1
|
Tian J, Liu Y, Gao W, Shi X, Cheng F, Xie B. NETs activate AIM2 to mediate synovial fibroblast pyroptosis and promote acute gouty arthritis development. Immunol Lett 2025; 275:107007. [PMID: 40267802 DOI: 10.1016/j.imlet.2025.107007] [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: 01/07/2025] [Revised: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Acute gouty arthritis is a metabolic disease characterized by hyperuricemia, with acute attacks involving neutrophil-released NETs activating immune responses through their major component, DNA, as danger-associated molecular patterns (DAMPs). OBJECTIVE To investigate whether DNA from NETs activates the AIM2 inflammasome in synovial fibroblasts during acute gouty arthritis attacks, inducing pyroptosis and exacerbating inflammation. METHODS The AIM2 gene knockdown mouse model of acute gouty arthritis was constructed, the joint pathological changes were observed by H&E staining, the synovium fibroblasts and neutrophils were sorted by flow cytometry, and the expressions of AIM2, Caspase-1 and GSDMD related proteins were detected by Western blot. The levels of TNF-α, IL-6, IL-1β and IL-18 in serum and cell supernatant were detected by ELISA. Neutrophils were induced to release NETs by urate, and NETs markers (dsDNA, MPO-DNA, NE-DNA) were detected by immunofluorescence (Cit-H3, PAD4) and ELISA. NETs media were co-cultured with synovial fibroblasts, cell activity and migration were evaluated by CCK8 and scrape assay, markers of synovitis (Thy1, VCAM-1, PDPN) were detected by immunofluorescence, and pyroptosis was evaluated by TUNEL and LDH release. By silencing or overexpression of AIM2 gene, Western blot and ELISA, the role of AIM2 in NETs induced pyrodeath and inflammatory response was investigated. RESULTS AIM2 gene knockdown significantly alleviated the symptoms of MSU-induced acute gouty arthritis in mice, reducing joint swelling and pathological damage. Expression levels of inflammatory factors (TNF-α, IL-6, IL-1β, IL-18) and cleaved Caspase-1/Caspase-1, GSDMD-NT/GSDMD) were decreased. It was found that neutrophils released NETs in response to sodium urate stimulation, manifested by significant upregulation of Cit-H3 and PAD4, as well as increased dsDNA, MPO-DNA, and NE-DNA complexes. NETs can induce inflammatory response in synovial fibroblasts, which is manifested as decreased cell activity and migration ability, increased release of inflammatory factors, and significantly increased markers of synovitis (Thy1, VCAM-1, PDPN). In addition, NETs induce scorch death of synovium fibroblasts by activating AIM2 inflammatories, which aggravates the inflammatory response, and AIM2 gene knockdown can effectively inhibit the scorch death and inflammatory response induced by NETs, indicating that NETs play a key role in the occurrence and development of gout arthritis through AIM2-mediated scorch death of synovium fibroblasts. CONCLUSION NETs-activated AIM2-mediated synovial fibroblast pyroptosis plays a crucial role in acute gouty arthritis, providing a new therapeutic target.
Collapse
Affiliation(s)
- Jing Tian
- Department of Orthopedics, General Hospital of Northern Theater, Shenyang City, Liaoning Province, China
| | - Ying Liu
- Department of Emergency, General Hospital of Northern Theater, Shenyang City, Liaoning Province, China
| | - Wei Gao
- Department of Endocrinology, General Hospital of Northern Theater, Shenyang City, Liaoning Province, China
| | - Xiuyun Shi
- Department of Emergency, General Hospital of Northern Theater, Shenyang City, Liaoning Province, China
| | - Feng Cheng
- Department of Emergency, General Hospital of Northern Theater, Shenyang City, Liaoning Province, China
| | - Bing Xie
- Department of Orthopedics, General Hospital of Northern Theater, Shenyang City, Liaoning Province, China.
| |
Collapse
|
2
|
Carrillo-Gálvez AB, Guerra-Valverde JA, Padial-Molina M, Martínez-Cuevas A, Abril-García D, Olaechea A, Martín-Morales N, O'Valle F, Galindo-Moreno P, Zurita F. Cross-talk between NLRP3 and AIM2 inflammasomes in macrophage activation by LPS and titanium ions. Mol Med 2025; 31:223. [PMID: 40490723 DOI: 10.1186/s10020-025-01290-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 05/30/2025] [Indexed: 06/11/2025] Open
Affiliation(s)
- Ana Belén Carrillo-Gálvez
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain.
- Instituto de Investigación Biosanitaria (ibs) de Granada, Granada, Spain.
| | - José Antonio Guerra-Valverde
- Instituto de Investigación Biosanitaria (ibs) de Granada, Granada, Spain
- Clinical Medicine and Public Health, University of Granada, Granada, Spain
| | - Miguel Padial-Molina
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs) de Granada, Granada, Spain
| | | | - Darío Abril-García
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs) de Granada, Granada, Spain
| | - Allinson Olaechea
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs) de Granada, Granada, Spain
- Clinical Medicine and Public Health, University of Granada, Granada, Spain
- GENYO. Centre for Genomics and Oncological Research: Pfizer, University of Granada/Andalusian Regional Government PTS Granada - Avenida de la Ilustración, Granada, 114 - 18016, Spain
| | - Natividad Martín-Morales
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs) de Granada, Granada, Spain
- Department of Pathology, School of Medicine, University of Granada, Granada, Spain
| | - Francisco O'Valle
- Instituto de Investigación Biosanitaria (ibs) de Granada, Granada, Spain
- Department of Pathology, School of Medicine, University of Granada, Granada, Spain
- Institute of Biopathology and Regenerative Medicine (IBIMER, CIBM), University of Granada, Granada, Spain
| | - Pablo Galindo-Moreno
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs) de Granada, Granada, Spain
| | - Federico Zurita
- Department of Genetics, University of Granada, Granada, Spain
- Institute of Biotechnology, University of Granada, Granada, Spain
| |
Collapse
|
3
|
Li P, Yang X, Liu Q, Zhang H, Luo Z. Bladder cancer biomarker analysis and drugtarget prediction based on pyroptosis-related genes. Discov Oncol 2025; 16:924. [PMID: 40415077 DOI: 10.1007/s12672-025-02754-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Bladder cancer (BC) is a common and lethal condition that presents a considerable risk to public health. Studies have demonstrated that inflammation is pivotal in the onset and advancement of BC. Pyroptosis is a type of programmed cell death distinguished by inflammatory reactions associated with innate immunity. Inhibiting inflammatory cytokine production and modulating pyroptosis-related pathways may provide a potential treatment approach for BC. We predicted and validated the Pyroptosis-related genes and potential biomarkers associated with BC, ultimately predicting therapeutic drugs based on the hub gene targets. METHODS The gene expression profiles for BC were acquired from the Gene Expression Omnibus (GEO) database. Bioinformatics analysis identified gene expression differences associated with pyroptosis in BC. The differently regulated pyroptosis-related genes were validated, and enrichment studies of specific biological processes and associated signaling pathways in BC were performed. Immune infiltration analysis and single-cell analysis were conducted to clarify the immune infiltration characteristics in BC. Therapeutic agents were forecasted based on critical gene targets. RESULTS In BC, 27 differentially expressed pyroptosis-related genes were discovered, with CASP8, NLRP3, CASP3, IL18, TP53, GSDME, IL1A, PYCARD, CYCS, and CASP9 recognized as key genes. Enrichment analysis revealed that the occurrence of pyroptosis was primarily associated with inflammation, activation of immune responses, and apoptosis. Additionally, data validation demonstrated that CASP8, NLRP3, CASP3, IL18, TP53, CYCS, and CASP9 were involved in the regulation of pyroptosis. The results of immune infiltration and single-cell analyses further validated that B-cells-memory, T-cells_CD8, T-cells_follicular-helper, Macrophages-M1, Dendritic_cells_activated, and Mast_cells_resting play significant roles in the immune processes of BC. The drug targeting predictions for pivotal genes identified Triethyl phosphate, Regorafenib, Ponatinib, Lenvatinib, Nintedanib, and Quercetin as potential key drugs or compounds for the treatment of BC. CONCLUSION This study elucidated the relationship between the development of BC and mechanisms of cellular senescence, apoptosis, and immunity. It clarified the roles of 27 genes associated with cellular senescence in BC and predicted that Triethyl phosphate, Regorafenib, Ponatinib, Lenvatinib, Nintedanib, and Quercetin may be key drugs or compounds for the treatment of BC.
Collapse
Affiliation(s)
- Ping Li
- Department of Oncology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Xuexi Yang
- Department of Oncology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Qin Liu
- Department of Human Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Hanchao Zhang
- Department of Urology, The Affiliated Hospital and Clinical Medical College of Chengdu University, Chengdu, Sichuan, China
| | - Zhumei Luo
- Department of Oncology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China.
| |
Collapse
|
4
|
Schmitt A, Andrews N, Yasuda K, Hodge M, Ryznar R. Acute Stress and Autoimmune Markers: Evaluating the Psychoneuroimmunology Axis in Firefighter Recruits. Int J Mol Sci 2025; 26:3945. [PMID: 40362185 PMCID: PMC12071583 DOI: 10.3390/ijms26093945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/12/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Chronic psychological stress is known to influence immune function and contribute to development of autoimmune disorders through dysregulated inflammatory responses. This study investigates relationships between acute stress, life trauma, and autoimmune salivary biomarkers in firefighter recruits during psychophysical stress training. Salivary samples were collected from firefighter recruits during two stress tests to evaluate responses to acute stress. Samples were obtained at three time points-pre-stress, post-stress, and recovery-across both tests. Cortisol was measured to characterize acute stress response (ASR) profiles, while immune function was assessed through the analyzing C-reactive Protein (CRP), Complement C4 (C4), Pigment Epithelium Derived Factor (PEDF), and Serum Amyloid P (SAP). Results showed significant changes in CRP, C4, and PEDF after stress inoculation. Higher previous life trauma was associated with lower baseline cortisol (r = -0.489) and delay in cortisol recovery (r = 0.514), suggesting a learned biological response, potentially protective against stress-induced dysregulation. Cluster analysis revealed four distinct cortisol ASR profiles which were found to have significantly different past life trauma (p = 0.031). These findings suggest that trauma history influences stress biomarker dynamics, potentially reflecting individualized adaptive or maladaptive responses. The insights gained may inform strategies to enhance stress resilience and mitigate autoimmune risk among high-stress populations.
