloproteinase production at the joint level in rheumatoid arthritis. A deregulated NLRP3 inflammasome activation occurs in the serous acini of salivary and lacrimal glands prone to Sjogren's syndrome, but also in the inflammatory process involving endothelial cells, leucocyte recruitment, and platelet plugging of vasculitides. Furthermore, organ-specific autoimmune diseases such as thyroiditis and hepatitis may display hyperactive NLRP3 inflammasomes at the level of resident immune cells within thyroid or liver, respectively. Therefore, it is not unexpected that preclinical studies have shown how specific inflammasome inhibitors may significantly overthrow the severity of different autoimmune diseases and slow down their trend towards an ominous progression. Specific markers of inflammasome activation could also reveal subclinical inflammatory components escaping conventional diagnostic approaches or improve monitoring of autoimmune diseases and personalizing their treatment.
Collapse
Affiliation(s)
- Valeria Carnazzo
- Department of Clinical Pathology, Santa Maria Goretti Hospital, Latina, Italy.
| | - Donato Rigante
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Università Cattolica Sacro Cuore, Rome, Italy.
| | - Giuliana Restante
- Department of Experimental Medicine, University "La Sapienza", Rome, Italy
| | - Valerio Basile
- Clinical Pathology Unit and Cancer Biobank, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Krizia Pocino
- Unit of Clinical Pathology, Ospedale San Pietro Fatebenefratelli, Rome, Italy
| | - Umberto Basile
- Department of Clinical Pathology, Santa Maria Goretti Hospital, Latina, Italy.
| |
Collapse
|
7
|
Wang C, Ye Y, Zhao M, Chen Q, Liu B, Ren W. Asparagine transporter supports macrophage inflammation via histone phosphorylation. SCIENCE ADVANCES 2025; 11:eads3506. [PMID: 40203093 PMCID: PMC11980831 DOI: 10.1126/sciadv.ads3506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Solute carrier (SLC) family is essential for immune responses; nevertheless, whether and how SLCs regulate macrophage inflammation remains unclear. Here, we demonstrate that K636 acetylation mediates high abundance of SLC6A14 in inflammatory macrophages. Notably, the pharmacological inhibition or genetic modulation of SLC6A14 reduces macrophage interleukin-1β (IL-1β) secretion dependently of lower asparagine uptake and subsequently enhanced nuclear LKB1. Mechanistically, nuclear LKB1 lessens MAPK pathway-mediated NLRP3 inflammasome activation by increased histone 3 S10/28 phosphorylation-dependent cyclin O transcription. Moreover, myeloid Slc6a14 deficiency alleviates pulmonary inflammation via suppressing inflammatory macrophage responses. Overall, these results uncover a network by which SLC6A14-mediated asparagine uptake orchestrates macrophage inflammation through histone phosphorylation, providing a crucial target for modulation of inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Muyang Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qingyi Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Bingnan Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Wenkai Ren
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
8
|
Zhang WG, Zheng XR, Yao Y, Sun WJ, Shao BZ. The role of NLRP3 inflammasome in multiple sclerosis: pathogenesis and pharmacological application. Front Immunol 2025; 16:1572140. [PMID: 40242770 PMCID: PMC11999851 DOI: 10.3389/fimmu.2025.1572140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Multiple sclerosis (MS) is widely acknowledged as a chronic inflammatory autoimmune disorder characterized by central nervous system (CNS) demyelination and neurodegeneration. The hyperactivation of immune and inflammatory responses is recognized as a pivotal factor contributing to the pathogenesis and progression of MS. Among various immune and inflammatory reactions, researchers have increasingly focused on the inflammasome, a complex of proteins. The initiation and activation of the inflammasome are intricately involved in the onset of MS. Notably, the NLRP3 inflammasome, the most extensively studied member of the inflammasome complex, is closely linked with MS. This review will delve into the roles of the NLRP3 inflammasome in the pathogenesis and progression of MS. Additionally, therapeutic strategies targeting the NLRP3 inflammasome for the treatment of MS, including natural compounds, autophagy regulators, and other small molecular compounds, will be detailed in this review.
Collapse
Affiliation(s)
- Wen-Gang Zhang
- The First Medical Center, General Hospital of the Chinese People’s Liberation Army, Beijing, China
| | - Xiao-Rui Zheng
- Medical Supplies Center, General Hospital of the Chinese People’s Liberation Army, Beijing, China
| | - Yi Yao
- The First Medical Center, General Hospital of the Chinese People’s Liberation Army, Beijing, China
| | - Wei-Jia Sun
- Medical Supplies Center, General Hospital of the Chinese People’s Liberation Army, Beijing, China
| | - Bo-Zong Shao
- The First Medical Center, General Hospital of the Chinese People’s Liberation Army, Beijing, China
| |
Collapse
|
9
|
Li G, Guan Y, Xu L, Peng G, Han Q, Wang T, Xu Z, Wen X, Lou H, Shen T. Total alkaloids from Thesium chinense inhibit lipopolysaccharide-induced respiratory inflammation by modulating Nrf2/NF-κB/NLRP3 signaling pathway. Chin J Nat Med 2025; 23:421-430. [PMID: 40274345 DOI: 10.1016/s1875-5364(25)60834-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/07/2024] [Accepted: 04/23/2024] [Indexed: 04/26/2025]
Abstract
Inflammation plays a pivotal role in the etiology and progression of various diseases. In traditional Chinese medicine, the whole plants of Thesium chinense Turcz. and its preparations (e.g. Bairui Granules) have been employed to manage inflammatory conditions. While flavonoids were previously considered the primary anti-inflammatory components, other potentially active constituents have been largely overlooked and not thoroughly investigated. This study presents a novel finding that the total alkaloids of T. chinense (BC-Alk) are potent active substances underlying the traditional and clinical applications of T. chinense and Bairui Granules as anti-inflammatory agents. UPLC-MS/MS analysis identified the composition of BC-Alk as quinolizidine alkaloids. The anti-inflammatory efficacy of BC-Alk was evaluated using a lipopolysaccharide (LPS)-induced lung inflammation model in mice. Results demonstrated that BC-Alk significantly mitigated LPS-induced lung inflammation, attenuated the overproduction of IL-1β and the overproduction of inflammatory factors (TNF-α), and ameliorated lung tissue hyperplasia in mice in vivo. Mechanistic studies in vitro revealed that BC-Alk upregulated the expression of Nrf2 and its downstream proteins NQO1 and glutamate-cystine ligase and modifier subunit (GCLM), inhibited NF-κB phosphorylation, and suppressed NLRP3 activation. Collectively, these findings indicate that BC-Alk exerts potent inhibitory effects against lung inflammation by modulating Nrf2, NF-κB, and NLRP3 pathways. This study provides new insights into the anti-inflammatory constituents of T. chinense and Bairui Granules.
Collapse
Affiliation(s)
- Guohui Li
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Department of Pharmacy, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan 250218, China
| | - Yueqin Guan
- Jiuhua Huayuan Pharmaceutical Co., Ltd., Chuzhou 239011, China
| | - Lintao Xu
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Guangcheng Peng
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Qingtong Han
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tian Wang
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Zhenpeng Xu
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xuesen Wen
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hongxiang Lou
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Tao Shen
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
10
|
Singh V, Ubaid S, Kashif M, Singh T, Singh G, Pahwa R, Singh A. Role of inflammasomes in cancer immunity: mechanisms and therapeutic potential. J Exp Clin Cancer Res 2025; 44:109. [PMID: 40155968 PMCID: PMC11954315 DOI: 10.1186/s13046-025-03366-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025] Open
Abstract
Inflammasomes are multi-protein complexes that detect pathogenic and damage-associated molecular patterns, activating caspase-1, pyroptosis, and the maturation of pro-inflammatory cytokines such as IL-1β and IL-18Within the tumor microenvironment, inflammasomes like NLRP3 play critical roles in cancer initiation, promotion, and progression. Their activation influences the crosstalk between innate and adaptive immunity by modulating immune cell recruitment, cytokine secretion, and T-cell differentiation. While inflammasomes can contribute to tumor growth and metastasis through chronic inflammation, their components also present novel therapeutic targets. Several inhibitors targeting inflammasome components- such as sensor proteins (e.g., NLRP3, AIM2), adaptor proteins (e.g., ASC), caspase-1, and downstream cytokines- are being explored to modulate inflammasome activity. These therapeutic strategies aim to modulate inflammasome activity to enhance anti-tumor immune responses and improve clinical outcomes. Understanding the role of inflammasomes in cancer immunity is crucial for developing interventions that effectively bridge innate and adaptive immune responses for better therapeutic outcomes.
Collapse
Affiliation(s)
- Vivek Singh
- Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Saba Ubaid
- Department of Biochemistry, King George'S Medical University (KGMU), U.P, Lucknow, 226003, India
| | - Mohammad Kashif
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Tanvi Singh
- Department of Biochemistry, King George'S Medical University (KGMU), U.P, Lucknow, 226003, India
| | - Gaurav Singh
- Department of Biochemistry, King George'S Medical University (KGMU), U.P, Lucknow, 226003, India
| | - Roma Pahwa
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anand Singh
- Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
11
|
Jung ES, Choi H, Mook-Jung I. Decoding microglial immunometabolism: a new frontier in Alzheimer's disease research. Mol Neurodegener 2025; 20:37. [PMID: 40149001 PMCID: PMC11948825 DOI: 10.1186/s13024-025-00825-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Alzheimer's disease (AD) involves a dynamic interaction between neuroinflammation and metabolic dysregulation, where microglia play a central role. These immune cells undergo metabolic reprogramming in response to AD-related pathology, with key genes such as TREM2, APOE, and HIF-1α orchestrating these processes. Microglial metabolism adapts to environmental stimuli, shifting between oxidative phosphorylation and glycolysis. Hexokinase-2 facilitates glycolytic flux, while AMPK acts as an energy sensor, coordinating lipid and glucose metabolism. TREM2 and APOE regulate microglial lipid homeostasis, influencing Aβ clearance and immune responses. LPL and ABCA7, both associated with AD risk, modulate lipid processing and cholesterol transport, linking lipid metabolism to neurodegeneration. PPARG further supports lipid metabolism by regulating microglial inflammatory responses. Amino acid metabolism also contributes to microglial function. Indoleamine 2,3-dioxygenase controls the kynurenine pathway, producing neurotoxic metabolites linked to AD pathology. Additionally, glucose-6-phosphate dehydrogenase regulates the pentose phosphate pathway, maintaining redox balance and immune activation. Dysregulated glucose and lipid metabolism, influenced by genetic variants such as APOE4, impair microglial responses and exacerbate AD progression. Recent findings highlight the interplay between metabolic regulators like REV-ERBα, which modulates lipid metabolism and inflammation, and Syk, which influences immune responses and Aβ clearance. These insights offer promising therapeutic targets, including strategies aimed at HIF-1α modulation, which could restore microglial function depending on disease stage. By integrating metabolic, immune, and genetic factors, this review underscores the importance of microglial immunometabolism in AD. Targeting key metabolic pathways could provide novel therapeutic strategies for mitigating neuroinflammation and restoring microglial function, ultimately paving the way for innovative treatments in neurodegenerative diseases.
Collapse
Affiliation(s)
- Eun Sun Jung
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Hayoung Choi
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Inhee Mook-Jung
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.
- Korea Dementia Research Center, Seoul, South Korea.
| |
Collapse
|
12
|
Tang X, Zeng T, Deng W, Zhao W, Liu Y, Huang Q, Deng Y, Xie W, Huang W. Gut microbe-derived betulinic acid alleviates sepsis-induced acute liver injury by inhibiting macrophage NLRP3 inflammasome in mice. mBio 2025; 16:e0302024. [PMID: 39887250 PMCID: PMC11898617 DOI: 10.1128/mbio.03020-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/06/2025] [Indexed: 02/01/2025] Open
Abstract
Sepsis-induced acute liver injury (SALI) is a prevalent and life-threatening complication associated with sepsis. The gut microbiota plays a crucial role in the maintenance of health and the development of diseases. The impact of physical exercise on gut microbiota modulation has been well-documented. However, the potential impact of gut microbiome on exercise training-induced protection against SALI remains uncertain. Here, we discovered exercise training ameliorated SALI and systemic inflammation in septic mice. Notably, gut microbiota pre-depletion abolished the protective effects of exercise training in SALI mice. Fecal microbiota transplantation treatment revealed that exercise training-associated gut microbiota contributed to the beneficial effect of exercise training on SALI. Exercise training modulated the metabolism of Ligilactobacillus and enriched betulinic acid (BA) levels in mice. Functionally, BA treatment conferred protection against SALI by inhibiting the hepatic inflammatory response in mice. BA bound and inactivated hnRNPA2B1, thus suppressing NLRP3 inflammasome activation in macrophages. Collectively, this study reveals gut microbiota is involved in the protective effects of exercise training against SALI, and gut microbiota-derived BA inhibits the hepatic inflammatory response via the hnRNPA2B1-NLRP3 axis, providing a potential therapeutic strategy for SALI. IMPORTANCE Sepsis is characterized by a dysregulated immune response to an infection that leads to multiple organ dysfunction. The occurrence of acute liver injury is frequently observed during the initial stage of sepsis and is directly linked to mortality in the intensive care unit. The preventive effect of physical exercise on SALI is well recognized, yet the underlying mechanism remains poorly elucidated. Exercise training alters the gut microbiome in mice, increasing the abundance of Ligilactobacillus and promoting the generation of BA. Additionally, BA supplementation can suppress the NLRP3 inflammasome activation in macrophages by directly binding to hnRNPA2B1, thereby mitigating SALI. These results highlight the beneficial role of gut microbiota-derived BA in inhibiting the hepatic inflammatory response, which represents a crucial stride toward implementing microbiome-based therapeutic strategies for the clinical management of sepsis.
Collapse
Affiliation(s)
- Xuheng Tang
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Tairan Zeng
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenyan Deng
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Wanning Zhao
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanan Liu
- Department of Critical Care Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Qiaobing Huang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiyu Deng
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Weidang Xie
- Department of Critical Care Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Huang
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Tian H, Zheng J, Wang F, Zhang W, Chen Y, Wang X, Wang X, Xi J, Hu J, Zhang Y. NLRP3 inflammasome promotes functional repair after spinal cord injury in mice by regulating autophagy and its mechanism. Int Immunopharmacol 2025; 149:114230. [PMID: 39922115 DOI: 10.1016/j.intimp.2025.114230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/25/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Inflammation at the injury site exacerbates tissue cell death following a spinal cord injury (SCI). Studies show that NLRP3 inflammasomes are crucial in the inflammation following Spinal Cord Injury, and NLRP3 inflammasomes have been shown to promote cells to undergo excessive autophagy in other diseases. Moreover, excessive autophagy levels could hinder functional repair post-SCI. In this regard, we hypothesized that inhibiting NLRP3 inflammasomes could reduce autophagy levels at the injury site, thus promoting functional repair post-SCI. METHODS Herein, a mouse SCI model was used for in vivo experiments, and an in vitro neuroinflammatory model created using LPS-activated BV2 cells was used for in vitro experiments. Histopathological staining was used to assess tissue repair. Western Blot (WB) and quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) were used to detect changes in relevant autophagy molecules, macrophage polarization-related markers and downstream inflammatory factors, and Immunofluorescence (IF) was used to detect changes in macrophage polarization. RESULTS Following SCI, the inhibition of NLRP3 inflammasomes resulting from intraperitoneal injection of MCC950 significantly reduced autophagy levels at the injury site, resulting in both histological and behavioral improvements. In addition, the phosphorylation of mTOR during inhibition of NLRP3 inflammasomes to reduce autophagy levels further improved the immune microenvironment at the injury site, and M2-type macrophages were significantly upregulated M2-type macrophages. Moreover, in vitro experiments yielded results consistent with those of in vivo experiments regarding changes in autophagy-related indexes and polarization-related markers. CONCLUSIONS Inhibition of NLRP3 inflammasomes can reduce autophagy level at the injury site to promote functional recovery and play a neuroprotective role. Moreover, phosphorylation of mTOR during the process of inhibition of NLRP3 inflammasomes to reduce autophagy, leading to reduced autophagy levels, could improve the immune microenvironment at the injury site, thus promoting functional recovery and histopathological repair post-SCI.
Collapse
Affiliation(s)
- Haozhe Tian
- School of Laboratory Medicine Bengbu Medical University Bengbu China; Anhui Key Laboratory of Tissue Transplantation at Bengbu Medical University Bengbu China
| | - Juan Zheng
- School of Laboratory Medicine Bengbu Medical University Bengbu China; Anhui Key Laboratory of Tissue Transplantation at Bengbu Medical University Bengbu China
| | - Fangli Wang
- School of Laboratory Medicine Bengbu Medical University Bengbu China; Anhui Key Laboratory of Tissue Transplantation at Bengbu Medical University Bengbu China
| | - Wenjing Zhang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases Bengbu China; Clinical laboratory of The First Affiliated Hospital of Bengbu Medical University Bengbu China
| | - Yuqing Chen
- School of Laboratory Medicine Bengbu Medical University Bengbu China
| | - Xiangshu Wang
- School of Laboratory Medicine Bengbu Medical University Bengbu China; Anhui Key Laboratory of Tissue Transplantation at Bengbu Medical University Bengbu China
| | - Xiaoxuan Wang
- School of Laboratory Medicine Bengbu Medical University Bengbu China; Anhui Key Laboratory of Tissue Transplantation at Bengbu Medical University Bengbu China
| | - Jin Xi
- Anhui Key Laboratory of Tissue Transplantation at Bengbu Medical University Bengbu China
| | - Jianguo Hu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases Bengbu China; Clinical laboratory of The First Affiliated Hospital of Bengbu Medical University Bengbu China.
| | - Yuxin Zhang
- School of Laboratory Medicine Bengbu Medical University Bengbu China; Anhui Key Laboratory of Tissue Transplantation at Bengbu Medical University Bengbu China.
| |
Collapse
|
14
|
Hao Z, Wang Y, Chen L, Zhou Y, Fang D, Yao W, Xiao L, Zhang Y. TRIM21 Promotes Endothelial Cell Activation via Accelerating SOCS3 Ubiquitination Degradation in Atherosclerosis. Cardiovasc Toxicol 2025; 25:395-410. [PMID: 39921798 DOI: 10.1007/s12012-025-09965-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/19/2025] [Indexed: 02/10/2025]
Abstract
Activated endothelial cells play an important role in the beginning of atherosclerotic disease by secreting various proteins and inflammatory cytokines. Ubiquitination is one of the most common post-translational changes in cells. However, the role and mechanisms of ubiquitination in endothelial cell activation remain poorly understood. In this study, we identified TRIM21 as an E3 ubiquitin ligase with increased expression in atherosclerotic disease and activated endothelial cells. Knockdown of TRIM21 resulted in reduced secretion of inflammatory factors and attenuated the pyroptosis of endothelial cells, inhibiting the progression of atherosclerosis. Mechanistically, TRIM21 could bind and ubiquitinate SOCS3, thereby enhancing NLRP3-mediated pyroptosis. Taken together, we found that endothelial TRIM21 activated the JAK/STAT3 pathway by degrading SOCS3, which in turn promoted NLRP3-mediated pyroptosis and aggravated atherosclerosis, revealing that TRIM21 may be a promising treatment target for the medical management of atherosclerosis.
Collapse
Affiliation(s)
- Zhenxuan Hao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yihuan Wang
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Linlin Chen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yanjun Zhou
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Dezhou Fang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Wenxiang Yao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Lili Xiao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Yanzhou Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
15
|
Ma C, Chen K, Li L, Jiang M, Zeng Z, Yin F, Yuan J, Jia Y, Zhang H. Epstein-Barr virus infection exacerbates ulcerative colitis by driving macrophage pyroptosis via the upregulation of glycolysis. PRECISION CLINICAL MEDICINE 2025; 8:pbaf002. [PMID: 40041420 PMCID: PMC11878796 DOI: 10.1093/pcmedi/pbaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 03/06/2025] Open
Abstract
Background Epstein-Barr virus (EBV) infection is associated with clinical symptoms, treatment response, need for surgical intervention, and an enhanced likelihood of lymphoma among patients with ulcerative colitis (UC). However, existing studies have primarily concentrated on the epidemiological and clinical associations between EBV and UC, leaving the mechanisms by which EBV exacerbates colitis poorly understood. Methods Clinical specimens of UC patients with EBV infection and a mouse model of dextran sulfate sodium-induced colitis with concurrent murine γ-herpesvirus 68 (MHV-68) infection were utilized to investigate the relationship between EBV infection and macrophage pyroptosis. In vivo, adoptive transfer of MHV-68-induced macrophages and macrophage depletion were performed to elucidate the underlying mechanisms. In vitro, myeloid leukemia mononuclear cells of human (THP-1) and macrophages derived from mouse bone marrow (BMDMs) were stimulated with EBV and MHV-68, respectively, to assess macrophage pyroptosis and glycolysis. Results EBV-induced activation of macrophage pyroptosis was positively correlated with clinical disease activity in UC patients. Furthermore, MHV-68 infection activated pyroptosis by upregulating gasdermin D, NLRP3, interleukin-1β, and interleukin-18 in colonic tissues and peritoneal macrophages of mice with colitis. In vitro, EBV and MHV-68 also mediated activation of pyroptosis in human THP-1 cells and mouse BMDMs, respectively. Additionally, the adoptive transfer of MHV-68-induced BMDMs aggravated murine colitis, whereas macrophage depletion attenuated MHV-68-induced intestinal injury. Mechanistically, MHV-68 promoted macrophage pyroptosis by upregulating glycolysis, while the glycolysis inhibitor, 2-deoxy-D-glucose, blocked this process in vitro. Conclusion EBV infection exacerbates UC by driving macrophage pyroptosis through upregulation of glycolysis, indicating a potential therapeutic approach to mitigate EBV-induced intestinal inflammation.