Collapse
Affiliation(s)
- Andrea Schmitt
- College of Osteopathic Medicine, Rocky Vista University, Englewood, CO 80112, USA; (A.S.); (N.A.); (K.Y.); (M.H.)
| | - Nathan Andrews
- College of Osteopathic Medicine, Rocky Vista University, Englewood, CO 80112, USA; (A.S.); (N.A.); (K.Y.); (M.H.)
| | - Krista Yasuda
- College of Osteopathic Medicine, Rocky Vista University, Englewood, CO 80112, USA; (A.S.); (N.A.); (K.Y.); (M.H.)
| | - Mitchell Hodge
- College of Osteopathic Medicine, Rocky Vista University, Englewood, CO 80112, USA; (A.S.); (N.A.); (K.Y.); (M.H.)
| | - Rebecca Ryznar
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Englewood, CO 80112, USA
| |
Collapse
|
5
|
Martin FP, Poulain C, Mulier JH, Motos A, Gourain V, Ogan I, Montassier E, Launey Y, Lasocki S, Cinotti R, Dahyot Fizelier C, Ranzani O, Reyes LF, Martin-Loeches I, Derde L, Torres A, Cremer O, Roquilly A. Identification and validation of robust hospital-acquired pneumonia subphenotypes associated with all-cause mortality: a multi-cohort derivation and validation. Intensive Care Med 2025; 51:692-707. [PMID: 40261385 DOI: 10.1007/s00134-025-07884-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/25/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE Despite optimal antimicrobial therapy, the treatment failure rate of hospital-acquired pneumonia (HAP) routinely reaches 40% in critically ill patients. Subphenotypes have been identified within sepsis and acute respiratory distress syndrome with important predictive and possibly therapeutic implications. We derived prognosis subphenotypes for HAP and explored whether they were associated with biological markers and response to treatment. METHODS We separately analysed data from four cohorts of critically ill patients in France (PNEUMOCARE, n = 511, ATLANREA, n = 401), Netherlands (MARS, n = 1351) and Europe-South America (ENIRRI, n = 900) to investigate HAP heterogeneity using unsupervised clustering based on clinical and routine biological variables available at HAP diagnosis. Then, we developed a machine learning-based workflow to create a simplified classification model using discovery data sets. This model was validated by applying it to an independent replication data set from an international randomized clinical trial comparing linezolid and tedizolid for the treatment of HAP (VITAL, n = 726 patients). The primary outcome was the association of subphenotypes with 28-day all-cause mortality. Secondary analyses included subphenotype associations with treatment failure at test-of-cure, respiratory microbiome and cytokine profiles in the ATLANREA subgroup, and treatment response in the VITAL trial. RESULTS We tested twelve metrics and determined that a two-cluster model best fits all cohorts. HAP subphenotype 2 had greater disease severity, lower body temperature, and worse PaO2/FiO2 ratio than subphenotype 1 patients. Although the prevalence of subphenotype 2 ranged from 26.9 to 66.9% across the four derivation cohorts, the rates of 28-day mortality and treatment failure at test-of-cure were consistently higher to subphenotype 1 (p < 0.01 for all comparisons). Subphenotype 2 was associated with greater respiratory microbiome dysbiosis and higher levels of proinflammatory cytokines in the ATLANREA cohort, as well as with statistically significant tedizolid effect modification in the VITAL trial (Relative Risk of treatment failure with tedizolid = 1.52; 95% CI 1.12-2.06 in subphenotype 1 vs. = 0.98; 95% CI 0.7-1.38 in subphenotype 2). CONCLUSIONS We identified two robust clinical subphenotypes by extensively analyzing HAP data sets. Their associations with respiratory microbiome composition, systemic inflammation, and treatment efficacy in independent data sets highlight their potential for prognostic value and predictive enrichment in future clinical trials aimed at personalized therapies.
Collapse
Affiliation(s)
- Florian Pierre Martin
- Inserm, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes Université, Nantes, France
| | - Cécile Poulain
- Inserm, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes Université, Nantes, France
- Service d'Anesthesie Réanimation, Institut de Recherche en Santé 2 Nantes Biotech, Nantes Université, CHU Nantes, Nantes, France
| | - Jelle Haitsma Mulier
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- Julius Centre for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ana Motos
- Inserm, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes Université, Nantes, France
- Servei de Pneumologia Hospital Clinic Fundació Clinic IDIBAPS, ICREA, CIBERES, Universitat de Barcelona, Barcelona, Spain
| | - Victor Gourain
- Inserm, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes Université, Nantes, France
| | - Ismaël Ogan
- Service d'Anesthesie Réanimation, Institut de Recherche en Santé 2 Nantes Biotech, Nantes Université, CHU Nantes, Nantes, France
| | - Emmanuel Montassier
- Inserm, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes Université, Nantes, France
- Service des Urgences, Nantes Université, CHU Nantes, Nantes, France
| | | | | | - Raphaël Cinotti
- Service d'Anesthesie Réanimation, Institut de Recherche en Santé 2 Nantes Biotech, Nantes Université, CHU Nantes, Nantes, France
- CHU Tours, INSERM, Methods in Patients-Centered Outcomes and HEalth Research, SPHERE, Nantes Université, Univ Tours, CHU Nantes, Nantes, France
| | | | - Otavio Ranzani
- Institut de Recerca Sant Pau (IR SANTPAU), Barcelona, Spain
- ISGlobal, Barcelona, Spain
| | - Luis Felipe Reyes
- Unisabana Center for Translational Science, School of Medicine, Universidad de La Sabana, Chia, Colombia
- Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | - Ignacio Martin-Loeches
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St. James' Hospital, Dublin, Ireland
| | - Lennie Derde
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- Julius Centre for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Antoni Torres
- Servei de Pneumologia Hospital Clinic Fundació Clinic IDIBAPS, ICREA, CIBERES, Universitat de Barcelona, Barcelona, Spain
| | - Olaf Cremer
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Antoine Roquilly
- Inserm, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes Université, Nantes, France.
- Service d'Anesthesie Réanimation, Institut de Recherche en Santé 2 Nantes Biotech, Nantes Université, CHU Nantes, Nantes, France.
| |
Collapse
|
6
|
Gorini F, Tonacci A, Sanmartin C, Venturi F. Phthalates and Non-Phthalate Plasticizers and Thyroid Dysfunction: Current Evidence and Novel Strategies to Reduce Their Spread in Food Industry and Environment. TOXICS 2025; 13:222. [PMID: 40137549 PMCID: PMC11945544 DOI: 10.3390/toxics13030222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Thyroid hormones (THs) play a crucial role in various biological functions, including metabolism, cell growth, and nervous system development, and any alteration involving the structure of the thyroid gland and TH secretion may result in thyroid disease. Growing evidence suggests that phthalate plasticizers, which are commonly used in a wide range of products (e.g., food packaging materials, children's toys, cosmetics, medical devices), can impact thyroid function, primarily affecting serum levels of THs and TH-related gene expression. Like phthalate compounds, recently introduced alternative plasticizers can leach from their source material into the environment, particularly into foods, although so far only a very limited number of studies have investigated their thyroid toxicity. This review aimed at summarizing the current knowledge on the role of phthalate and non-phthalate plasticizers in thyroid dysfunction and disease, describing the major biological mechanisms underlying this relationship. We will also focus on the food industry as one of the main players for the massive spread of such compounds in the human body, in turn conveyed by edible compounds. Given the increasing worldwide use of plasticizers and the essential role of THs in humans, novel strategies should be envisaged to reduce this burden on the thyroid and, in general, on human health.
Collapse
Affiliation(s)
- Francesca Gorini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy;
| | - Alessandro Tonacci
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy;
| | - Chiara Sanmartin
- Department of Agriculture, Food and Environment, University of Pisa, 56124 Pisa, Italy; (C.S.); (F.V.)
| | - Francesca Venturi
- Department of Agriculture, Food and Environment, University of Pisa, 56124 Pisa, Italy; (C.S.); (F.V.)
| |
Collapse
|
7
|
Chen Z, Behrendt R, Wild L, Schlee M, Bode C. Cytosolic nucleic acid sensing as driver of critical illness: mechanisms and advances in therapy. Signal Transduct Target Ther 2025; 10:90. [PMID: 40102400 PMCID: PMC11920230 DOI: 10.1038/s41392-025-02174-2] [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: 06/20/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Nucleic acids from both self- and non-self-sources act as vital danger signals that trigger immune responses. Critical illnesses such as acute respiratory distress syndrome, sepsis, trauma and ischemia lead to the aberrant cytosolic accumulation and massive release of nucleic acids that are detected by antiviral innate immune receptors in the endosome or cytosol. Activation of receptors for deoxyribonucleic acids and ribonucleic acids triggers inflammation, a major contributor to morbidity and mortality in critically ill patients. In the past decade, there has been growing recognition of the therapeutic potential of targeting nucleic acid sensing in critical care. This review summarizes current knowledge of nucleic acid sensing in acute respiratory distress syndrome, sepsis, trauma and ischemia. Given the extensive research on nucleic acid sensing in common pathological conditions like cancer, autoimmune disorders, metabolic disorders and aging, we provide a comprehensive summary of nucleic acid sensing beyond critical illness to offer insights that may inform its role in critical conditions. Additionally, we discuss potential therapeutic strategies that specifically target nucleic acid sensing. By examining nucleic acid sources, sensor activation and function, as well as the impact of regulating these pathways across various acute diseases, we highlight the driving role of nucleic acid sensing in critical illness.
Collapse
Affiliation(s)
- Zhaorong Chen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Rayk Behrendt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Lennart Wild
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
8
|
Baatarjav C, Komada T, Gunji Y, Komori S, Aizawa H, Nagi-Miura N, Karasawa T, Takahashi M. AIM2 targeting of nuclear DNA leakage in dendritic cells exacerbates vasculitis in a murine model of Kawasaki disease. Am J Physiol Heart Circ Physiol 2025; 328:H509-H517. [PMID: 39903175 DOI: 10.1152/ajpheart.00901.2024] [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: 12/30/2024] [Revised: 01/11/2025] [Accepted: 01/31/2025] [Indexed: 02/06/2025]
Abstract
Kawasaki disease (KD) is an acute vasculitis that mostly affects children and is characterized by inflammation of medium-sized arteries, particularly the coronary arteries. The absent in melanoma 2 (AIM2) inflammasome senses cytosolic dsDNA and regulates IL-1β-driven inflammation. We investigated the role of AIM2 in Candida albicans water-soluble fraction (CAWS)-induced vasculitis in a murine model mimicking KD. Aim2-/- mice exhibited reduced vasculitis, inflammatory cell infiltration, and vascular fibrosis in the aorta and coronary arteries. In addition, dsDNA damage was detected in Dectin-2+ cells infiltrating vasculitis areas. In vitro experiments showed that CAWS induced dsDNA damage in Dectin-2+ bone marrow-derived dendritic cells (BMDCs) isolated from wild-type (WT) and Aim2-/- mice. Furthermore, CAWS induces nuclear membrane deformation and DNA leakage into the cytosol, leading to AIM2 inflammasome activation and subsequent IL-1β production in WT BMDC. These findings suggest that AIM2 inflammasome activation in dendritic cells, triggered by dsDNA damage and leakage, contributes to the development of CAWS-induced vasculitis, and provides important insights into the inflammatory mechanisms underlying KD.NEW & NOTEWORTHY The AIM2 inflammasome in dendritic cells is a significant component of the murine model of Kawasaki disease-like vasculitis induced by CAWS injection. The AIM2 deficiency reduces vasculitis via reduced inflammatory cell infiltration and vascular fibrosis in CAWS-induced vasculitis. CAWS induces the damage and leakage of nuclear DNA in dendritic cells, which triggers AIM2 inflammasome activation, leading to an IL-1β-driven inflammatory response.