Collapse
Affiliation(s)
- Chunxiang Ma
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Lab of Inflammatory Bowel Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kexin Chen
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Lab of Inflammatory Bowel Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lili Li
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Lab of Inflammatory Bowel Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mingshan Jiang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Lab of Inflammatory Bowel Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhen Zeng
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Lab of Inflammatory Bowel Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fang Yin
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Lab of Inflammatory Bowel Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Yuan
- Department of Gastroenterology, West China Tianfu Hospital, Sichuan University, Chengdu 610213, China
| | - Yongbin Jia
- Lab of Inflammatory Bowel Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hu Zhang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Lab of Inflammatory Bowel Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Gastroenterology, West China Tianfu Hospital, Sichuan University, Chengdu 610213, China
| |
Collapse
|
16
|
Munshi S, Alarbi AM, Zheng H, Kuplicki R, Burrows K, Figueroa-Hall LK, Victor TA, Aupperle RL, Khalsa SS, Paulus MP, Teague TK, Savitz J. Increased expression of ER stress, inflammasome activation, and mitochondrial biogenesis-related genes in peripheral blood mononuclear cells in major depressive disorder. Mol Psychiatry 2025; 30:574-586. [PMID: 39174649 PMCID: PMC12054637 DOI: 10.1038/s41380-024-02695-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 08/09/2024] [Indexed: 08/24/2024]
Abstract
A subset of major depressive disorder (MDD) is characterized by immune system dysfunction, but the intracellular origin of these immune changes remains unclear. Here we tested the hypothesis that abnormalities in endoplasmic reticulum (ER) stress, inflammasome activity and mitochondrial biogenesis contribute to the development of systemic inflammation in MDD. RT-qPCR was used to measure mRNA expression of key organellar genes from peripheral blood mononuclear cells (PBMCs) isolated from 186 MDD and 67 healthy control (HC) subjects. The comparative CT (2-ΔΔCT) method was applied to quantify mRNA expression using GAPDH as the reference gene. After controlling for age, sex, BMI, and medication status using linear regression models, expression of the inflammasome (NLRC4 and NLRP3) and the ER stress (XBP1u, XBP1s, and ATF4) genes was found to be significantly increased in the MDD versus the HC group. Sensitivity analyses excluding covariates yielded similar results. After excluding outliers, expression of the inflammasome genes was no longer statistically significant but expression of the ER stress genes (XBP1u, XBP1s, and ATF4) remained significant and the mitochondrial biogenesis gene, MFN2, was significantly increased in the MDD group. NLRC4 and MFN2 were positively correlated with serum C-reactive protein concentrations, while ASC trended significant. The altered expression of inflammasome activation, ER stress, and mitochondrial biogenesis pathway components suggest that dysfunction of these organelles may play a role in the pathogenesis of MDD.
Collapse
Affiliation(s)
- Soumyabrata Munshi
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA.
| | - Ahlam M Alarbi
- Integrative Immunology Center, Department of Surgery and Department of Psychiatry, University of Oklahoma - School of Community Medicine, 4502 E. 41st St., Tulsa, OK, 74135, USA
| | - Haixia Zheng
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - Rayus Kuplicki
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
| | - Kaiping Burrows
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
| | - Leandra K Figueroa-Hall
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - Teresa A Victor
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
| | - Robin L Aupperle
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - Sahib S Khalsa
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California at Los Angeles, 300 UCLA Medical Plaza, Los Angeles, CA, 90095, USA
| | - Martin P Paulus
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - T Kent Teague
- Integrative Immunology Center, Department of Surgery and Department of Psychiatry, University of Oklahoma - School of Community Medicine, 4502 E. 41st St., Tulsa, OK, 74135, USA
- Department of Biochemistry and Microbiology, Center for Health Sciences, Oklahoma State University, 1111 W. 17th St., Tulsa, OK, 74107, USA
| | - Jonathan Savitz
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| |
Collapse
|
17
|
Wei S, Han C, Mo S, Huang H, Luo X. Advancements in programmed cell death research in antitumor therapy: a comprehensive overview. Apoptosis 2025; 30:401-421. [PMID: 39487314 DOI: 10.1007/s10495-024-02038-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
Cell death is a normal physiological process within cells that involves multiple pathways, such as normal DNA damage, cell cycle arrest, and programmed cell death (PCD). Cell death has been a hot spot of research in tumor-related fields, especially programmed cell death, which is a key form of cell death and is classified into different types according to the mechanism of occurrence, such as apoptosis, autophagy, necroptosis, pyroptosis, ferroptosis, and disulfidptosis. Given the important role of PCD in maintaining tissue homeostasis and inhibiting tumorigenesis and development, more and more basic and clinical studies are devoted to revealing its potential application in anti-tumor strategies. The purpose of this review is to systematically review the regulatory mechanisms of PCD and to summarize the latest research progress of anti-tumor treatment strategies based on PCD.
Collapse
Affiliation(s)
- Shuxin Wei
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China
| | - Shutian Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Hailian Huang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China
| | - Xiaoling Luo
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China.
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, China.
| |
Collapse
|
18
|
Mao J, Xia W, Wu Y, Li M, Zhao Y, Zhai P, Zhang Y, Zan T, Cui W, Sun X. Biosynthesis of Lysosomally Escaped Apoptotic Bodies Inhibits Inflammasome Synthesis in Macrophages. RESEARCH (WASHINGTON, D.C.) 2025; 8:0581. [PMID: 39850366 PMCID: PMC11754539 DOI: 10.34133/research.0581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/07/2024] [Accepted: 12/22/2024] [Indexed: 01/25/2025]
Abstract
Hyperglycemia and bacterial colonization in diabetic wounds aberrantly activate Nod-like receptor protein 3 (NLRP3) in macrophages, resulting in extensive inflammatory infiltration and impaired wound healing. Targeted suppression of the NLRP3 inflammasome shows promise in reducing macrophage inflammatory disruptions. However, challenges such as drug off-target effects and degradation via lysosomal capture remain during treatment. In this study, engineered apoptotic bodies (BHB-dABs) derived from adipose stem cells loaded with β-hydroxybutyric acid (BHB) were synthesized via biosynthesis. These vesicles target M1-type macrophages, which highly express the folic acid receptor in the inflammatory microenvironment, and facilitate lysosomal escape through 1,2-distearoyl-sn-propyltriyl-3-phosphatidylethanolamine-polyethylene glycol functionalization, which may enhance the efficacy of NLRP3 inhibition for managing diabetic wounds. In vitro studies demonstrated the biocompatibility of BHB-dABs, their selective targeting of M1-type macrophages, and their ability to release BHB within the inflammatory microenvironment via folic acid and folic acid receptor signaling. These nanovesicles exhibited lysosomal escape, anti-inflammatory, mitochondrial protection, and endothelial cell vascularization properties. In vivo experiments demonstrated that BHB-dABs enhance the recovery of diabetic wound inflammation and angiogenesis, accelerating wound healing. These functionalized apoptotic bodies efficiently deliver NLRP3 inflammasome inhibitors using a dual strategy of targeting macrophages and promoting lysosomal escape. This approach represents a novel therapeutic strategy for effectively treating chronic diabetic wounds.
Collapse
Affiliation(s)
- Jiayi Mao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Wenzheng Xia
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Yanglin Wu
- Department of Orthopaedics, Shanghai Tenth People’s Hospital,
Tongji University School of Medicine, Shanghai 200072, P. R. China
| | - Minxiong Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Yun Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Peisong Zhai
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuguang Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Xiaoming Sun
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| |
Collapse
|
19
|
Xi Z, Shu L, Xiao L, Fang X, Dai M, Wang J, Wu Y, Zhang J, Bao M. Macrophage NLRP3 inflammasome mediates the effects of sympathetic nerve on cardiac remodeling in obese rats. Mol Cell Endocrinol 2025; 596:112417. [PMID: 39557185 DOI: 10.1016/j.mce.2024.112417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 11/20/2024]
Abstract
Obesity-associated cardiac remodeling is characterized by cardiac sympathetic nerve over-activation and pro-inflammatory macrophage infiltration. We identified norepinephrine (NE), a sympathetic neurotransmitter, as a pro-inflammatory effector to activate macrophage NLRP3 inflammasome, which contributed to cardiac inflammation. In vivo, Sprague-Dawley (SD) rats were fed a high-fat diet (HFD) for 12 weeks to establish obese rat models. Obese rats exhibited marked cardiac hypertrophy compared to normal rats. The expression of NLRP3 and interleukin (IL)-1β was upregulated, accompanied by CD68+NLRP3+ macrophage infiltration in the hearts of the obese rats. The obese rats also showed increased sympathetic nerve activity. β-adrenergic receptor (AR) inhibition mitigated these changes. In vitro, sympathetic neurotransmitter NE significantly exacerbated palmitic acid (PA)-induced macrophage polarization toward pro-inflammatory type and NLRP3 inflammasome activation in THP-1 macrophages. It was further found that the pro-inflammatory role of NE is dependent on the activation of protein kinase A (PKA) and subsequently inhibition of β-arrestin2, which is an important regulator of the nuclear factor-kappa B (NF-κB) pathway. This study identifies the neuro-immune axis as an important mediator in obesity-associated cardiac remodeling. Targeting the neuro-immune system may open therapeutic opportunities for the treatment of cardiac remodeling in obesity.
Collapse
Affiliation(s)
- Zhaoqing Xi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Ling Shu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Lingling Xiao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Xuesheng Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Mingyan Dai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jing Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China; State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100000, China
| | - Yuan Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Junxia Zhang
- Department of Endocrinology, Taikang Tongji (Wuhan) Hospital, Wuhan, Hubei, 430050, China.
| | - Mingwei Bao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| |
Collapse
|
20
|
Gao H, Sun M, Gao H, Sun Y, Chen W, Dong N. Genome-wide screen based on 2DG activated NLRP3 inflammasome reveals the priming signal of TLR2/4 to IKKβ but not IKKα. Int Immunopharmacol 2025; 145:113781. [PMID: 39657538 DOI: 10.1016/j.intimp.2024.113781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/27/2024] [Accepted: 12/01/2024] [Indexed: 12/12/2024]
Abstract
NLRP3 inflammasome activation is a pivotal area of research in innate immunity, yet the precise priming and activation signal remain unclear. In this study, we demonstrate that glycolysis inhibitor 2-Deoxy-D-glucose (2DG) triggers NLRP3-driven pyroptosis in human leukemia monocyte THP-1 cells by interfering glycosylation rather than glycolysis, which occurs independent of potassium efflux but requires the involvement of glycolysis rate-limiting enzyme PFKP. Using a CRISPR-Cas9 mediated large-scale screen, with 2DG as a new tool for probing NLRP3 activation, we identified that TLR2, rather than TLR4, initiates a rapid and robust priming signal for NLRP3 inflammasome activation. Importantly, both TLR2 and TLR4 depend entirely on MyD88, but not TRIF, for signal transduction. Furthermore, we discovered that TAK1, IKKβ and NEMO, but not IKKα, are essential for the priming signal. Additionally, we observed that deficiency in the linear ubiquitin assembly complex (LUBAC) subunits HOIP and HOIL-1, but not SHARPIN, is sufficient to inhibit 2DG-induced pyroptotic cell death. Collectively, our study reveals some common mechanism in the NLRP3 priming signals, as well as specific mechanisms upstream of NLRP3 triggered by 2DG, and underscores the potential of 2DG as a trigger to facilitate further detailed analysis of the underlying mechanisms of NLRP3 inflammasome activation. One Sentence Summary: Priming signal by IKKβ is essential for NLRP3 activation.
Collapse
Affiliation(s)
- Hui Gao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2#, Haidian District, Beijing 100193, China
| | - Mengning Sun
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2#, Haidian District, Beijing 100193, China
| | - Hang Gao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2#, Haidian District, Beijing 100193, China
| | - Yi Sun
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2#, Haidian District, Beijing 100193, China
| | - Wenjuan Chen
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2#, Haidian District, Beijing 100193, China
| | - Na Dong
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2#, Haidian District, Beijing 100193, China.
| |
Collapse
|
21
|
Kim JK, Sapkota A, Roh T, Jo EK. The intricate interactions between inflammasomes and bacterial pathogens: Roles, mechanisms, and therapeutic potentials. Pharmacol Ther 2025; 265:108756. [PMID: 39581503 DOI: 10.1016/j.pharmthera.2024.108756] [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: 07/08/2024] [Revised: 10/06/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
Inflammasomes are intracellular multiprotein complexes that consist of a sensor, an adaptor, and a caspase enzyme to cleave interleukin (IL)-1β and IL-18 into their mature forms. In addition, caspase-1 and -11 activation results in the cleavage of gasdermin D to form pores, thereby inducing pyroptosis. Activation of the inflammasome and pyroptosis promotes host defense against pathogens, whereas dysregulation of the inflammasome can result in various pathologies. Inflammasomes exhibit versatile microbial signal detection, directly or indirectly, through cellular processes, such as ion fluctuations, reactive oxygen species generation, and the disruption of intracellular organelle function; however, bacteria have adaptive strategies to manipulate the inflammasome by altering microbe-associated molecular patterns, intercepting innate pathways with secreted effectors, and attenuating inflammatory and cell death responses. In this review, we summarize recent advances in the diverse roles of the inflammasome during bacterial infections and discuss how bacteria exploit inflammasome pathways to establish infections or persistence. In addition, we highlight the therapeutic potential of harnessing bacterial immune subversion strategies against acute and chronic bacterial infections. A more comprehensive understanding of the significance of inflammasomes in immunity and their intricate roles in the battle between bacterial pathogens and hosts will lead to the development of innovative strategies to address emerging threats posed by the expansion of drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Asmita Sapkota
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Taylor Roh
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
22
|
Pandey A, Li Z, Gautam M, Ghosh A, Man SM. Molecular mechanisms of emerging inflammasome complexes and their activation and signaling in inflammation and pyroptosis. Immunol Rev 2025; 329:e13406. [PMID: 39351983 PMCID: PMC11742652 DOI: 10.1111/imr.13406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Inflammasomes are multi-protein complexes that assemble within the cytoplasm of mammalian cells in response to pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), driving the secretion of the pro-inflammatory cytokines IL-1β and IL-18, and pyroptosis. The best-characterized inflammasome complexes are the NLRP3, NAIP-NLRC4, NLRP1, AIM2, and Pyrin canonical caspase-1-containing inflammasomes, and the caspase-11 non-canonical inflammasome. Newer inflammasome sensor proteins have been identified, including NLRP6, NLRP7, NLRP9, NLRP10, NLRP11, NLRP12, CARD8, and MxA. These inflammasome sensors can sense PAMPs from bacteria, viruses and protozoa, or DAMPs in the form of mitochondrial damage, ROS, stress and heme. The mechanisms of action, physiological relevance, consequences in human diseases, and avenues for therapeutic intervention for these novel inflammasomes are beginning to be realized. Here, we discuss these emerging inflammasome complexes and their putative activation mechanisms, molecular and signaling pathways, and physiological roles in health and disease.
Collapse
Affiliation(s)
- Abhimanu Pandey
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Zheyi Li
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Manjul Gautam
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Aritra Ghosh
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Si Ming Man
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| |
Collapse
|
23
|
Anand PK. From fat to fire: The lipid-inflammasome connection. Immunol Rev 2025; 329:e13403. [PMID: 39327931 PMCID: PMC11744241 DOI: 10.1111/imr.13403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Inflammasomes are multiprotein complexes that play a crucial role in regulating immune responses by governing the activation of Caspase-1, the secretion of pro-inflammatory cytokines, and the induction of inflammatory cell death, pyroptosis. The inflammasomes are pivotal in effective host defense against a range of pathogens. Yet, overt activation of inflammasome signaling can be detrimental. The most well-studied NLRP3 inflammasome has the ability to detect a variety of stimuli including pathogen-associated molecular patterns, environmental irritants, and endogenous stimuli released from dying cells. Additionally, NLRP3 acts as a key sensor of cellular homeostasis and can be activated by disturbances in diverse metabolic pathways. Consequently, NLRP3 is considered a key player linking metabolic dysregulation to numerous inflammatory disorders such as gout, diabetes, and atherosclerosis. Recently, compelling studies have highlighted a connection between lipids and the regulation of NLRP3 inflammasome. Lipids are integral to cellular processes that serve not only in maintaining the structural integrity and subcellular compartmentalization, but also in contributing to physiological equilibrium. Certain lipid species are known to define NLRP3 subcellular localization, therefore directly influencing the site of inflammasome assembly and activation. For instance, phosphatidylinositol 4-phosphate plays a crucial role in NLRP3 localization to the trans Golgi network. Moreover, new evidence has demonstrated the roles of lipid biosynthesis and trafficking in activation of the NLRP3 inflammasome. This review summarizes and discusses these emerging and varied roles of lipid metabolism in inflammasome activation. A deeper understanding of lipid-inflammasome interactions may open new avenues for therapeutic interventions to prevent or treat chronic inflammatory and autoimmune conditions.
Collapse
Affiliation(s)
- Paras K. Anand
- Department of Infectious Disease, Faculty of MedicineImperial College LondonLondonUK
| |
Collapse
|
24
|
Zhang L, Gao Y, Zhou H, Liang X, Jiang X, Gong W, Sun Y, Zhang D, Wang X, Nauwynck H, Bai J, Jiang P. PRRSV-2 nsp2 Ignites NLRP3 inflammasome through IKKβ-dependent dispersed trans-Golgi network translocation. PLoS Pathog 2025; 21:e1012915. [PMID: 39869629 PMCID: PMC11801707 DOI: 10.1371/journal.ppat.1012915] [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: 11/29/2024] [Revised: 02/06/2025] [Accepted: 01/17/2025] [Indexed: 01/29/2025] Open
Abstract
The NLRP3 inflammasome is a fundamental component of the innate immune system, yet its excessive activation is intricately associated with viral pathogenesis. Porcine reproductive and respiratory syndrome virus type 2 (PRRSV-2), belonging to the family Arteriviridae, triggers dysregulated cytokine release and interstitial pneumonia, which can quickly escalate to acute respiratory distress and death. However, a mechanistic understanding of PRRSV-2 progression remains unclear. Here, we screen that PRRSV-2 nsp2 activates the NLRP3 inflammasome, thereby instigating a state of hyperinflammation. Mechanistically, PRRSV-2 nsp2 interacts with the nucleotide-binding and oligomerization (NACHT) domain of NLRP3, augmenting IKKβ recruitment to driving NLRP3 translocation to the dispersed trans-Golgi network (dTGN) for oligomerization. This process facilitates ASC polymerization, culminating in the activation of the NLRP3 inflammasome. In addition, the IKKβ-dependent NLRP3 translocation to the dTGN is pivotal for pseudorabies virus (PRV) and encephalomyocarditis virus (EMCV)-induced inflammatory responses. Collectively, these results elucidate a novel mechanism of NLRP3 inflammasome activation during PRRSV-2 infection, providing valuable insights into PRRSV-2 pathogenesis.
Collapse
Affiliation(s)
- Lujie Zhang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yanni Gao
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Haoran Zhou
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiao Liang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiaolin Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wenqin Gong
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yangyang Sun
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Desheng Zhang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xianwei Wang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hans Nauwynck
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan Merelbeke, Belgium
| | - Juan Bai
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, PR China
| | - Ping Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, PR China
| |
Collapse
|
25
|
Wang Q, Yang S, Zhang X, Zhang S, Chen L, Wang W, Chen N, Yan J. Inflammasomes in neurodegenerative diseases. Transl Neurodegener 2024; 13:65. [PMID: 39710713 PMCID: PMC11665095 DOI: 10.1186/s40035-024-00459-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/27/2024] [Indexed: 12/24/2024] Open
Abstract
Inflammasomes represent a crucial component of the innate immune system, which respond to threats by recognizing different molecules. These are known as pathogen-associated molecular patterns (PAMPs) or host-derived damage-associated molecular patterns (DAMPs). In neurodegenerative diseases and neuroinflammation, the accumulation of misfolded proteins, such as beta-amyloid and alpha-synuclein, can lead to inflammasome activation, resulting in the release of interleukin (IL)-1β and IL-18. This activation also induces pyroptosis, the release of inflammatory mediators, and exacerbates neuroinflammation. Increasing evidence suggests that inflammasomes play a pivotal role in neurodegenerative diseases. Therefore, elucidating and investigating the activation and regulation of inflammasomes in these diseases is of paramount importance. This review is primarily focused on evidence indicating that inflammasomes are activated through the canonical pathway in these diseases. Inflammasomes as potential targets for treating neurodegenerative diseases are also discussed.
Collapse
Affiliation(s)
- Qianchen Wang
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xuan Zhang
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shanshan Zhang
- China Three Gorges University College of Medicine and Health Sciences, Yichang, 443002, China
| | - Liping Chen
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Wanxue Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Naihong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jiaqing Yan
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
26
|
Yanushkevich S, Zieminska A, Gonzalez J, Añazco F, Song R, Arias-Cavieres A, Granados ST, Zou J, Rao Y, Concepcion AR. Recent advances in the structure, function and regulation of the volume-regulated anion channels and their role in immunity. J Physiol 2024. [PMID: 39709525 DOI: 10.1113/jp285200] [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: 10/07/2024] [Accepted: 11/20/2024] [Indexed: 12/23/2024] Open
Abstract
Volume-regulated anion channels (VRACs) are heteromeric complexes formed by proteins of the leucine-rich repeat-containing 8 (LRRC8) family. LRRC8A (also known as SWELL1) is the core subunit required for VRAC function, and it must combine with one or more of the other paralogues (i.e. LRRC8B-E) to form functional heteromeric channels. VRACs were discovered in T lymphocytes over 35 years ago and are found in virtually all vertebrate cells. Initially, these anion channels were characterized for their role in Cl- efflux during the regulatory volume decrease process triggered when cells are subjected to hypotonic challenges. However, substantial evidence suggests that VRACs also transport small molecules under isotonic conditions. These findings have expanded the research on VRACs to explore their functions beyond volume regulation. In innate immune cells, VRACs promote inflammation by modulating the transport of immunomodulatory cyclic dinucleotides, itaconate and ATP. In adaptive immune cells, VRACs suppress their function by taking up cyclic dinucleotides to activate the STING signalling pathway. In this review, we summarize the current understanding of LRRC8 proteins in immunity and discuss recent progress in their structure, function, regulation and mechanisms for channel activation and gating. Finally, we also examine potential immunotherapeutic applications of VRAC modulation.