Collapse
Affiliation(s)
- Chintogtokh Baatarjav
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Takanori Komada
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Yoshitaka Gunji
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Satoko Komori
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Hidetoshi Aizawa
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Noriko Nagi-Miura
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Tadayoshi Karasawa
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
9
|
Suresh Kumar Meena Kumari M, Liu P, Nitchman MS, Chaudhary S, Jump K, Morales Y, Miller EA, Shecter I, Stadecker MJ, Kalantari P. NLRP3 and AIM2 inflammasomes exacerbate the pathogenic Th17 cell response to eggs of the helminth Schistosoma mansoni. PLoS Pathog 2025; 21:e1012108. [PMID: 40100932 PMCID: PMC11918320 DOI: 10.1371/journal.ppat.1012108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 02/09/2025] [Indexed: 03/20/2025] Open
Abstract
Infection with the helminth Schistosoma mansoni can cause exacerbated morbidity and mortality via a pathogenic host CD4 T cell-mediated immune response directed against parasite egg antigens, with T helper (Th) 17 cells playing a major role in the development of severe granulomatous hepatic immunopathology. The role of inflammasomes in intensifying disease has been reported; however, neither the types of caspases and inflammasomes involved, nor their impact on the Th17 response are known. Here we show that enhanced egg-induced IL-1β secretion and pyroptotic cell death required both caspase-1 and caspase-8 as well as NLRP3 and AIM2 inflammasome activation. Schistosome genomic DNA activated AIM2, whereas reactive oxygen species, potassium efflux and cathepsin B, were the major activators of NLRP3. NLRP3 and AIM2 deficiency led to a significant reduction in pathogenic Th17 responses, suggesting their crucial and non-redundant role in promoting inflammation. Additionally, we show that NLRP3- and AIM2-induced IL-1β suppressed IL-4 and protective Type I IFN (IFN-I) production, which further enhanced inflammation. IFN-I signaling also curbed inflammasome- mediated IL-1β production suggesting that these two antagonistic pathways shape the severity of disease. Lastly, Gasdermin D (Gsdmd) deficiency resulted in a marked decrease in egg-induced granulomatous inflammation. Our findings establish NLRP3/AIM2-Gsdmd axis as a central inducer of pathogenic Th17 responses which is counteracted by IFN-I pathway in schistosomiasis.
Collapse
Affiliation(s)
- Madhusoodhanan Suresh Kumar Meena Kumari
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Pengyu Liu
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Megan S. Nitchman
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Santoshi Chaudhary
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Kaile Jump
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Yoelkys Morales
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Emily A. Miller
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Ilana Shecter
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Miguel J. Stadecker
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Parisa Kalantari
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
10
|
Fusco L, Gazzi A, Giro L, Schefer RB, D'Almeida SM, Cagliani R, Zoccheddu M, Uyar R, Besbinar Ö, Çelik D, Yilmazer A, Mitrano DM, Orecchioni M, Delogu LG. Nanoplastics: Immune Impact, Detection, and Internalization after Human Blood Exposure by Single-Cell Mass Cytometry. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413413. [PMID: 39449193 PMCID: PMC11938024 DOI: 10.1002/adma.202413413] [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: 09/06/2024] [Revised: 10/07/2024] [Indexed: 10/26/2024]
Abstract
The increasing exposure to nanoplastics (NPs) raises significant concerns for human health, primarily due to their potential bioaccumulative properties. While NPs have recently been detected in human blood, their interactions with specific immune cell subtypes and their impact on immune regulation remain unclear. In this proof-of-concept study, model palladium-doped polystyrene NPs (PS-Pd NPs) are utilized to enable single-cell mass cytometry (CyTOF) detection. The size-dependent impact of carboxylate polystyrene NPs (50-200 nm) is investigated across 15 primary immune cell subpopulations using CyTOF. By taking advantage of Pd-doping for detecting PS-Pd NPs, this work evaluates their impact on human immune-cells at the single-cell level following blood exposure. This work traces PS-Pd NPs in 37 primary immune-cell subpopulations from human blood, quantifying the palladium atom count per cell by CyTOF while simultaneously assessing the impact of PS-Pd NPs on cell viability, functionality, and uptake. These results demonstrate that NPs can interact with, interfere with, and translocate into several immune cell subpopulations after exposure. In vivo distribution experiments in mice further confirmed their accumulation in immune cells within the liver, blood, and spleen, particularly in monocytes, macrophages, and dendritic cells. These findings provide valuable insights into the impact of NPs on human health.
Collapse
Affiliation(s)
- Laura Fusco
- ImmuneNano‐labDepartment of Biomedical SciencesUniversity of PaduaPadua35131Italy
| | - Arianna Gazzi
- ImmuneNano‐labDepartment of Biomedical SciencesUniversity of PaduaPadua35131Italy
| | - Linda Giro
- ImmuneNano‐labDepartment of Biomedical SciencesUniversity of PaduaPadua35131Italy
- Immunology Center of GeorgiaAugusta UniversityAugusta30912United States
| | - Roman B. Schefer
- Department of Environmental Systems ScienceETH ZürichZürich8092Switzerland
| | | | - Roberta Cagliani
- ImmuneNano‐labDepartment of Biomedical SciencesUniversity of PaduaPadua35131Italy
| | - Martina Zoccheddu
- Immunology Center of GeorgiaAugusta UniversityAugusta30912United States
- Georgia Cancer Center Integrated Genomics coreAugusta UniversityAugusta30912United States
| | - Recep Uyar
- Stem Cell InstituteAnkara UniversityAnkara06100Turkey
| | - Ömur Besbinar
- Stem Cell InstituteAnkara UniversityAnkara06100Turkey
| | | | - Acelya Yilmazer
- Stem Cell InstituteAnkara UniversityAnkara06100Turkey
- Department of Biomedical EngineeringFaculty of EngineeringAnkara UniversityAnkara06100Turkey
| | - Denise M. Mitrano
- Department of Environmental Systems ScienceETH ZürichZürich8092Switzerland
| | - Marco Orecchioni
- Immunology Center of GeorgiaAugusta UniversityAugusta30912United States
- Department of Pharmacology & ToxicologyAugusta UniversityAugusta30912United States
| | - Lucia Gemma Delogu
- ImmuneNano‐labDepartment of Biomedical SciencesUniversity of PaduaPadua35131Italy
- Department of Biological SciencesKhalifa University of Science & TechnologyAbu Dhabi127788UAE
| |
Collapse
|
11
|
Ding D, Liu H, Zhang L, Zhang G, Wei Y, Zhang W, Yang X, Li M, Yin G, Guo W, Chen X, Huang Z, Gao R. AIM2 promotes the progression of HNSCC via STAT1 mediated transcription and IL-17/MAPK signaling. Cell Signal 2025; 127:111545. [PMID: 39638137 DOI: 10.1016/j.cellsig.2024.111545] [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: 08/02/2024] [Revised: 11/10/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Chronic inflammation has been recognized as one of the hallmarks of head and neck squamous cell carcinoma (HNSCC), Absent In Melanoma 2(AIM2) has emerged as important regulators of chronic inflammatory, and participated in initiation, progression of kinds of human cancers. Nonetheless, the biological functions and underlying mechanisms of AIM2 in HNSCC remain inadequately understood. Based on the bioinformatics analysis of public databases, we identified elevated AIM2 expression in HNSCC, which positively correlates with disease stage and HPV infection, thereby possessing both diagnostic and prognostic significance. Immunohistochemistry on clinical samples revealed that AIM2 expression was frequently upregulated in cancerous tissues compared to paracancerous tissues, exhibiting a significant association with Ki-67 expression. Modulating AIM2 expression in HNSCC cell lines through transfection with inhibitors or mimics demonstrated that ectopic AIM2 expression enhances cell growth, migration, tumorigenesis, and metastasis both in vitro and in vivo. A dual luciferase reporter assay indicated that the transcription factor STAT1 can bind directly to the AIM2 promoter region and activate its transcription. The STAT1 inhibitor, fludarabine, reduces AIM2 expression and subsequently diminishes cell proliferation. Mechanistically, AIM2 exerts its tumor-promoting effects through the IL-17-MAPK signaling pathway. Collectively, our data demonstrate that AIM2, transcriptionally activated by STAT1, exhibits oncogenic functions by promoting the IL-17-MAPK signaling pathway, suggesting that AIM2 may be a new intervention targets for the diagnostic and treatment of HNSCC.
Collapse
Affiliation(s)
- Dong Ding
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, PR China; NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, PR China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences&Peking Union Medical College, Beijing 100021, PR China; Departments of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui Province 230001, PR China
| | - Hongfei Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, PR China
| | - Liping Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, PR China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences&Peking Union Medical College, Beijing 100021, PR China
| | - Guoxin Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, PR China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences&Peking Union Medical College, Beijing 100021, PR China
| | - Yumin Wei
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, PR China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences&Peking Union Medical College, Beijing 100021, PR China
| | - Wenlong Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, PR China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences&Peking Union Medical College, Beijing 100021, PR China
| | - Xingjiu Yang
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, PR China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences&Peking Union Medical College, Beijing 100021, PR China
| | - Mengyuan Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, PR China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences&Peking Union Medical College, Beijing 100021, PR China
| | - Gaofei Yin
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, PR China
| | - Wei Guo
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, PR China
| | - Xiaohong Chen
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, PR China.
| | - Zhigang Huang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, PR China.
| | - Ran Gao
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, PR China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences&Peking Union Medical College, Beijing 100021, PR China.
| |
Collapse
|
12
|
Soe YM, Sim SL, Kumari S. Innate Immune Sensors and Cell Death-Frontiers Coordinating Homeostasis, Immunity, and Inflammation in Skin. Viruses 2025; 17:241. [PMID: 40006996 PMCID: PMC11861910 DOI: 10.3390/v17020241] [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: 12/13/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
The skin provides a life-sustaining interface between the body and the external environment. A dynamic communication among immune and non-immune cells in the skin is essential to ensure body homeostasis. Dysregulated cellular communication can lead to the manifestation of inflammatory skin conditions. In this review, we will focus on the following two key frontiers in the skin: innate immune sensors and cell death, as well as their cellular crosstalk in the context of skin homeostasis and inflammation. This review will highlight the recent advancements and mechanisms of how these pathways integrate signals and orchestrate skin immunity, focusing on inflammatory skin diseases and skin infections in mice and humans.
Collapse
Affiliation(s)
| | | | - Snehlata Kumari
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Woolloongabba, Brisbane, QLD 4102, Australia; (Y.M.S.); (S.L.S.)
| |
Collapse
|
13
|
Colarusso C, Terlizzi M, Di Caprio S, Falanga A, D’Andria E, d’Emmanuele di Villa Bianca R, Sorrentino R. Role of the AIM2 Inflammasome in Cancer: Potential Therapeutic Strategies. Biomedicines 2025; 13:395. [PMID: 40002808 PMCID: PMC11852973 DOI: 10.3390/biomedicines13020395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Absent in melanoma 2 (AIM2) is a member of the innate immune sensors that recognizes cytosolic nucleic acids, leading to inflammasome assembly. In recent years, several studies in the oncology field have highlighted the presence of cytoplasmic double-stranded DNA (dsDNA) following necrosis and/or genomic instability, which is typical of malignant transformation. The recognition of dsDNA by the AIM2 inflammasome either in cancer cells or in immune cells can further exacerbate inflammatory processes on the basis of cancer progression. In this context, the role of AIM2 in cancer is still controversial in that some authors assume that AIM2 activation has pro-tumor activities, while others define it as anti-tumor. This discrepancy may be due to the nature of the cells where AIM2 is expressed or the histology of the tumor. This review aims to provide an overview of the controversial role of AIM2 in cancer, taking into consideration the pharmacological tools currently available to modulate AIM2 activity in cancer.