Collapse
Affiliation(s)
- Sergei Yanushkevich
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Aleksandra Zieminska
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Joshua Gonzalez
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Francisca Añazco
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Richard Song
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA
| | | | - Sara T Granados
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Junyi Zou
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Yan Rao
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Axel R Concepcion
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
- University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL, USA
| |
Collapse
|
27
|
Cornejo-Báez AA, Zenteno-Cuevas R, Luna-Herrera J. Association Between Diabetes Mellitus-Tuberculosis and the Generation of Drug Resistance. Microorganisms 2024; 12:2649. [PMID: 39770852 PMCID: PMC11728438 DOI: 10.3390/microorganisms12122649] [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: 10/17/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains one of the leading infectious causes of death globally, with drug resistance presenting a significant challenge to control efforts. The interplay between type 2 diabetes mellitus (T2DM) and TB introduces additional complexity, as T2DM triples the risk of active TB and exacerbates drug resistance development. This review explores how T2DM-induced metabolic and immune dysregulation fosters the survival of Mtb, promoting persistence and the emergence of multidrug-resistant strains. Mechanisms such as efflux pump activation and the subtherapeutic levels of isoniazid and rifampicin in T2DM patients are highlighted as key contributors to resistance. We discuss the dual syndemics of T2DM-TB, emphasizing the role of glycemic control and innovative therapeutic strategies, including efflux pump inhibitors and host-directed therapies like metformin. This review underscores the need for integrated diagnostic, treatment, and management approaches to address the global impact of T2DM-TB comorbidity and drug resistance.
Collapse
Affiliation(s)
- Axhell Aleid Cornejo-Báez
- Laboratorio de Inmunoquímica II, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Col. Casco de Santo Tomas, Delegación Miguel Hidalgo, Mexico City C.P. 11340, Mexico;
- Instituto de Salud Pública, Universidad Veracruzana, Av. Luis Castelazo Ayala s/n, A.P. 57, Col. Industrial Animas, Xalapa C.P. 91190, Veracruz, Mexico
| | - Roberto Zenteno-Cuevas
- Instituto de Salud Pública, Universidad Veracruzana, Av. Luis Castelazo Ayala s/n, A.P. 57, Col. Industrial Animas, Xalapa C.P. 91190, Veracruz, Mexico
| | - Julieta Luna-Herrera
- Laboratorio de Inmunoquímica II, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Col. Casco de Santo Tomas, Delegación Miguel Hidalgo, Mexico City C.P. 11340, Mexico;
| |
Collapse
|
28
|
Shang P, Gan M, Wei Z, Hu S, Song L, Feng J, Chen L, Niu L, Wang Y, Zhang S, Shen L, Zhu L, Zhao Y. Advances in research on the impact and mechanisms of pathogenic microorganism infections on pyroptosis. Front Microbiol 2024; 15:1503130. [PMID: 39735183 PMCID: PMC11671501 DOI: 10.3389/fmicb.2024.1503130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 12/02/2024] [Indexed: 12/31/2024] Open
Abstract
Pyroptosis, also known as inflammatory necrosis, is a form of programmed cell death characterized by the activation of gasdermin proteins, leading to the formation of pores in the cell membrane, continuous cell swelling, and eventual membrane rupture. This process results in the release of intracellular contents, including pro-inflammatory cytokines like IL-1β and IL-18, which subsequently trigger a robust inflammatory response. This process is a crucial component of the body's innate immune response and plays a significant role in combating infections. There are four main pathways through which pathogenic microorganisms induce pyroptosis: the canonical inflammasome pathway, the non-canonical inflammasome pathway, the apoptosis-associated caspase-mediated pathway, and the granzyme-mediated pathway. This article provides a brief overview of the effects and mechanisms of pathogen infections on pyroptosis.
Collapse
Affiliation(s)
- Pan Shang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Chengdu, China
| | - Mailin Gan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Chengdu, China
| | - Ziang Wei
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Chengdu, China
| | - Shijie Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Chengdu, China
| | - Lei Song
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Chengdu, China
| | - Jinkang Feng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Chengdu, China
| | - Lei Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Chengdu, China
| | - Lili Niu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Chengdu, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, China
| | - Shunhua Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Chengdu, China
| | - Linyuan Shen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, China
| | - Li Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, China
| | - Ye Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
29
|
Luo L, Zhuang X, Fu L, Dong Z, Yi S, Wang K, Jiang Y, Zhao J, Yang X, Hei F. The role of the interplay between macrophage glycolytic reprogramming and NLRP3 inflammasome activation in acute lung injury/acute respiratory distress syndrome. Clin Transl Med 2024; 14:e70098. [PMID: 39623879 PMCID: PMC11612265 DOI: 10.1002/ctm2.70098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/26/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a severe respiratory condition associated with elevated morbidity and mortality. Understanding their complex pathophysiological mechanisms is crucial for developing new preventive and therapeutic strategies. Recent studies highlight the significant role of inflammation involved in ALI/ARDS, particularly the hyperactivation of the NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome in macrophages. This activation drives pulmonary inflammation by releasing inflammatory signalling molecules and is linked to metabolic reprogramming, marked by increased glycolysis and reduced oxidative phosphorylation. However, the relationship between NLRP3 inflammasome activation and macrophage glycolytic reprogramming in ALI/ARDS, as well as the molecular mechanisms regulating these processes, remain elusive. This review provides a detailed description of the interactions and potential mechanisms linking NLRP3 inflammasome activation with macrophage glycolytic reprogramming, proposing that glycolytic reprogramming may represent a promising therapeutic target for mitigating inflammatory responses in ALI/ARDS. KEY POINTS: NLRP3 inflammasome activation is pivotal in mediating the excessive inflammatory response in ALI/ARDS. Glycolytic reprogramming regulates NLRP3 inflammasome activation. Therapeutic potential of targeting glycolytic reprogramming to inhibit NLRP3 inflammasome activation in ALI/ARDS.
Collapse
Affiliation(s)
- Lan Luo
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Xiaoli Zhuang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Lin Fu
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Ziyuan Dong
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Shuyuan Yi
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Kan Wang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yu Jiang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Ju Zhao
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Xiaofang Yang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Feilong Hei
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
30
|
Vizuete AFK, Fróes F, Seady M, Caurio AC, Ramires Junior OV, Leite AKO, Farias CP, Wyse AT, Gonçalves CA. Targeting glycolysis for neuroprotection in early LPS-induced neuroinflammation. Brain Behav Immun Health 2024; 42:100901. [PMID: 39583162 PMCID: PMC11582448 DOI: 10.1016/j.bbih.2024.100901] [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: 07/01/2024] [Revised: 09/21/2024] [Accepted: 10/27/2024] [Indexed: 11/26/2024] Open
Abstract
Neuroinflammation is a pathophysiological feature of numerous neurological and psychiatric disorders. The immune response in the central nervous system, driven by microglia and astrocytes, leads to metabolic reprogramming towards aerobic glycolysis, a phenomenon known as the Warburg effect. The control of metabolic reprogramming via immunomodulation may represent a potential therapeutic target for providing protection against neuroinflammation, which contributes to neuronal dysfunction and death in several neurological disorders. For this purpose, we investigated putative neuroprotective effects of the downregulation of aerobic glycolysis using the 3PO inhibitor, and the downregulation of neuroinflammation using MCC950, in the early LPS-induced neuroinflammation model. The LPS-induced shift towards glycolysis, inflammatory and glial changes (IL-1β, NF-κB, COX2, Iba1, GFAP) were reversed by 3PO, which improved animal behavior. Additionally, MCC950 (an NLRP3 inhibitor) downregulated TLR4/Akt/p38 MAPK/NF-κB/STAT3 signaling, expressions of COX2 and IL-1β, and the astrocyte reactivity (decreasing GFAP) induced by early neuroinflammation, resulting in low glucose uptake. Our data support the occurrence of the Warburg effect during early neuroinflammation and suggest potential new approaches for the treatment of brain injury, given the role of neuroinflammation in such events.
Collapse
Affiliation(s)
- Adriana Fernanda K. Vizuete
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fernanda Fróes
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Marina Seady
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Aline Castro Caurio
- Post Graduate Program in Biochemistry, Unipampa (Universidade Federal do Pampa) Campus Uruguaiana, Uruguaina, Rio Grande do Sul, Brazil
| | - Osmar Vieira Ramires Junior
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse's Lab), Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana Karla Oliveira Leite
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse's Lab), Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduate Program in Translational Neuroscience (PGNET), National Institute of Translational Neuroscience, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Clarissa Penha Farias
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse's Lab), Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduate Program in Translational Neuroscience (PGNET), National Institute of Translational Neuroscience, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Angela T.S. Wyse
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse's Lab), Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos-Alberto Gonçalves
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
31
|
Tan H, Zhang S, Zhang Z, Zhang J, Wang Z, Liao J, Qiu X, Jia E. Neutrophil extracellular traps promote M1 macrophage polarization in gouty inflammation via targeting hexokinase-2. Free Radic Biol Med 2024; 224:540-553. [PMID: 39277122 DOI: 10.1016/j.freeradbiomed.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/19/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Peptidylarginine deiminase 4 (PAD4)-dependent neutrophil extracellular trap (NET) formation is a new neutrophil death mechanism. Increased NET formation has been demonstrated to be associated with gouty inflammation. Macrophages release proinflammatory mediators and chemokines in acute gouty inflammation and subsequently lead to inflammatory cascades. However, whether NETs regulate macrophage function and polarization and further contribute to gout development remains unclear. Herein, we investigated the relationship between monosodium urate (MSU) crystal-induced NETs and macrophages and the associated mechanisms in gouty inflammation. Elevated NET formation and CD86+ macrophage infiltration were observed in human gouty arthritis (GA). In vitro, MSU crystal-induced NETs or NET-associated histone H3 treatments modulated nod-like receptor protein 3 (NLRP3) inflammasome activation, M1 polarization, and metabolic changes in macrophages. These effects were eliminated by hexokinase-2 (HK-2) silencing. Moreover, NET formation and inflammation were significantly reduced in PAD4-/- GA mice. Pharmacological inhibition of NET formation with Cl-Amidine or NET degradation with DNase Ⅰ significantly reduced M1 polarization of macrophages and ameliorated inflammation in GA mice. In sum, MSU crystal-induced NETs promote M1 polarization and NLRP3 activation in macrophages via targeting HK-2. Cell-free DNA and histone H3 may be the driving elements behind the NET-induced M1 macrophage polarization, NLRP3 activation, and metabolic changes. Targeting NETs could be a potential therapeutic strategy for gout flare.
Collapse
Affiliation(s)
- Haibo Tan
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, 518033, PR China
| | - Shan Zhang
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, 518033, PR China
| | - Zhihao Zhang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Department of Rheumatism, Shenzhen, 518033, PR China
| | - Jianyong Zhang
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, 518033, PR China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Department of Rheumatism, Shenzhen, 518033, PR China
| | - Ziyu Wang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Department of Rheumatism, Shenzhen, 518033, PR China
| | - Junlan Liao
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, 518033, PR China
| | - Xia Qiu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Department of Rheumatism, Shenzhen, 518033, PR China
| | - Ertao Jia
- The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangdong Second Hospital of Traditional Chinese Medicine, Department of Rheumatism, Guangzhou, 510000, PR China.
| |
Collapse
|
32
|
Chang CP, Wu CW, Chern Y. Metabolic dysregulation in Huntington's disease: Neuronal and glial perspectives. Neurobiol Dis 2024; 201:106672. [PMID: 39306013 DOI: 10.1016/j.nbd.2024.106672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a mutant huntingtin protein with an abnormal CAG/polyQ expansion in the N-terminus of HTT exon 1. HD is characterized by progressive neurodegeneration and metabolic abnormalities, particularly in the brain, which accounts for approximately 20 % of the body's resting metabolic rate. Dysregulation of energy homeostasis in HD includes impaired glucose transporters, abnormal functions of glycolytic enzymes, changes in tricarboxylic acid (TCA) cycle activity and enzyme expression in the basal ganglia and cortical regions of both HD mouse models and HD patients. However, current understanding of brain cell behavior during energy dysregulation and its impact on neuron-glia crosstalk in HD remains limited. This review provides a comprehensive summary of the current understanding of the differences in glucose metabolism between neurons and glial cells in HD and how these differences contribute to disease development compared with normal conditions. We also discuss the potential impact of metabolic shifts on neuron-glia communication in HD. A deeper understanding of these metabolic alterations may reveal potential therapeutic targets for future drug development.