Collapse
Affiliation(s)
- Chiara Colarusso
- Department of Pharmacy (DIFARMA), University of Salerno, 84084 Fisciano, SA, Italy; (C.C.); (M.T.); (S.D.C.); (A.F.); (E.D.)
| | - Michela Terlizzi
- Department of Pharmacy (DIFARMA), University of Salerno, 84084 Fisciano, SA, Italy; (C.C.); (M.T.); (S.D.C.); (A.F.); (E.D.)
| | - Simone Di Caprio
- Department of Pharmacy (DIFARMA), University of Salerno, 84084 Fisciano, SA, Italy; (C.C.); (M.T.); (S.D.C.); (A.F.); (E.D.)
| | - Anna Falanga
- Department of Pharmacy (DIFARMA), University of Salerno, 84084 Fisciano, SA, Italy; (C.C.); (M.T.); (S.D.C.); (A.F.); (E.D.)
| | - Emmanuel D’Andria
- Department of Pharmacy (DIFARMA), University of Salerno, 84084 Fisciano, SA, Italy; (C.C.); (M.T.); (S.D.C.); (A.F.); (E.D.)
| | | | - Rosalinda Sorrentino
- Department of Pharmacy (DIFARMA), University of Salerno, 84084 Fisciano, SA, Italy; (C.C.); (M.T.); (S.D.C.); (A.F.); (E.D.)
| |
Collapse
|
14
|
Lu L, Shuang R, Cao F, Sun Z, Wei Q, Gao T, Gu X, Wen K, Cheng X, Gu M. Emodin Inhibits AIM2 Inflammasome Activation via Modulating K27-Linked Polyubiquitination to Attenuate Renal Fibrosis. Phytother Res 2025; 39:551-563. [PMID: 39558552 DOI: 10.1002/ptr.8390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/20/2024]
Abstract
Chronic kidney diseases (CKD) is a serious threat to people's health with renal fibrosis as the major pathological feature. The absent in melanoma 2 (AIM2) has recently been proposed to play a critical role in CKD. Emodin is a major bioactive compound from rhubarb, which is widely used for clinical treatment of renal disease. The aim of this study is to elucidate the effect of emodin on unilateral ureteral obstruction (UUO) model mice and its association with the AIM2 inflammasome. In this study, we established the UUO-induced mice renal interstitial fibrosis in vivo and bone marrow-derived macrophages (BMDMs) model in vitro. The BUN, SCr, TNF-α, IL-1β in serum were examined. The degree of renal damage and fibrosis were determined by histological assessment. Immunofluorescence, western blot, and Co-IP were used to determine the mechanisms of emodin against CKD. Emodin could improve UUO-induced abnormal renal function and histopathological abnormalities. It could also ameliorate renal fibrosis, evidenced by inhibiting the expression of α-SMA, TGF-β1, FN, and collagen I. Mechanistically, emodin significantly suppressed AIM2 inflammasome as well as its components including ASC, cleaved caspase-1, and IL-1β both in vivo and in vitro. Further studies demonstrated that emodin inhibited K27-linked polyubiquitination of AIM2 by targeting on K64 sites of the lysine residues. In summary, emodin could hinder the activation of AIM2 inflammasome in UUO model mice through K27-linked polyubiquitination to reduce renal fibrosis. Emodin is a possible therapeutic option for CKD treatment.
Collapse
Affiliation(s)
- Lidan Lu
- Department of Gynaecology, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Ruonan Shuang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Fang Cao
- Department of Nephrology, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Zhongwen Sun
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qingxue Wei
- Department of Nephrology, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Tiantian Gao
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuejing Gu
- Department of Nephrology, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Kejian Wen
- Department of Nephrology, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Xiaolan Cheng
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mingjia Gu
- Department of Nephrology, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| |
Collapse
|
15
|
Su Z, Lan J, Wang Y, Ma N, Yang J, Liang D, Zeng H, Yang M. Lactylation-driven ALKBH5 diminishes macrophage NLRP3 inflammasome activation in patients with G6PT deficiency. J Allergy Clin Immunol 2025:S0091-6749(25)00117-4. [PMID: 39900266 DOI: 10.1016/j.jaci.2025.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND Neutropenia represents an important clinical problem in patients with glycogen storage disease type Ib, characterized by genetic deficiency in glucose-6-phosphate translocase (G6PT/SLC37A4). However, the role of G6PT in macrophages has not been elucidated. OBJECTIVE We sought to investigate the function of G6PT in macrophage inflammation. METHODS Functional assays (including immunoblotting, real-time quantitative PCR, flow cytometry, immunofluorescence staining, and enzyme-linked immunosorbent assay) and RNA sequencing were performed. RESULTS We find that macrophages from patients deficient in G6PT exhibited diminished NLRP3 inflammasome activation. Mechanistically, deficiency of G6PT promotes glycolysis and lactate production in macrophages. Lactate accumulation potently induces ALKBH5 upregulation via H3K18 lactylation. ALKBH5 decreases m6A modification on NLRP3 messenger RNA, attenuating its transcript stability and thus inhibiting inflammasome activation. Further, treating G6PT-deficient macrophages with an inhibitor of the lactate dehydrogenase to lower their lactate levels restores NLRP3 inflammasome activation and rescues bacterial handling defect. CONCLUSION These findings reveal a previously unknown pathogenic mechanism of lactylation-driven defective NLRP3 inflammasome signaling and subsequent impaired antimicrobial activity as driving factors in these inflammatory disorders, indicating that glycolysis/lactate/histone lactylation cascade may be a potential therapeutic target for glycogen storage disease type Ib.
Collapse
Affiliation(s)
- Zexiong Su
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China; Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiaoli Lan
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ying Wang
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ni Ma
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jing Yang
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Danxia Liang
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hanshi Zeng
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Min Yang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China; Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
16
|
Shiri Aghbash P, Rasizadeh R, Sadri Nahand J, Bannazadeh Baghi H. The role of immune cells and inflammasomes in Modulating cytokine responses in HPV-Related cervical cancer. Int Immunopharmacol 2025; 145:113625. [PMID: 39637578 DOI: 10.1016/j.intimp.2024.113625] [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: 07/04/2024] [Revised: 10/26/2024] [Accepted: 11/10/2024] [Indexed: 12/07/2024]
Abstract
One of the most frequent cancers associated with gynecological malignancies is cervical cancer. Nearly 99% of cervical tumor lesions are produced by prolonged infection with hr-HPV and almost 70% of cases are related to HPV-16 and HPV-18. The human immune system has a crucial role in defending against infections caused by HPV infection. As an illustration, elevation in neutrophils reduces T cell antitumor activity, which in turn results in the development of malignancies and subsequently inhibits immune system function. HPV-infected cells, also, express a significant number of genes related to pro-inflammatory mediators including IL-1β. Moreover, inflammasomes, which are multi-protein complexes, owing the production of the pro-inflammatory cytokines including IL-1β and IL-18 in response to viral infections. In other words, these multi-protein complexes have a crucial role in tumor immunity regulation through the secretion of pro-inflammatory cytokines and induction of antigen presentation and maturation by APCs including dendritic cells. In this study, we attempted to investigate the inflammasome's general role in the initiation and advancement of cervical cancer, as well as a summary of the pathways connected to the possible participation of inflammasomes in the pathological process of cervical carcinoma and immune cell engagement. Novel strategy techniques that target the inflammatory reaction of tumor-related antigens may be created with an understanding of inflammasome-dependent pathways to accomplish tumor immunotherapy and cervical tumor treatment.
Collapse
Affiliation(s)
- Parisa Shiri Aghbash
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reyhaneh Rasizadeh
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
17
|
Hollis A, Lukens JR. Role of inflammasomes and neuroinflammation in epilepsy. Immunol Rev 2025; 329:e13421. [PMID: 39523682 PMCID: PMC11744240 DOI: 10.1111/imr.13421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Epilepsy is a brain disorder characterized by recurrent seizures, which are brief episodes of abnormal electrical activity in the brain and involuntary movement that can lead to physical injury and loss of consciousness. Seizures are canonically accompanied by increased inflammatory cytokine production that promotes neuroinflammation, brain pathology, and seizure propagation. Understanding the source of pro-inflammatory cytokines which promote seizure pathogenesis could be a gateway to precision epilepsy drug design. This review discusses the inflammasome in epilepsy including its role in seizure propagation and negative impacts on brain health. The inflammasome is a multiprotein complex that coordinates IL-1β and IL-18 production in response to tissue damage, cellular stress, and infection. Clinical evidence for inflammasome signaling in epileptogenesis is reviewed followed by a discussion of emerging strategies to modulate inflammasome activity in epilepsy.
Collapse
Affiliation(s)
- Ava Hollis
- Center for Brain Immunology and Glia (BIG), Department of NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - John R. Lukens
- Center for Brain Immunology and Glia (BIG), Department of NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
18
|
Weymouth L, Smith AR, Lunnon K. DNA Methylation in Alzheimer's Disease. Curr Top Behav Neurosci 2025; 69:149-178. [PMID: 39455499 DOI: 10.1007/7854_2024_530] [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: 10/28/2024]
Abstract
To date, DNA methylation is the best characterized epigenetic modification in Alzheimer's disease. Involving the addition of a methyl group to the fifth carbon of the cytosine pyrimidine base, DNA methylation is generally thought to be associated with the silencing of gene expression. It has been hypothesized that epigenetics may mediate the interaction between genes and the environment in the manifestation of Alzheimer's disease, and therefore studies investigating DNA methylation could elucidate novel disease mechanisms. This chapter comprehensively reviews epigenomic studies, undertaken in human brain tissue and purified brain cell types, focusing on global methylation levels, candidate genes, epigenome wide approaches, and recent meta-analyses. We discuss key differentially methylated genes and pathways that have been highlighted to date, with a discussion on how new technologies and the integration of multiomic data may further advance the field.
Collapse
Affiliation(s)
- Luke Weymouth
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Adam R Smith
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Katie Lunnon
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
| |
Collapse
|
19
|
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: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 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
|
20
|
Coll RC, Schroder K. Inflammasome components as new therapeutic targets in inflammatory disease. Nat Rev Immunol 2025; 25:22-41. [PMID: 39251813 DOI: 10.1038/s41577-024-01075-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2024] [Indexed: 09/11/2024]
Abstract
Inflammation drives pathology in many human diseases for which there are no disease-modifying drugs. Inflammasomes are signalling platforms that can induce pathological inflammation and tissue damage, having potential as an exciting new class of drug targets. Small-molecule inhibitors of the NLRP3 inflammasome that are now in clinical trials have demonstrated proof of concept that inflammasomes are druggable, and so drug development programmes are now focusing on other key inflammasome molecules. In this Review, we describe the potential of inflammasome components as candidate drug targets and the novel inflammasome inhibitors that are being developed. We discuss how the signalling biology of inflammasomes offers mechanistic insights for therapeutic targeting. We also discuss the major scientific and technical challenges associated with drugging these molecules during preclinical development and clinical trials.
Collapse
Affiliation(s)
- Rebecca C Coll
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK.
| | - Kate Schroder
- Institute for Molecular Bioscience (IMB), The University of Queensland, St Lucia, Queensland, Australia.
| |
Collapse
|
21
|
Trembath HE, LaBella ME, Kearney JF, Hariharan A, Zarmer S, Nabors M, McCabe I, Zhao RT, Meyers M, Kim HJ, Yeh JJ. New Onset Diabetes in Pancreatic Adenocarcinoma Does Not Correlate With Molecular Subtype. J Surg Oncol 2024. [PMID: 39711008 DOI: 10.1002/jso.28044] [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: 11/17/2024] [Accepted: 11/23/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND AND OBJECTIVES Studies show that new onset diabetes mellitus (DM) (NOD) predates the diagnosis of PDAC by up to 2 years. Two tumor-intrinsic molecular subtypes of PDAC that are prognostic and predictive of chemotherapy response have been described and validated. We hypothesize that patients with NOD may have different molecular subtypes and prognoses. METHODS This is a single-institution study of patients who underwent resection for PDAC from 2009 to 2022 with de-identified samples available for sequencing. Demographic and clinical factors were examined using bivariate and multivariate analysis. RESULTS A total of 97 patients met inclusion criteria: 70 with no history of DM, 11 with longstanding DM (> 2 years), and 16 with NOD. The demographics between groups were overall similar. After controlling for age, sex, race, BMI, and tobacco history, NOD was not a significant predictor of PDAC subtype. There were no survival differences between groups. Transcriptomic analysis suggests the upregulation of inflammatory and immune activation and regulation pathways in NOD. CONCLUSIONS As continued interest in NOD and PDAC mounts, we are the first to examine if NOD may be associated with molecular subtypes and outcomes. Further investigation into the underlying pathophysiology of the NOD group is still needed.
Collapse
Affiliation(s)
- Hannah E Trembath
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michelle E LaBella
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joseph F Kearney
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Arthi Hariharan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sandra Zarmer
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mariaelena Nabors
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ian McCabe
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ryan T Zhao
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael Meyers
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Hong Jin Kim
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jen Jen Yeh
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
22
|
Arrè V, Negro R, Giannelli G. The role of inflammasomes in hepatocellular carcinoma: Mechanisms and therapeutic insights. Ann Hepatol 2024; 30:101772. [PMID: 39701280 DOI: 10.1016/j.aohep.2024.101772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024]
Abstract
Hepatocellular carcinoma is among the most frequent forms of primary liver cancer and develops within a context of chronic inflammation, frequently associated with a multitude of risk factors, including viral infections, metabolic dysfunction-associated fatty liver disease, metabolic dysfunction-associated steatohepatitis and liver fibrosis. The tumor microenvironment is crucial for the progression of HCC, as immune cells, tumor-associated fibroblasts and hepatic stellate cells interact to promote chronic inflammation and tumor spread. Inflammasomes, the multiprotein complexes that launch the innate immune response, emerge as important mediators in the pathogenesis of HCC. Among others, the inflammasome Nucleotide-binding oligomerization domain, Leucine rich Repeat (NLR) and Pyrin (NLRP) 3 (NLRP3), and absent in melanoma 2 (AIM2), exhibit a dual role in HCC background. It has been reported that they can exert oncosuppressive functions by triggering the inflammatory death of cancer cells. Vice versa, chronic activation contributes to the development of a pro-tumorigenic environment, thus supporting tumor growth. In addition, other inflammasomes such as Nucleotide-binding oligomerization domain, Leucine rich Repeat (NLR) and Pyrin (NLRP) 6 and 12 (NLRP6 and NLRP12, respectively) regulate HCC onset and progression, although more experimental evidence is required. This review focuses on the molecular mechanisms underpinning the inflammasome's contribution to the onset, progression and spread of HCC. Moreover, we will explore the potential therapeutic approaches currently under investigation, which aim to improve the efficacy and reduce the side effects of the treatments currently available. Targeting inflammasomes may be a promising therapeutic strategy for the treatment of HCC, offering new opportunities to improve patient prognosis.
Collapse
Affiliation(s)
- Valentina Arrè
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy.
| | - Roberto Negro
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy.
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, "S. de Bellis", IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy.
| |
Collapse
|
23
|
Sun R, Chu J, Li P. Inflammasomes and idiopathic inflammatory myopathies. Front Immunol 2024; 15:1449969. [PMID: 39723212 PMCID: PMC11668653 DOI: 10.3389/fimmu.2024.1449969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/15/2024] [Indexed: 12/28/2024] Open
Abstract
Idiopathic inflammatory myopathies (IIM) are a group of systemic autoimmune diseases characterized by muscle weakness and elevated serum creatine kinase levels. Recent research has highlighted the role of the innate immune system, particularly inflammasomes, in the pathogenesis of IIM. This review focuses on the role of inflammasomes, specifically NLRP3 and AIM2, and their associated proteins in the development of IIM. We discuss the molecular mechanisms of pyroptosis, a programmed cell death pathway that triggers inflammation, and its association with IIM. The NLRP3 inflammasome, in particular, has been implicated in muscle fiber necrosis and the subsequent release of damage-associated molecular patterns (DAMPs), leading to inflammation. We also explore the potential therapeutic implications of targeting the NLRP3 inflammasome with inhibitors such as glyburide and MCC950, which have shown promise in reducing inflammation and improving muscle function in preclinical models. Additionally, we discuss the role of caspases, particularly caspase-1, in the canonical pyroptotic pathway associated with IIM. The understanding of these mechanisms offers new avenues for therapeutic intervention and a better comprehension of IIM pathophysiology.
Collapse
Affiliation(s)
- Rui Sun
- Department of Rheumatology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Jiyan Chu
- Department of Rheumatology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
- Graduate School, Dalian Medical University, Dalian, Liaoning, China
| | - Ping Li
- Department of Rheumatology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| |
Collapse
|
24
|
Scalavino V, Piccinno E, Giannelli G, Serino G. Inflammasomes in Intestinal Disease: Mechanisms of Activation and Therapeutic Strategies. Int J Mol Sci 2024; 25:13058. [PMID: 39684769 DOI: 10.3390/ijms252313058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
NOD-like receptors (NLRs) are a family of cytosolic pattern recognition receptors (PRRs) implicated in the innate immune sensing of pathogens and damage signals. NLRs act as sensors in multi-protein complexes called inflammasomes. Inflammasome activity is necessary for the maintenance of intestinal homeostasis, although their aberrant activation contributes to the pathogenesis of several gastrointestinal diseases. In this review, we summarize the main features of the predominant types of inflammasomes involved in gastrointestinal immune responses and their implications in intestinal disease, including Irritable Bowel Syndrome (IBS), Inflammatory Bowel Disease (IBD), celiac disease, and Colorectal Cancer (CRC). In addition, we report therapeutic discoveries that target the inflammasome pathway, highlighting promising novel therapeutic strategies in the treatment of intestinal diseases. Collectively, our understanding of the mechanisms of intestinal inflammasome activation and their interactions with other immune pathways appear to be not fully elucidated. Moreover, the clinical relevance of the efficacy of inflammasome inhibitors has not been evaluated. Despite these limitations, a greater understanding of the effectiveness, specificity, and reliability of pharmacological and natural inhibitors that target inflammasome components could be an opportunity to develop new therapeutic options for the treatment of intestinal disease.
Collapse
Affiliation(s)
- Viviana Scalavino
- National Institute of Gastroenterology S. De Bellis, IRCCS Research Hospital, Via Turi 27, 70013 Castellana Grotte, BA, Italy
| | - Emanuele Piccinno
- National Institute of Gastroenterology S. De Bellis, IRCCS Research Hospital, Via Turi 27, 70013 Castellana Grotte, BA, Italy
| | - Gianluigi Giannelli
- National Institute of Gastroenterology S. De Bellis, IRCCS Research Hospital, Via Turi 27, 70013 Castellana Grotte, BA, Italy
| | - Grazia Serino
- National Institute of Gastroenterology S. De Bellis, IRCCS Research Hospital, Via Turi 27, 70013 Castellana Grotte, BA, Italy
| |
Collapse
|
25
|
Wei S, Wu L, Xiang Z, Yang X, Pei D, Jiang L, Du Z. EIF2AK2 protein targeted activation of AIM2-mediated PANoptosis promotes sepsis-induced acute kidney injury. Ren Fail 2024; 46:2403649. [PMID: 39311631 PMCID: PMC11421145 DOI: 10.1080/0886022x.2024.2403649] [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: 04/08/2024] [Revised: 08/22/2024] [Accepted: 09/07/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Acute kidney injury (AKI) frequently occurs as a complication of sepsis. PANoptosis refers to a type of inflammatory programmed cell death that exhibits key characteristics of apoptosis, necroptosis, and pyroptosis. Here, we evaluated the role of absent in melanoma 2 (AIM2) and eukaryotic translation initiation factor 2 alpha kinase 2 (EIF2AK2) in septic AKI. METHODS A septic AKI model was created through cecal ligation and puncture (CLP), while an in vitro model was developed using lipopolysaccharide (LPS)-stimulated HK2 cells. Hematoxylin and eosin (HE), Periodic acid-Schiff (PAS), and TUNEL staining were conducted to assess kidney injury in mice. Levels of serum creatinine (Scr) and blood urea nitrogen (BUN) were detected by kits. Gene expression was detected utilizing RT-qPCR, and Western blot was used to test protein levels. Immunofluorescence was employed to measure EIF2AK2 and AIM2 expression in mouse kidney tissue. Lactate dehydrogenase (LDH) activity assay was conducted to evaluate cytotoxicity. Co-immunoprecipitation (Co-IP) was performed to verify the binding relationship between EIF2AK2 and AIM2. RESULTS AIM2 expression was increased in the renal tissue of mice subjected to CLP. Activation of the inflammasome and PANoptosis were observed in the renal tissue of CLP mice. AIM2 depletion attenuated PANoptosis in LPS-treated HK-2 cells. Additionally, EIF2AK2 could directly target AIM2, leading to a positive regulation of AIM2 expression. Notably, EIF2AK2 induced PANoptosis through upregulating AIM2 in HK-2 cells stimulated by LPS. CONCLUSIONS Our results revealed the important role of EIF2AK2-induced AIM2 upregulation in the activation of PANoptosis during septic AKI.