Collapse
Affiliation(s)
- Ching-Pang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan; Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ching-Wen Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
33
|
Atalay Ekiner S, Gęgotek A, Skrzydlewska E. Inflammasome activity regulation by PUFA metabolites. Front Immunol 2024; 15:1452749. [PMID: 39290706 PMCID: PMC11405227 DOI: 10.3389/fimmu.2024.1452749] [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: 06/21/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Oxidative stress and the accompanying chronic inflammation constitute an important metabolic problem that may lead to pathology, especially when the body is exposed to physicochemical and biological factors, including UV radiation, pathogens, drugs, as well as endogenous metabolic disorders. The cellular response is associated, among others, with changes in lipid metabolism, mainly due to the oxidation and the action of lipolytic enzymes. Products of oxidative fragmentation/cyclization of polyunsaturated fatty acids (PUFAs) [4-HNE, MDA, 8-isoprostanes, neuroprostanes] and eicosanoids generated as a result of the enzymatic metabolism of PUFAs significantly modify cellular metabolism, including inflammation and the functioning of the immune system by interfering with intracellular molecular signaling. The key regulators of inflammation, the effectiveness of which can be regulated by interacting with the products of lipid metabolism under oxidative stress, are inflammasome complexes. An example is both negative or positive regulation of NLRP3 inflammasome activity by 4-HNE depending on the severity of oxidative stress. 4-HNE modifies NLRP3 activity by both direct interaction with NLRP3 and alteration of NF-κB signaling. Furthermore, prostaglandin E2 is known to be positively correlated with both NLRP3 and NLRC4 activity, while its potential interference with AIM2 or NLRP1 activity is unproven. Therefore, the influence of PUFA metabolites on the activity of well-characterized inflammasome complexes is reviewed.
Collapse
Affiliation(s)
| | - Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
34
|
Leishman S, Aljadeed NM, Qian L, Cockcroft S, Behmoaras J, Anand PK. Fatty acid synthesis promotes inflammasome activation through NLRP3 palmitoylation. Cell Rep 2024; 43:114516. [PMID: 39024103 DOI: 10.1016/j.celrep.2024.114516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/31/2024] [Accepted: 07/01/2024] [Indexed: 07/20/2024] Open
Abstract
Despite its significance, the role of lipid metabolism in NLRP3 inflammasome remains elusive. Here, we reveal a critical role for fatty acid synthase (FASN) in NLRP3 inflammasome activation. We demonstrate that pharmacological or genetic depletion of FASN dampens NLRP3 activation in primary mouse and human macrophages and in mice. This disruption in NLRP3 activation is contingent upon FASN activity. Accordingly, abolishing cellular palmitoylation, a post-translational modification in which the FASN product palmitate is reversibly conjugated to cysteine residues of target proteins, blunts inflammasome signaling. Correspondingly, an acyl-biotin exchange assay corroborated NLRP3 palmitoylation. Mechanistically, Toll-like receptor (TLR) ligation introduces palmitoylation at NLRP3 Cys898, permitting NLRP3 translocation to dispersed trans-Golgi network (dTGN) vesicles, the site of inflammasome assembly, upon NLRP3 activation. Accordingly, the NLRP3 Cys898 mutant exhibits reduced palmitoylation, limited translocation to the dTGN compartment, and diminished inflammasome activation. These results underscore mechanistic insights through which lipid metabolism licenses NLRP3 inflammasome assembly and activation.
Collapse
Affiliation(s)
- Stuart Leishman
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
| | - Najd M Aljadeed
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
| | - Liyunhe Qian
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
| | - Shamshad Cockcroft
- Department of Neuroscience, Physiology and Pharmacology, Division of Biosciences, University College London, London WC1E 6JJ, UK
| | - Jacques Behmoaras
- Programme in Cardiovascular and Metabolic Disorders and Centre for Computational Biology, Duke-NUS Medical School Singapore, Singapore
| | - Paras K Anand
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
35
|
Wang K, Liu J, Yue J, Zhou L, Mao H, Li J, Sun Z, Chen Z, Zhang L. Nlrp3 inflammasome drives regulatory T cell depletion to accelerate periapical bone erosion. Int Endod J 2024; 57:1110-1123. [PMID: 38441141 DOI: 10.1111/iej.14062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 07/03/2024]
Abstract
AIM Apical periodontitis is an inflammatory disorder triggered by an immune response to bacterial infection, leading to the periapical tissue damage and alveolar resorption. However, the underlying mechanisms driving this process remain elusive, due to the complex and interconnected immune microenvironment within the local lesion site. In this study, the influence of Nlrp3 inflammasome-mediated immune response on the apical periodontitis was investigated. METHODOLOGY RNA sequencing, immunohistochemistry and ELISA assay were performed to investigate the activation of Nlrp3 inflammasome signalling pathways in the human periapical tissues, including radicular cysts, periapical granulomas and healthy oral mucosa. A mouse model of apical periodontitis was established to study the role of Nlrp3 knockout in periapical bone resorption and Treg cell stability, and the underlying mechanism was explored through in vitro experiments. In vivo Treg cell adoptive transfer was performed to investigate the effects of Treg cells on the progression of apical periodontitis. RESULTS Our findings find that the hyperactivated Nlrp3 inflammasome is present in human periapical lesions and plays a vital role in the immune-related periapical bone loss. Using a mouse model of apical periodontitis, we observe that Nlrp3 deficiency is resistant to bone resorption. This protection was accompanied by elevated generation and infiltration of local Treg cells that displayed a notable ability to suppress RANKL-dependent osteoclast differentiation. In terms of the mechanism of action, Nlrp3 deficiency directly inhibits the osteoclast differentiation and bone loss through JNK/MAPK and NF-κB pathways. In addition, Nlrp3 induces pyroptosis in the stem cells from apical papilla (SCAPs), and the subsequent release of cytokines affects the stability of Treg cell in periapical lesions, leading indirectly to enhanced bone resorption. In turn, adoptive transfer of both Nlrp3-deficient and wild-type Treg cells effectively prevent the bone erosion during apical periodontitis. CONCLUSIONS Together, our data identify that the Nlrp3 inflammasome modulates the Treg cell stability and osteoclastogenesis in the periapical inflammatory microenvironment, thus determining the progression of bone erosion.
Collapse
Affiliation(s)
- Konghuai Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jiayi Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Junli Yue
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lu Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hanqing Mao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jiaqi Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhijun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
36
|
Chen F, Li S, Liu M, Qian C, Shang Z, Song X, Jiang W, Tu C. Targeting BRD4 mitigates hepatocellular lipotoxicity by suppressing the NLRP3 inflammasome activation and GSDMD-mediated hepatocyte pyroptosis. Cell Mol Life Sci 2024; 81:295. [PMID: 38977508 PMCID: PMC11335218 DOI: 10.1007/s00018-024-05328-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 07/10/2024]
Abstract
Nod-like receptor family pyrin-containing protein 3 (NLRP3) inflammasome plays a pathologic role in metabolic dysfunction-associated steatohepatitis (MASH), but the molecular mechanism regulating the NLRP3 inflammasome activation in hepatocellular lipotoxicity remains largely unknown. Bromodomain-containing protein 4 (BRD4) has emerged as a key epigenetic reader of acetylated lysine residues in enhancer regions that control the transcription of key genes. The aim of this study is to investigate if and how BRD4 regulated the NLRP3 inflammasome activation and pyroptosis in MASH. Using the AML12 and primary mouse hepatocytes stimulated by palmitic acid (PA) as an in vitro model of hepatocellular lipotoxicity, we found that targeting BRD4 by genetic knockdown or a selective BRD4 inhibitor MS417 protected against hepatosteatosis; and this protective effect was attributed to inhibiting the activation of NLRP3 inflammasome and reducing the expression of Caspase-1, gasdermin D (GSDMD), interleukin (IL)-1β and IL-6. Moreover, BRD4 inhibition limited the voltage-dependent anion channel-1 (VDAC1) expression and oligomerization in PA-treated AML12 hepatocytes, thereby suppressing the NLRP3 inflammasome activation. Additionally, the expression of BRD4 enhanced in MASH livers of humans. Mechanistically, BRD4 was upregulated during hepatocellular lipotoxicity that in turn modulated the active epigenetic mark H3K27ac at the promoter regions of the Vdac and Gsdmd genes, thereby enhancing the expression of VDAC and GSDMD. Altogether, our data provide novel insights into epigenetic mechanisms underlying BRD4 activating the NLRP3 inflammasome and promoting GSDMD-mediated pyroptosis in hepatocellular lipotoxicity. Thus, BRD4 might serve as a novel therapeutic target for the treatment of MASH.
Collapse
Affiliation(s)
- Fangyuan Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shuyu Li
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Min Liu
- Department of Gastroenterology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Cheng Qian
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhiyin Shang
- Department of Gastroenterology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Xu Song
- Department of Pathology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Wei Jiang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Department of Gastroenterology and Hepatology, Zhongshan Hospital (Xiamen), Fudan University, Shanghai, 361015, China.
| | - Chuantao Tu
- Department of Gastroenterology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
37
|
Sun Y, Li F, Liu Y, Qiao D, Yao X, Liu GS, Li D, Xiao C, Wang T, Chi W. Targeting inflammasomes and pyroptosis in retinal diseases-molecular mechanisms and future perspectives. Prog Retin Eye Res 2024; 101:101263. [PMID: 38657834 DOI: 10.1016/j.preteyeres.2024.101263] [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: 07/30/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Retinal diseases encompass various conditions associated with sight-threatening immune responses and are leading causes of blindness worldwide. These diseases include age-related macular degeneration, diabetic retinopathy, glaucoma and uveitis. Emerging evidence underscores the vital role of the innate immune response in retinal diseases, beyond the previously emphasized T-cell-driven processes of the adaptive immune system. In particular, pyroptosis, a newly discovered programmed cell death process involving inflammasome formation, has been implicated in the loss of membrane integrity and the release of inflammatory cytokines. Several disease-relevant animal models have provided evidence that the formation of inflammasomes and the induction of pyroptosis in innate immune cells contribute to inflammation in various retinal diseases. In this review article, we summarize current knowledge about the innate immune system and pyroptosis in retinal diseases. We also provide insights into translational targeting approaches, including novel drugs countering pyroptosis, to improve the diagnosis and treatment of retinal diseases.