Collapse
Affiliation(s)
- Siwei Wei
- Department of Anesthesiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha City, China
| | - Lei Wu
- Department of Anesthesiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha City, China
| | - Zhen Xiang
- Department of Anesthesiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha City, China
| | - Xiaoxiao Yang
- Department of Anesthesiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha City, China
| | - Dongjie Pei
- Department of Anesthesiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha City, China
| | - Liubing Jiang
- Department of Anesthesiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha City, China
| | - Zhen Du
- Department of Anesthesiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha City, China
| |
Collapse
|
26
|
Gu F, Wang Z, Ding H, Tao X, Zhang J, Dai K, Li X, Shen H, Li H, Chen Z, Wang Z. Microglial mitochondrial DNA release contributes to neuroinflammation after intracerebral hemorrhage through activating AIM2 inflammasome. Exp Neurol 2024; 382:114950. [PMID: 39278588 DOI: 10.1016/j.expneurol.2024.114950] [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: 07/01/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/18/2024]
Abstract
Intracerebral hemorrhage (ICH) is a severe disease that often leads to disability and death. Neuroinflammatory response is a key causative factor of early secondary brain injury after ICH. AIM2 is a DNA-sensing protein that recognizes cytosolic double-stranded DNA and take a significant part in neuroinflammation. Mitochondrial DNA participates in the translation of proteins such as the respiratory chain in the mitochondria. Whether mtDNA is involved in forming AIM2 inflammasome after ICH remains unclear. We used mice to construct ICH model in vivo and we used BV2 microglial cells treated with oxyhemoglobin to simulate ICH in vitro. Following lentiviral transfection to overexpress AIM2 antagonist P202, a notable decrease was observed in the levels of AIM2 inflammasome-associated proteins, leading to a reduction in dead neurons surrounding the hematoma and an enhancement in long-term and short-term behavior of neurological deficits. We further explored whether mtDNA took part in the AIM2 activation after ICH. The cytosolic mtDNA level was down-regulated by the mitochondrial division protector Mdivi-1 and up-regulated by transfection of mtDNA into cytoplasm. We found the expression level of AIM2 inflammasome-related proteins and inflammatory cytokines release were regulated by the cytosolic mtDNA level. In conclusion, after ICH, the mtDNA content in the cytoplasm of microglia around the hematoma rises, causing AIM2 inflammation leading to neuronal apoptosis, which leads to neurological deficits in mice. On the other hand, P202 was able to block inflammatory vesicle activation and improve neurological function by preventing the interaction between AIM2 protein and mitochondrial DNA.
Collapse
Affiliation(s)
- Feng Gu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Zongqi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haojie Ding
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xinyu Tao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Juyi Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Kun Dai
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Zhouqing Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| |
Collapse
|
27
|
Anderson MJ, den Hartigh AB, Loomis WP, Fink SL. Broad-spectrum inflammasome inhibition by thiomuscimol. Cell Death Discov 2024; 10:470. [PMID: 39550359 PMCID: PMC11569204 DOI: 10.1038/s41420-024-02238-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024] Open
Abstract
Inflammasome formation, arising from pathogen or internal activating signals, is a key step in canonical pyroptosis, a gasdermin-mediated inflammatory cell death. Inhibition of pyroptosis has great clinical relevance due to its involvement in many different disease states. Current inhibitors of pyroptosis either only inhibit the final lytic step, which still allows inflammatory signal release, or only inhibit a single inflammasome, which does not account for inherent redundancy in activation of other inflammatory pathways. Here, we show that thiomuscimol, a structural analog of the lysis inhibitor muscimol, exhibits unique inhibitory activity upstream of plasma membrane rupture. We find that thiomuscimol inhibits inflammasome formation, as well as downstream caspase-1 activation, initiated by multiple pyroptotic signals, regardless of whether NLR recruitment of caspase-1 to the inflammasome relies on the ASC adapter protein. The ability of thiomuscimol to block multiple different inflammasomes opens the door for development of therapeutics with increased applications to broadly inhibit pyroptosis in multiple pathological settings.
Collapse
Affiliation(s)
- Marisa J Anderson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Andreas B den Hartigh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Wendy P Loomis
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Susan L Fink
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
28
|
Tang C, Jia F, Wu M, Wang Y, Lu X, Li J, Ding Y, Chen W, Chen X, Han F, Xu H. Elastase-targeting biomimic nanoplatform for neurovascular remodeling by inhibiting NETosis mediated AlM2 inflammasome activation in ischemic stroke. J Control Release 2024; 375:404-421. [PMID: 39288890 DOI: 10.1016/j.jconrel.2024.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
Neutrophil elastase (NE) is a protease released by activated neutrophils in the brain parenchyma after cerebral ischemia, which plays a pivotal role in the regulation of neutrophil extracellular traps (NETs) formation. The excess NETs could lead to blood-brain barrier (BBB) breakdown, overwhelming neuroinflammation, and neuronal injury. While the potential of targeting neutrophils and inhibiting NE activity to mitigate ischemic stroke (IS) pathology has been recognized, effective strategies that inhibit NETs formation remain under-explored. Herein, a biomimic multifunctional nanoplatform (HM@ST/TeTeLipos) was developed for active NE targeting and IS treatment. The core of the HM@ST/TeTeLipos consisted of sivelestat-loaded ditelluride-containing liposomes with ROS-responsive and NE-inhibiting properties. The outer shell was composed of platelet-neutrophil hybrid membrane vesicles (HMVs), which acted to hijack neutrophils and neutralize proinflammatory cytokines. Our studies revealed that HM@ST/TeTeLipos could effectively inhibit NE activity, thereby suppressing the release of NETs, impeding the activation of the AIM2 inflammasome, and consequently redirecting the immune response away from a pro-inflammatory M1 microglia phenotype. This resulted in enhanced neurovascular remodeling, reduced BBB disruption, and diminished neuroinflammation, ultimately promoting neuron survival. We believe that this innovative approach holds significant potential for improving the treatment of IS and various NE-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Chunming Tang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Feng Jia
- Department of Neurosurgery, Yancheng NO.1 People's Hospital, The Affiliated Yancheng First Hospital of Nanjing University Medical School, 224008, China
| | - Min Wu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yanling Wang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiaowei Lu
- Department of Geriatric Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jinyu Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yan Ding
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Weilin Chen
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Xufeng Chen
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Feng Han
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Huae Xu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
29
|
Xu T, Zhong X, Luo N, Ma W, Hao P. Review of Excessive Cytosolic DNA and Its Role in AIM2 and cGAS-STING Mediated Psoriasis Development. Clin Cosmet Investig Dermatol 2024; 17:2345-2357. [PMID: 39464745 PMCID: PMC11512523 DOI: 10.2147/ccid.s476785] [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: 05/27/2024] [Accepted: 10/07/2024] [Indexed: 10/29/2024]
Abstract
In psoriasis, keratinocytes are triggered by factors, such as infection or tissue damage, to release DNA, which thereby activates plasmacytoid dendritic cells and macrophages to induce inflammation, thickened epidermis, and parakeratosis. The recognition of double-stranded (ds)DNA facilitates the activation of cytoplasmic DNA sensors absent in melanoma 2 (AIM2) inflammasome assembly and cyclic guanosine monophosphate adenosine monophosphate (cGAMP) synthase (cGAS) - stimulator of interferon gene (STING) pathway, both of which play a pivotal role in mediating the inflammatory response and driving the progression of psoriasis. Additionally, secreted proinflammatory cytokines can stimulate further DNA release from keratinocytes. Notably, the activation of AIM2 and cGAS-STING signaling pathways also mediates programmed cell death, potentially enhancing DNA overproduction. As a result, excessive DNA can activate these pathways, amplifying persistent inflammatory responses that contribute to the maintenance of psoriasis. Several studies have validated that targeting DNA and its mediated activation of AIM2 and cGAS-STING offers promising therapeutic strategies for psoriasis. Here, we postulate a hypothesis that excessive cytosolic DNA can activate AIM2 and cGAS-STING, mediating inflammation and programmed cell death, ultimately fostering DNA accumulation and contributing to the development of psoriasis.
Collapse
Affiliation(s)
- Tongtong Xu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xiaojing Zhong
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Nana Luo
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Wenyi Ma
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Pingsheng Hao
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| |
Collapse
|
30
|
Campbell C, Mayatra JM, Neve AJ, Fletcher JM, Johnston DGW. Inflammasomes: emerging therapeutic targets in hidradenitis suppurativa? Br J Dermatol 2024; 191:670-679. [PMID: 38913409 DOI: 10.1093/bjd/ljae262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/22/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
Hidradenitis suppurativa (HS) is a chronic inflammatory skin disease characterized by recurrent inflammatory lesions, which affect skin and hair follicles in intertriginous areas. HS has a multifactorial aetiology resulting in barrier dysfunction associated with aberrant immune activation. There is increased evidence for the role of inflammasomes in the pathophysiology of inflammatory skin diseases, including HS. Inflammasomes are multiprotein complexes activated following exposure to danger signals, including microbial ligands and components of damaged host cells. Inflammasome activation induces many signalling cascades and subsequent cleavage of proinflammatory cytokines - most notably interleukin (IL)-1β - which have a role in HS pathogenesis. Limited immunotherapies are approved for treating moderate-to-severe HS, with variable response rates influenced by disease heterogeneity. Inflammasomes represent attractive targets to suppress multiple inflammatory pathways in HS, including IL-1β and IL-17. This review aims to summarize the role of inflammasomes in HS and to evaluate evidence for inflammasomes as therapeutic targets for HS treatment.
Collapse
Affiliation(s)
- Ciara Campbell
- Discipline of Anatomy, Trinity College Dublin, Dublin, Ireland
- Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin
| | - Jay M Mayatra
- Discipline of Anatomy, Trinity College Dublin, Dublin, Ireland
| | - Ashish J Neve
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Jean M Fletcher
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin
| | - Daniel G W Johnston
- Discipline of Anatomy, Trinity College Dublin, Dublin, Ireland
- Charles Institute of Dermatology, University College Dublin, Dublin, Ireland
| |
Collapse
|
31
|
Martinez JC, Morandini F, Fitzgibbons L, Sieczkiewicz N, Bae SJ, Meadow ME, Hillpot E, Cutting J, Paige V, Biashad SA, Simon M, Sedivy J, Seluanov A, Gorbunova V. cGAS deficient mice display premature aging associated with de-repression of LINE1 elements and inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617645. [PMID: 39416083 PMCID: PMC11482887 DOI: 10.1101/2024.10.10.617645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Aging-associated inflammation, or 'inflammaging" is a driver of multiple age-associated diseases. Cyclic GMP-AMP Synthase (cGAS) is a cytosolic DNA sensor that functions to activate interferon response upon detecting viral DNA in the cytoplasm. cGAS contributes to inflammaging by responding to endogenous signals such as damaged DNA or LINE1 (L1) cDNA which forms in aged cells. While cGAS knockout mice are viable their aging has not been examined. Unexpectedly, we found that cGAS knockout mice exhibit accelerated aging phenotype associated with induction of inflammation. Transcription of L1 elements was increased in both cGAS knockout mice and in cGAS siRNA knockdown cells associated with high levels of cytoplasmic L1 DNA and expression of ORF1 protein. Cells from cGAS knockout mice showed increased chromatin accessibility and decreased DNA methylation on L1 transposons. Stimulated emission depletion microscopy (STED) showed that cGAS forms nuclear condensates that co-localize with H3K9me3 heterochromatin marks, and H3K9me3 pattern is disrupted in cGAS knockout cells. Taken together these results suggest a previously undescribed role for cGAS in maintaining heterochromatin on transposable elements. We propose that loss of cGAS leads to loss of chromatin organization, de-repression of transposable elements and induction of inflammation resulting in accelerated aging.
Collapse
Affiliation(s)
- John C Martinez
- Translational Biomedical Sciences Program, University of Rochester, NY, 14627, USA
- Department of Biology, University of Rochester, NY, 14627, USA
| | | | | | | | - Sung Jae Bae
- Department of Biology, University of Rochester, NY, 14627, USA
| | | | - Eric Hillpot
- Department of Biology, University of Rochester, NY, 14627, USA
| | - Joseph Cutting
- Department of Biology, University of Rochester, NY, 14627, USA
| | - Victoria Paige
- Department of Biology, University of Rochester, NY, 14627, USA
| | | | - Matthew Simon
- Department of Biology, University of Rochester, NY, 14627, USA
| | - John Sedivy
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, RI, 02912, USA
| | - Andrei Seluanov
- Department of Biology, University of Rochester, NY, 14627, USA
- Department of Medicine, University of Rochester, NY, 14627, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, NY, 14627, USA
- Department of Medicine, University of Rochester, NY, 14627, USA
| |
Collapse
|
32
|
Xu C, Jing W, Liu C, Yuan B, Zhang X, Liu L, Zhang F, Chen P, Liu Q, Wang H, Du X. Cytoplasmic DNA and AIM2 inflammasome in RA: where they come from and where they go? Front Immunol 2024; 15:1343325. [PMID: 39450183 PMCID: PMC11499118 DOI: 10.3389/fimmu.2024.1343325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Rheumatoid arthritis is a chronic autoimmune disease of undetermined etiology characterized by symmetric synovitis with predominantly destructive and multiple joint inflammation. Cytoplasmic DNA sensors that recognize protein molecules that are not themselves or abnormal dsDNA fragments play an integral role in the generation and perpetuation of autoimmune diseases by activating different signaling pathways and triggering innate immune signaling pathways and host defenses. Among them, melanoma deficiency factor 2 (AIM2) recognizes damaged DNA and double-stranded DNA and binds to them to further assemble inflammasome, initiating the innate immune response and participating in the pathophysiological process of rheumatoid arthritis. In this article, we review the research progress on the source of cytoplasmic DNA, the mechanism of assembly and activation of AIM2 inflammasome, and the related roles of other cytoplasmic DNA sensors in rheumatoid arthritis.
Collapse
Affiliation(s)
- Conghui Xu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Cui Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Bo Yuan
- Department of Acupuncture and Pain, Affiliated Hospital of Gansu University of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Xinghua Zhang
- Department of Acupuncture, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Limei Liu
- Department of Zheng's Acupuncture, Affiliated Hospital of Gansu University of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Fengfan Zhang
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Ping Chen
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Qiang Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Haidong Wang
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Xiaozheng Du
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
33
|
Varela Martins T, Silva de Melo BM, Toller-Kawahisa JE, da Silva GVL, Aníbal Silva CE, Paiva IM, Públio GA, Rosa MH, da Silva Souza C, Zamboni DS, Cunha FQ, Cunha TM, Ryffel B, Riteau N, Alves-Filho JC. The DNA sensor AIM2 mediates psoriasiform inflammation by inducing type 3 immunity. JCI Insight 2024; 9:e171894. [PMID: 39352743 PMCID: PMC11601563 DOI: 10.1172/jci.insight.171894] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/18/2024] [Indexed: 11/09/2024] Open
Abstract
Psoriasis is a chronic and recurrent inflammatory skin disease characterized by abnormal proliferation and differentiation of keratinocytes and activation of immune cells. However, the molecular driver that triggers this immune response in psoriatic skin remains unclear. The inflammation-related gene absent in melanoma 2 (AIM2) was identified as a susceptibility gene/locus associated with psoriasis. In this study, we investigated the role of AIM2 in the pathophysiology of psoriasis. We found elevated levels of mitochondrial DNA in patients with psoriasis, along with high expression of AIM2 in both the human psoriatic epidermis and a mouse model of psoriasis induced by topical imiquimod (IMQ) application. Genetic ablation of AIM2 reduced the development of IMQ-induced psoriasis by decreasing the production of type 3 cytokines (such as IL-17A and IL-23) and infiltration of immune cells into the inflammatory site. Furthermore, we demonstrate that IL-17A induced AIM2 expression in keratinocytes. Finally, the genetic absence of inflammasome components downstream AIM2, ASC, and caspase-1 alleviated IMQ-induced skin inflammation. Collectively, our data show that AIM2 is involved in developing psoriasis through its canonical activation.
Collapse
Affiliation(s)
- Timna Varela Martins
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
- Immune Health Laboratory, Regulation of host responses and immune health, IRL2029, French National Centre for Scientific Research (CNRS) and Ribeirão Preto Medical School (FMRP) of the Sao Paulo University (USP), Sao Paulo, Brazil
- INEM, CNRS, UMR7355 and University, Orleans, France
| | - Bruno Marcel Silva de Melo
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Juliana Escher Toller-Kawahisa
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Gabriel Victor Lucena da Silva
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Conceição Elidianne Aníbal Silva
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Isadora Marques Paiva
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Gabriel Azevedo Públio
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Marcos Henrique Rosa
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | | | - Dario Simões Zamboni
- Department of Cell Biology, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Fernando Q. Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | | | - Nicolas Riteau
- Immune Health Laboratory, Regulation of host responses and immune health, IRL2029, French National Centre for Scientific Research (CNRS) and Ribeirão Preto Medical School (FMRP) of the Sao Paulo University (USP), Sao Paulo, Brazil
- INEM, CNRS, UMR7355 and University, Orleans, France
| | - José C. Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
- Immune Health Laboratory, Regulation of host responses and immune health, IRL2029, French National Centre for Scientific Research (CNRS) and Ribeirão Preto Medical School (FMRP) of the Sao Paulo University (USP), Sao Paulo, Brazil
| |
Collapse
|
34
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Overview of pyroptosis mechanism and in-depth analysis of cardiomyocyte pyroptosis mediated by NF-κB pathway in heart failure. Biomed Pharmacother 2024; 179:117367. [PMID: 39214011 DOI: 10.1016/j.biopha.2024.117367] [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: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The pyroptosis of cardiomyocytes has become an essential topic in heart failure research. The abnormal accumulation of these biological factors, including angiotensin II, advanced glycation end products, and various growth factors (such as connective tissue growth factor, vascular endothelial growth factor, transforming growth factor beta, among others), activates the nuclear factor-κB (NF-κB) signaling pathway in cardiovascular diseases, ultimately leading to pyroptosis of cardiomyocytes. Therefore, exploring the underlying molecular biological mechanisms is essential for developing novel drugs and therapeutic strategies. However, our current understanding of the precise regulatory mechanism of this complex signaling pathway in cardiomyocyte pyroptosis is still limited. Given this, this study reviews the milestone discoveries in the field of pyroptosis research since 1986, analyzes in detail the similarities, differences, and interactions between pyroptosis and other cell death modes (such as apoptosis, necroptosis, autophagy, and ferroptosis), and explores the deep connection between pyroptosis and heart failure. At the same time, it depicts in detail the complete pathway of the activation, transmission, and eventual cardiomyocyte pyroptosis of the NF-κB signaling pathway in the process of heart failure. In addition, the study also systematically summarizes various therapeutic approaches that can inhibit NF-κB to reduce cardiomyocyte pyroptosis, including drugs, natural compounds, small molecule inhibitors, gene editing, and other cutting-edge technologies, aiming to provide solid scientific support and new research perspectives for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
35
|
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
|
36
|
Li Y, Tian L, Li S, Chen X, Lei F, Bao J, Wu Q, Wen Y, Jie Y. Disrupted mitochondrial transcription factor A expression promotes mitochondrial dysfunction and enhances ocular surface inflammation by activating the absent in melanoma 2 inflammasome. Free Radic Biol Med 2024; 222:106-121. [PMID: 38797339 DOI: 10.1016/j.freeradbiomed.2024.05.032] [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: 03/28/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
PURPOSE Severe dry eye disease causes ocular surface damage, which is highly associated with mitochondrial dysfunction. Mitochondrial transcription factor A (TFAM) is essential for packaging mitochondrial DNA (mtDNA) and is crucial for maintaining mitochondrial function. Herein, we aimed to explore the effect of a decreased TFAM expression on ocular surface damage. METHODS Female C57BL/6 mice were induced ocular surface injury by topical administrating benzalkonium chloride (BAC). Immortalized human corneal epithelial cells (HCECs) were stimulated by tert-butyl hydroperoxide (t-BHP) to create oxidative stress damage. HCECs with TFAM knockdown were established. RNA sequencing was employed to analyze the whole-genome expression. Mitochondrial changes were measured by transmission electron microscopy, Seahorse metabolic flux analysis, mitochondrial membrane potential, and mtDNA copy number. TFAM expression and inflammatory cytokines were determined using RT-qPCR, immunohistochemistry, immunofluorescence, and immunoblotting. RESULTS In both the corneas of BAC-treated mice and t-BHP-induced HCECs, we observed impaired TFAM expression, accompanied by mitochondrial structure and function defects. TFAM downregulation in HCECs suppressed mitochondrial respiratory capacity, reduced mtDNA content, induced mtDNA leakage into the cytoplasm, and led to inflammation. RNA sequencing revealed the absent in melanoma 2 (AIM2) inflammasome was activated in the corneas of BAC-treated mice. The AIM2 inflammasome activation was confirmed in TFAM knockdown HCECs. TFAM knockdown in t-BHP-stimulated HCECs aggravated mitochondrial dysfunction and the AIM2 inflammasome activation, thereby further triggering the secretion of inflammatory factors such as interleukin (IL) -1β and IL-18. CONCLUSIONS TFAM reduction impaired mitochondrial function, activated AIM2 inflammasome and promoted ocular surface inflammation, revealing an underlying molecular mechanism for ocular surface disorders.
Collapse
Affiliation(s)
- Yaqiong Li
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Lei Tian
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Siyuan Li
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Xiaoniao Chen
- Department of Ophthalmology, The Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China.
| | - Fengyang Lei
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Jiayu Bao
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Qianru Wu
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Ya Wen
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Ying Jie
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| |
Collapse
|
37
|
Wang X, Qian J, Li Y, Meng Y, Cheng R, Ren N, Fei Y. Protective effects of forsythoside A against severe acute pancreatitis- induced brain injury in mice. Biomed Pharmacother 2024; 178:117301. [PMID: 39137650 DOI: 10.1016/j.biopha.2024.117301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024] Open
Abstract
OBJECTIVES This study aimed to evaluate the therapeutic effects of forsythoside A (FA) on brain injury induced by severe acute pancreatitis (SAP) using a murine model. METHODS Mice were induced with 3.5 % sodium taurocholate to model SAP-induced brain injury (SAP-IBI) and were randomly assigned to four distinct treatment regimens: the SAP-IBI model group (SAP-IBI), low-dose FA treatment group (FA L+SI), middle-dose FA treatment group (FA M+SI), and high-dose FA treatment group (FA H+SI). A sham-operation group (SO) served as a negative control. Serum levels of interleukin-1β (IL-1β) and IL-18 were quantified via ELISA, and serum amylase levels were assessed using optical turbidimetry. mRNA expression levels of AIM2, ASC, Caspase-1, and GAPDH in hippocampal brain tissue were measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Protein levels of NLRP3, GSDMD, IL-1β, and IL-18 in hippocampal brain tissue were evaluated using Western blotting. Neurological function in surviving mice was assessed through modified neurological severity scores (mNSS). Transmission electron microscopy (TEM) provided ultrastructural analysis of the hippocampus. Additionally, water content and pathological changes in hippocampal brain tissue were examined 24 hours post-operation, along with other relevant indicators. RESULTS At 24 hours post-operation, the FA H+SI group exhibited significantly reduced levels of serum amylase, IL-1β, and IL-18, along with decreased expression of AIM2, ASC, and Caspase-1 mRNA. Furthermore, NLRP3 protein levels, water content, pancreas and hippocampal brain pathological scores, and mNSS were significantly lower compared to the SAP-IBI group (P<0.01). CONCLUSIONS FA demonstrates protective effects against SAP-IBI in mice, suggesting potential therapeutic benefits.