Collapse
Affiliation(s)
- Yimeng Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Fan Li
- Eye Center, Zhongshan City People's Hospital, Zhongshan, 528403, China
| | - Yunfei Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Dijie Qiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Xinyu Yao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Guei-Sheung Liu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, 3002, Australia; Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Dequan Li
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chuanle Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Tao Wang
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Guangming District, Shenzhen, 518132, China; School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao You'anMen Street, Beijing, 100069, China
| | - Wei Chi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| |
Collapse
|
38
|
Wang Z, Luo W, Zhang G, Li H, Zhou F, Wang D, Feng X, Xiong Y, Wu Y. FoxO1 knockdown inhibits RANKL-induced osteoclastogenesis by blocking NLRP3 inflammasome activation. Oral Dis 2024; 30:3272-3285. [PMID: 37927112 DOI: 10.1111/odi.14800] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023]
Abstract
OBJECTIVES This study aimed to elucidate the connection between osteoclastic forkhead transcription factor O1 (FoxO1) and periodontitis and explore the underlying mechanism by which FoxO1 knockdown regulates osteoclast formation. MATERIALS AND METHODS A conventional ligature-induced periodontitis model was constructed to reveal the alterations in the proportion of osteoclastic FoxO1 in periodontitis via immunofluorescence staining. Additionally, RNA sequencing (RNA-seq) was performed to explore the underlying mechanisms of FoxO1 knockdown-mediated osteoclastogenesis, followed by western blotting, quantitative polymerase chain reaction, and enzyme-linked immunosorbent assay. RESULTS FoxO1+ osteoclasts were enriched in the alveolar bone in experimental periodontitis. Moreover, FoxO1 knockdown led to impaired osteoclastogenesis with low expression of osteoclast differentiation-related genes, accompanied by an insufficient osteoclast maturation phenotype. Mechanistically, RNA-seq revealed that the nuclear factor kappa B (NF-κB) and nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome signaling pathways were inhibited in FoxO1-knockdown osteoclasts. Consistent with this, MCC950, an effective inhibitor of the NLRP3 inflammasome, substantially attenuated osteoclast formation. CONCLUSIONS FoxO1 knockdown contributed to the inhibition of osteoclastogenesis by effectively suppressing NF-κB signaling and NLRP3 inflammasome activation. This prospective study reveals the role of FoxO1 in mediating osteoclastogenesis and provides a viable therapeutic target for periodontitis treatment.
Collapse
Affiliation(s)
- Zhanqi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenxin Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guorui Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Haiyun Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Feng Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dongyang Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuan Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yingying Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Zhou T, Fang YL, Tian TT, Wang GX. Pathological mechanism of immune disorders in diabetic kidney disease and intervention strategies. World J Diabetes 2024; 15:1111-1121. [PMID: 38983817 PMCID: PMC11229953 DOI: 10.4239/wjd.v15.i6.1111] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/29/2024] [Accepted: 04/15/2024] [Indexed: 06/11/2024] Open
Abstract
Diabetic kidney disease is one of the most severe chronic microvascular complications of diabetes and a primary cause of end-stage renal disease. Clinical studies have shown that renal inflammation is a key factor determining kidney damage during diabetes. With the development of immunological technology, many studies have shown that diabetic nephropathy is an immune complex disease, and that most patients have immune dysfunction. However, the immune response associated with diabetic nephropathy and autoimmune kidney disease, or caused by ischemia or infection with acute renal injury, is different, and has a com-plicated pathological mechanism. In this review, we discuss the pathogenesis of diabetic nephropathy in immune disorders and the intervention mechanism, to provide guidance and advice for early intervention and treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Tong Zhou
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun 130021, Jilin Province, China
| | - Yi-Lin Fang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun 130021, Jilin Province, China
| | - Tian-Tian Tian
- School of Public Health, Jilin University, Changchun 130021, Jilin Province, China
| | - Gui-Xia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
40
|
Herwerth M, Wyss MT, Schmid NB, Condrau J, Ravotto L, Mateos Melero JM, Kaech A, Bredell G, Thomas C, Stadelmann C, Misgeld T, Bennett JL, Saab AS, Jessberger S, Weber B. Astrocytes adopt a progenitor-like migratory strategy for regeneration in adult brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.18.594292. [PMID: 38798654 PMCID: PMC11118580 DOI: 10.1101/2024.05.18.594292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Mature astrocytes become activated upon non-specific tissue damage and contribute to glial scar formation. Proliferation and migration of adult reactive astrocytes after injury is considered very limited. However, the regenerative behavior of individual astrocytes following selective astroglial loss, as seen in astrocytopathies, such as neuromyelitis optica spectrum disorder, remains unexplored. Here, we performed longitudinal in vivo imaging of cortical astrocytes after focal astrocyte ablation in mice. We discovered that perilesional astrocytes develop a remarkable plasticity for efficient lesion repopulation. A subset of mature astrocytes transforms into reactive progenitor-like (REPL) astrocytes that not only undergo multiple asymmetric divisions but also remain in a multinucleated interstage. This regenerative response facilitates efficient migration of newly formed daughter cell nuclei towards unoccupied astrocyte territories. Our findings define the cellular principles of astrocyte plasticity upon focal lesion, unravelling the REPL phenotype as a fundamental regenerative strategy of mature astrocytes to restore astrocytic networks in the adult mammalian brain. Promoting this regenerative phenotype bears therapeutic potential for neurological conditions involving glial dysfunction.
Collapse
|
41
|
Wu J, Sun X, Jiang P. Metabolism-inflammasome crosstalk shapes innate and adaptive immunity. Cell Chem Biol 2024; 31:884-903. [PMID: 38759617 DOI: 10.1016/j.chembiol.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 05/19/2024]
Abstract
Inflammasomes are a central component of innate immunity and play a vital role in regulating innate immune response. Activation of inflammasomes is also indispensable for adaptive immunity, modulating the development and response of adaptive immunity. Recently, increasing studies have shown that metabolic alterations and adaptations strongly influence and regulate the differentiation and function of the immune system. In this review, we will take a holistic view of how inflammasomes bridge innate and adaptive (especially T cell) immunity and how inflammasomes crosstalk with metabolic signals during the immune responses. And, special attention will be paid to the metabolic control of inflammasome-mediated interactions between innate and adaptive immunity in disease. Understanding the metabolic regulatory functions of inflammasomes would provide new insights into future research directions in this area and may help to identify potential targets for inflammasome-associated diseases and broaden therapeutic avenues.
Collapse
Affiliation(s)
- Jun Wu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, Fujian, China; State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Xuan Sun
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Peng Jiang
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
42
|
Yurakova TR, Gorshkova EA, Nosenko MA, Drutskaya MS. Metabolic Adaptations and Functional Activity of Macrophages in Homeostasis and Inflammation. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:817-838. [PMID: 38880644 DOI: 10.1134/s0006297924050043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 06/18/2024]
Abstract
In recent years, the role of cellular metabolism in immunity has come into the focus of many studies. These processes form a basis for the maintenance of tissue integrity and homeostasis, as well as represent an integral part of the immune response, in particular, inflammation. Metabolic adaptations not only ensure energy supply for immune response, but also affect the functions of immune cells by controlling transcriptional and post-transcriptional programs. Studying the immune cell metabolism facilitates the search for new treatment approaches, especially for metabolic disorders. Macrophages, innate immune cells, are characterized by a high functional plasticity and play a key role in homeostasis and inflammation. Depending on the phenotype and origin, they can either perform various regulatory functions or promote inflammation state, thus exacerbating the pathological condition. Furthermore, their adaptations to the tissue-specific microenvironment influence the intensity and type of immune response. The review examines the effect of metabolic reprogramming in macrophages on the functional activity of these cells and their polarization. The role of immunometabolic adaptations of myeloid cells in tissue homeostasis and in various pathological processes in the context of inflammatory and metabolic diseases is specifically discussed. Finally, modulation of the macrophage metabolism-related mechanisms reviewed as a potential therapeutic approach.
Collapse
Affiliation(s)
- Taisiya R Yurakova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Ekaterina A Gorshkova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Maxim A Nosenko
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, D02F306, Ireland
| | - Marina S Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
- Division of Immunobiology and Biomedicine, Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, 354340, Russia
| |
Collapse
|
43
|
Feng Y, Jin C, Wang T, Chen Z, Ji D, Zhang Y, Zhang C, Zhang D, Peng W, Sun Y. The Uridylyl Transferase TUT7-Mediated Accumulation of Exosomal miR-1246 Reprograms TAMs to Support CRC Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304222. [PMID: 38342611 PMCID: PMC11022710 DOI: 10.1002/advs.202304222] [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: 06/25/2023] [Revised: 12/06/2023] [Indexed: 02/13/2024]
Abstract
Tumor-associated macrophages (TAMs) play a crucial role in promoting tumor growth and dissemination, motivating a search for key targets to interfere with the activation of TAMs or reprogram TAMs into the tumor-suppressive type. To gain insight into the mechanisms of macrophage polarization, a designed co-culture system is established, allowing for the education of macrophages in a manner that closely mimics the intricacies of TAMs in the tumor immune microenvironment (TIME). Through database mining, exosomal miR-1246 is identified and is then validated. Exosomal miR-1246-driven polarization of TAMs disrupts the infiltration and function of CD8+ T cells. Mechanically, the amassment of exosomal miR-1246 stems from TUT7-mediated degradation of small noncoding RNA, a process stabilized by SNRPB, but not the precursor of miR-1246. Moreover, an Exo-motif is present in the exosomal miR-1246 sequence, enabling it to bind with the exosomal sorting protein hnRNPA2B1. RNA-seq analysis reveals that exogenous miR-1246 modulates the polarization of TAMs at a post-transcriptional level, emphasizing the pivotal role of the NLRP3 in macrophage polarization. In conclusion, the findings underscore the importance of exosomal miR-1246 as a trigger of macrophage reprogramming and uncover a novel mechanism for its enhanced presence in the TIME.
Collapse
Affiliation(s)
- Yifei Feng
- Department of General SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjing210029China
| | - Chi Jin
- Department of General SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjing210029China
| | - Tuo Wang
- Department of General SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjing210029China
| | - Zhihao Chen
- Department of General SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjing210029China
| | - Dongjian Ji
- Department of General SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjing210029China
| | - Yue Zhang
- Department of General SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjing210029China
| | - Chuan Zhang
- Department of General SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjing210029China
| | - Dongsheng Zhang
- Department of General SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjing210029China
| | - Wen Peng
- Department of General SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjing210029China
| | - Yueming Sun
- Department of General SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjing210029China
| |
Collapse
|
44
|
Zuo Z, Shi J, Wang Y, Yin Z, Wang Z, Yang Z, Jia B, Sun Y. The transcriptomic landscape of canonical activation of NLRP3 inflammasome from bone marrow-derived macrophages. Biochem Biophys Res Commun 2024; 694:149409. [PMID: 38141558 DOI: 10.1016/j.bbrc.2023.149409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
The NLRP3 inflammasome has gained significant attention due to its participation in diverse cellular processes. Nevertheless, the detailed framework of the canonical NLRP3 inflammasome assembly still remains unrevealed. This study aims to elucidate the transcriptomic landscape of the various stages involved in the canonical activation of the NLRP3 inflammasome in BMDMs by integrating RNA-seq, bioinformatics, and molecular dynamics analyses. The model for the canonical activation of the NLRP3 inflammasome was confirmed through morphological observations, functional assessments (ELISA and LDH), and protein detection (western blot). Subsequently, cells were subjected to RNA sequencing following three groups: control, priming (LPS 500 ng/ml, 4 h), and activation (LPS 500 ng/ml, 4 h; ATP 5 mM, 1 h). A total of 9116 differentially expressed genes (DEGs) were identified, which exerted regulatory effects on various pathways, including cell metabolism, ion fluxes, post-translational modifications, and organelles. Subsequently, six hub genes (Sirt3, Stat3, Syk, Trpm2, Tspo, and Txnip) were identified via integrating literature review and database screening. Finally, the three-dimensional structures of these six hub proteins were obtained using the MD-optimized RoseTTAFold and Gromacs simulations (at least 200 ns). In summary, our research offers novel insights into the transcriptomic-level understanding of the assembly of the canonical NLRP3 inflammasome.
Collapse
Affiliation(s)
- Zhuo Zuo
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Jiajia Shi
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Yaxing Wang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Zhongqian Yin
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Zhe Wang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Zhouqi Yang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Bin Jia
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China
| | - Yulong Sun
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province, 710072, China.
| |
Collapse
|
45
|
Munshi S, Alarbi A, Zheng H, Kuplicki R, Burrows K, Figueroa-Hall L, Victor T, Aupperle R, Khalsa S, Paulus M, Teague TK, Savitz J. Increased expression of ER stress, inflammasome activation, and mitochondrial biogenesis-related genes in peripheral blood mononuclear cells in major depressive disorder. RESEARCH SQUARE 2024:rs.3.rs-3564760. [PMID: 38260352 PMCID: PMC10802690 DOI: 10.21203/rs.3.rs-3564760/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
A subset of major depressive disorder (MDD) is characterized by immune system dysfunction, but the intracellular origin of these immune changes remains unclear. Here we tested the hypothesis that abnormalities in the endoplasmic reticulum (ER) stress, inflammasome activity and mitochondrial biogenesis contribute to the development of systemic inflammation in MDD. RT-qPCR was used to measure mRNA expression of key organellar genes from peripheral blood mononuclear cells (PBMCs) isolated from 186 MDD and 67 healthy control (HC) subjects. The comparative CT (2-ΔΔCT) method was applied to quantify mRNA expression using GAPDH as the reference gene. After controlling for age, sex, BMI, and medication status using linear regression models, expression of the inflammasome (NLRC4 and NLRP3) and the ER stress (XBP1u, XBP1s, and ATF4) genes was found to be significantly increased in the MDD versus the HC group. After excluding outliers, expression of the inflammasome genes was no longer statistically significant but expression of the ER stress genes (XBP1u, XBP1s, and ATF4) and the mitochondrial biogenesis gene, MFN2, was significantly increased in the MDD group. ASC and MFN2 were positively correlated with serum C-reactive protein concentrations. The altered expression of inflammasome activation, ER stress, and mitochondrial biogenesis pathway components suggest that dysfunction of these organelles may play a role in the pathogenesis of MDD.
Collapse
|
46
|
Kruglov V, Jang IH, Camell CD. Inflammaging and fatty acid oxidation in monocytes and macrophages. IMMUNOMETABOLISM (COBHAM, SURREY) 2024; 6:e00038. [PMID: 38249577 PMCID: PMC10798594 DOI: 10.1097/in9.0000000000000038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
Fatty acid oxidation (FAO), primarily known as β-oxidation, plays a crucial role in breaking down fatty acids within mitochondria and peroxisomes to produce cellular energy and preventing metabolic dysfunction. Myeloid cells, including macrophages, microglia, and monocytes, rely on FAO to perform essential cellular functions and uphold tissue homeostasis. As individuals age, these cells show signs of inflammaging, a condition that includes a chronic onset of low-grade inflammation and a decline in metabolic function. These lead to changes in fatty acid metabolism and a decline in FAO pathways. Recent studies have shed light on metabolic shifts occurring in macrophages and monocytes during aging, correlating with an altered tissue environment and the onset of inflammaging. This review aims to provide insights into the connection of inflammatory pathways and altered FAO in macrophages and monocytes from older organisms. We describe a model in which there is an extended activation of receptor for advanced glycation end products, nuclear factor-κB (NF-κB) and the nod-like receptor family pyrin domain containing 3 inflammasome within macrophages and monocytes. This leads to an increased level of glycolysis, and also promotes pro-inflammatory cytokine production and signaling. As a result, FAO-related enzymes such as 5' AMP-activated protein kinase and peroxisome proliferator-activated receptor-α are reduced, adding to the escalation of inflammation, accumulation of lipids, and heightened cellular stress. We examine the existing body of literature focused on changes in FAO signaling within macrophages and monocytes and their contribution to the process of inflammaging.
Collapse
Affiliation(s)
- Victor Kruglov
- Department of Biochemistry, Molecular Biology, and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - In Hwa Jang
- Department of Biochemistry, Molecular Biology, and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Christina D. Camell
- Department of Biochemistry, Molecular Biology, and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
47
|
Wu J, Yan Y. SIAH1 Promotes the Pyroptosis of Cardiomyocytes in Diabetic Cardiomyopathy via Regulating IκB-α/NF-κВ Signaling. Crit Rev Eukaryot Gene Expr 2024; 34:45-57. [PMID: 38842203 DOI: 10.1615/critreveukaryotgeneexpr.2024052773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Inflammation-mediated dysfunction of cardiomyocytes is the main cause of diabetic cardiomyopathy (DCM). The present study aimed to investigate the roles of siah E3 ubiquitin protein ligase 1 (SIAH1) in DCM. The online dataset GSE4172 was used to analyze the differentially expressed genes in myocardial inflammation of DCM patients. RT-qPCR was conducted to detect mRNA levels. Enzyme-Linked Immunosorbent Assay (ELISA) was performed to detect cytokine release. Western blot was used to detect protein expression. Lactate dehydrogenase (LDH) assay was used to determine cytotoxicity. In vitro ubiquitination assay was applied to determine the ubiquitination of nuclear factor kappa B inhibitor alpha (1κВ-α). Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay was used to detect the death of cardiomyocytes. Flow cytometry was applied for determining cardiomyocyte pyroptosis. The results showed that SIAH1 was overexpressed in human inflammatory cardiomyopathy. High expression of SIAH1 was associated with inflammatory response. SIAH1 was also overexpressed lipopolysaccharide (LPS)-induced inflammatory cardiomyopathy model in vitro. However, SIAH1 knockdown suppressed the inflammatory-related pyroptosis of cardiomyocytes. SIAH1 promoted the ubiquitination of 1κВ-α and activated nuclear factor kappa В (NF-κВ) signaling, which promoted the pyroptosis of cardiomyocytes. In conclusion, SIAH1 exacerbated the progression of human inflammatory cardiomyopathy via inducing the ubiquitination of 1κВ-α and activation of NF-κВ signaling. Therefore, SIAHI/IκB-α/NF-κB signaling may be a potential target for human inflammatory cardiomyopathy.
Collapse
Affiliation(s)
| | - Yaoming Yan
- Laboratory Department, Peking University Shenzhen Hospital, Shenzhen 518036, China
| |
Collapse
|
48
|
Wang Y, Ji X, Wang X, Sun M, Li C, Wu D. The injectable hydrogel loading cannabidiol to regulate macrophage polarization in vitro for the treatment of chronic enteritis. J Appl Biomater Funct Mater 2024; 22:22808000241289022. [PMID: 39385453 DOI: 10.1177/22808000241289022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024] Open
Abstract
OBJECTIVE Chronic bowel disease has the characteristics of high recurrence rate, prolonged and non-healing, and the incidence has increased year by year in recent years. Cannabidiol (CBD) has significant anti-inflammatory and antioxidant activities, but it is limited by its characteristics of fat solubility and low bioavailability. This study aims to treat chronic inflammatory bowel disease by preparing a CBD-loaded hydrogel system (GelMA + CBD) that can deliver CBD in situ and improve its bioavailability through slow release. METHOD The study designed and constructed GelMA + CBD, and its surface morphology was observed by scanning electron microscopy, and its pore size, swelling rate and release rate were evaluated to evaluate its bioactivity and biosafety. The expression of various inflammatory factors was detected by ELISA, and the expression of protein and reactive oxygen species were observed by laser confocal microscopy to evaluate their anti-inflammatory and antioxidant properties. RESULTS Our study found that GelMA + CBD with biosafety, could make CBD be slowly released, and effectively inhibit the M1-type polarization of macrophages in vitro, and promote the M2-type polarization. In addition, GelMA + CBD can also reduce the expression of pro-inflammatory factors (such as iNOS) in macrophages, and increase the expression of anti-inflammatory factors (such as Arg-1), clear intracellular reactive oxygen species (ROS), and relieve oxidative stress. CONCLUSION The vitro experiments have confirmed that the CBD-loaded hydrogel system has good biosafety, and can alleviate inflammation by regulating the polarization direction of macrophages, and then inhibiting the secretion of pro-inflammatory factors, laying a strong foundation for the treatment of chronic enteritis.
Collapse
Affiliation(s)
- Ye Wang
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, China
- Tianjin First Central Hospital, Tianjin, China
| | - Xingming Ji
- Tianjin First Central Hospital, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xinyi Wang
- Tianjin First Central Hospital, Tianjin, China
| | - Mengyu Sun
- Tianjin First Central Hospital, Tianjin, China
| | - Cheng Li
- Tianjin First Central Hospital, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Dongmei Wu
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, China
| |
Collapse
|
49
|
Qin C, Xie T, Yeh WW, Savas AC, Feng P. Metabolic Enzymes in Viral Infection and Host Innate Immunity. Viruses 2023; 16:35. [PMID: 38257735 PMCID: PMC10820379 DOI: 10.3390/v16010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Metabolic enzymes are central players for cell metabolism and cell proliferation. These enzymes perform distinct functions in various cellular processes, such as cell metabolism and immune defense. Because viral infections inevitably trigger host immune activation, viruses have evolved diverse strategies to blunt or exploit the host immune response to enable viral replication. Meanwhile, viruses hijack key cellular metabolic enzymes to reprogram metabolism, which generates the necessary biomolecules for viral replication. An emerging theme arising from the metabolic studies of viral infection is that metabolic enzymes are key players of immune response and, conversely, immune components regulate cellular metabolism, revealing unexpected communication between these two fundamental processes that are otherwise disjointed. This review aims to summarize our present comprehension of the involvement of metabolic enzymes in viral infections and host immunity and to provide insights for potential antiviral therapy targeting metabolic enzymes.
Collapse
Affiliation(s)
- Chao Qin
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | | | | | | | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
50
|
Bornancin F, Dekker C. A phospho-harmonic orchestra plays the NLRP3 score. Front Immunol 2023; 14:1281607. [PMID: 38022631 PMCID: PMC10654991 DOI: 10.3389/fimmu.2023.1281607] [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: 08/22/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
NLRP3 is a prototypical sensor protein connecting cellular stress to pro-inflammatory signaling. A complex array of regulatory steps is required to switch NLRP3 from an inactive state into a primed entity that is poised to assemble an inflammasome. Accumulating evidence suggests that post-translational mechanisms are critical. In particular, phosphorylation/dephosphorylation and ubiquitylation/deubiquitylation reactions have been reported to regulate NLRP3. Taken individually, several post-translational modifications appear to be essential. However, it remains difficult to understand how they may be coordinated, whether there is a unique sequence of regulatory steps accounting for the functional maturation of NLRP3, or whether the sequence is subject to variations depending on cell type, the stimulus, and other parameters such as the cellular context. This review will focus on the regulation of the NLRP3 inflammasome by phosphorylation and dephosphorylation, and on kinases and phosphatases that have been reported to modulate NLRP3 activity. The aim is to try to integrate the current understanding and highlight potential gaps for further studies.
Collapse
Affiliation(s)
| | - Carien Dekker
- Discovery Sciences Department, Novartis Biomedical Research, Basel, Switzerland
| |
Collapse
|