Collapse
Affiliation(s)
- Xiaohong Wang
- Department of Gastroenterology, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China.
| | - Jing Qian
- Department of General Surgery, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China
| | - Yanjie Li
- Department of Neurology, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China
| | - Yun Meng
- Department of Gastroenterology, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China
| | - Ruizhi Cheng
- Department of Gastroenterology, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China
| | - Nana Ren
- Department of Gastroenterology, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China
| | - Ying Fei
- Department of Gastroenterology, Yizheng Hospital of Nanjing Drum Tower Hospital Group, Yizheng, Jiangsu 211900, China
| |
Collapse
|
38
|
Wu Q, Du J, Bae EJ, Choi Y. Pyroptosis in Skeleton Diseases: A Potential Therapeutic Target Based on Inflammatory Cell Death. Int J Mol Sci 2024; 25:9068. [PMID: 39201755 PMCID: PMC11354934 DOI: 10.3390/ijms25169068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Skeletal disorders, including fractures, osteoporosis, osteoarthritis, rheumatoid arthritis, and spinal degenerative conditions, along with associated spinal cord injuries, significantly impair daily life and impose a substantial burden. Many of these conditions are notably linked to inflammation, with some classified as inflammatory diseases. Pyroptosis, a newly recognized form of inflammatory cell death, is primarily triggered by inflammasomes and executed by caspases, leading to inflammation and cell death through gasdermin proteins. Emerging research underscores the pivotal role of pyroptosis in skeletal disorders. This review explores the pyroptosis signaling pathways and their involvement in skeletal diseases, the modulation of pyroptosis by other signals in these conditions, and the current evidence supporting the therapeutic potential of targeting pyroptosis in treating skeletal disorders, aiming to offer novel insights for their management.
Collapse
Affiliation(s)
- Qian Wu
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea (J.D.)
| | - Jiacheng Du
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea (J.D.)
| | - Eun Ju Bae
- School of Pharmacy, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Yunjung Choi
- Division of Rheumatology, Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| |
Collapse
|
39
|
Mu KL, Ran F, Peng LQ, Zhou LL, Wu YT, Shao MH, Chen XG, Guo CM, Luo QM, Wang TJ, Liu YC, Liu G. Identification of diagnostic biomarkers of rheumatoid arthritis based on machine learning-assisted comprehensive bioinformatics and its correlation with immune cells. Heliyon 2024; 10:e35511. [PMID: 39170142 PMCID: PMC11336745 DOI: 10.1016/j.heliyon.2024.e35511] [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: 01/11/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by inflammatory cell infiltration, which can lead to chronic disability, joint destruction and loss of function. At present, the pathogenesis of RA is still unclear. The purpose of this study is to explore the potential biomarkers and immune molecular mechanisms of rheumatoid arthritis through machine learning-assisted bioinformatics analysis, in order to provide reference for the early diagnosis and treatment of RA disease. Methods RA gene chips were screened from the public gene GEO database, and batch correction of different groups of RA gene chips was performed using Strawberry Perl. DEGs were obtained using the limma package of R software, and functional enrichment analysis such as gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), disease ontology (DO), and gene set (GSEA) were performed. Three machine learning methods, least absolute shrinkage and selection operator regression (LASSO), support vector machine recursive feature elimination (SVM-RFE) and random forest tree (Random Forest), were used to identify potential biomarkers of RA. The validation group data set was used to verify and further confirm its expression and diagnostic value. In addition, CIBERSORT algorithm was used to evaluate the infiltration of immune cells in RA and control samples, and the correlation between confirmed RA diagnostic biomarkers and immune cells was analyzed. Results Through feature screening, 79 key DEGs were obtained, mainly involving virus response, Parkinson's pathway, dermatitis and cell junction components. A total of 29 hub genes were screened by LASSO regression, 34 hub genes were screened by SVM-RFE, and 39 hub genes were screened by Random Forest. Combined with the three algorithms, a total of 12 hub genes were obtained. Through the expression and diagnostic value verification in the validation group data set, 7 genes that can be used as diagnostic biomarkers for RA were preliminarily confirmed. At the same time, the correlation analysis of immune cells found that γδT cells, CD4+ memory activated T cells, activated dendritic cells and other immune cells were positively correlated with multiple RA diagnostic biomarkers, CD4+ naive T cells, regulatory T cells and other immune cells were negatively correlated with multiple RA diagnostic biomarkers. Conclusions The results of novel characteristic gene analysis of RA showed that KYNU, EVI2A, CD52, C1QB, BATF, AIM2 and NDC80 had good diagnostic and clinical value for the diagnosis of RA, and were closely related to immune cells. Therefore, these seven DEGs may become new diagnostic markers and immunotherapy markers for RA.
Collapse
Affiliation(s)
| | | | - Le-qiang Peng
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Ling-li Zhou
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Yu-tong Wu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Ming-hui Shao
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Xiang-gui Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Chang-mao Guo
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Qiu-mei Luo
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Tian-jian Wang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Yu-chen Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Gang Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| |
Collapse
|
40
|
Liu D, Qin H, Gao Y, Sun M, Wang M. Cardiovascular disease: Mitochondrial dynamics and mitophagy crosstalk mechanisms with novel programmed cell death and macrophage polarisation. Pharmacol Res 2024; 206:107258. [PMID: 38909638 DOI: 10.1016/j.phrs.2024.107258] [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: 02/20/2024] [Revised: 06/08/2024] [Accepted: 06/08/2024] [Indexed: 06/25/2024]
Abstract
Several cardiovascular illnesses are associated with aberrant activation of cellular pyroptosis, ferroptosis, necroptosis, cuproptosis, disulfidptosis, and macrophage polarisation as hallmarks contributing to vascular damage and abnormal cardiac function. Meanwhile, these three novel forms of cellular dysfunction are closely related to mitochondrial homeostasis. Mitochondria are the main organelles that supply energy and maintain cellular homeostasis. Mitochondrial stability is maintained through a series of regulatory pathways, such as mitochondrial fission, mitochondrial fusion and mitophagy. Studies have shown that mitochondrial dysfunction (e.g., impaired mitochondrial dynamics and mitophagy) promotes ROS production, leading to oxidative stress, which induces cellular pyroptosis, ferroptosis, necroptosis, cuproptosis, disulfidptosis and macrophage M1 phenotypic polarisation. Therefore, an in-depth knowledge of the dynamic regulation of mitochondria during cellular pyroptosis, ferroptosis, necroptosis, cuproptosis, disulfidptosis and macrophage polarisation is necessary to understand cardiovascular disease development. This paper systematically summarises the impact of changes in mitochondrial dynamics and mitophagy on regulating novel cellular dysfunctions and macrophage polarisation to promote an in-depth understanding of the pathogenesis of cardiovascular diseases and provide corresponding theoretical references for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Dandan Liu
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Hewei Qin
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China; Department of Rehabilitation Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 450002, China.
| | - Yang Gao
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Mengyan Sun
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Mengnan Wang
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| |
Collapse
|
41
|
Wang Y, Fang N, Wang Y, Geng Y, Li Y. Activating MC4R Promotes Functional Recovery by Repressing Oxidative Stress-Mediated AIM2 Activation Post-spinal Cord Injury. Mol Neurobiol 2024; 61:6101-6118. [PMID: 38277117 DOI: 10.1007/s12035-024-03936-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Abstract
Spinal cord injury (SCI) is a destructive neurological trauma that induces permanent sensory and motor impairment as well as a deficit in autonomic physiological function. Melanocortin receptor 4 (MC4R) is a G protein-linked receptor that is extensively expressed in the neural system and contributes to inhibiting inflammation, regulating mitochondrial function, and inducing programmed cell death. However, the effect of MC4R in the modulation of oxidative stress and whether this mechanism is related to the role of absent in melanoma 2 (AIM2) in SCI are not confirmed yet. In the current study, we demonstrated that MC4R is significantly increased in the neurons of spinal cords after trauma and oxidative stimulation of cells. Further, activation of MC4R by RO27-3225 effectively improved functional recovery, inhibited AIM2 activation, maintained mitochondrial homeostasis, repressed oxidative stress, and prevented Drp1 translocation to the mitochondria. Meanwhile, treating Drp1 inhibitors would be beneficial in reducing AIM2 activation, and activating AIM2 could abolish the protective effect of MC4R on neuron homeostasis. In conclusion, we demonstrated that MC4R protects against neural injury through a novel process by inhibiting mitochondrial dysfunction, oxidative stress, as well as AIM2 activation, which may serve as an available candidate for SCI therapy.
Collapse
Affiliation(s)
- Yongli Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Orthopaedics, Huzhou Central Hospital, Huzhou Basic and Clinical Translation of Orthopaedics Key Laboratory, Huzhou, Zhejiang, China
| | - Nongtao Fang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yikang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yibo Geng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yao Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
42
|
Liu X, Zhou Q, Meng J, Zuo H, Li R, Zhang R, Lu H, Zhang Z, Li H, Zeng S, Tian M, Wang H, Hu K, Li N, Mao L, Hou S. Autophagy-mediated activation of the AIM2 inflammasome enhances M1 polarization of microglia and exacerbates retinal neovascularization. MedComm (Beijing) 2024; 5:e668. [PMID: 39081514 PMCID: PMC11286542 DOI: 10.1002/mco2.668] [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: 02/16/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a retinal neovascularization (RNV) disease that is characterized by abnormal blood vessel development in the retina. Importantly, the etiology of ROP remains understudied. We re-analyzed previously published single-cell data and discovered a strong correlation between microglia and RNV diseases, particularly ROP. Subsequently, we found that reactive oxygen species reduced autophagy-dependent protein degradation of absent in melanoma 2 (AIM2) in hypoxic BV2 cells, leading to increased AIM2 protein accumulation. Furthermore, we engineered AIM2 knockout mice and observed that the RNV was significantly reduced compared to wild-type mice. In vitro vascular function assays also demonstrated diminished angiogenic capabilities following AIM2 knockdown in hypoxic BV2 cells. Mechanistically, AIM2 enhanced the M1-type polarization of microglia via the ASC/CASP1/IL-1β pathway, resulting in RNV. Notably, the administration of recombinant protein IL-1β exacerbated angiogenesis, while its inhibition ameliorated the condition. Taken together, our study provides a novel therapeutic target for ROP and offers insight into the interaction between pyroptosis and autophagy.
Collapse
Affiliation(s)
- Xianyang Liu
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Qian Zhou
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Jiayu Meng
- Sichuan Provincial Key Laboratory for Human Disease Gene StudySichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Hangjia Zuo
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Ruonan Li
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Rui Zhang
- Department of OphthalmologyQilu HospitalCheeloo College of MedicineShandong UniversityJinanChina
| | - Huiping Lu
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Zhi Zhang
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Hongshun Li
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Shuhao Zeng
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Meng Tian
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijing Tongren HospitalCapital Medical UniversityBeijingChina
| | - Hong Wang
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijing Tongren HospitalCapital Medical UniversityBeijingChina
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of OphthalmologyChongqingChina
| | - Na Li
- Department of Laboratory Medicine, Beijing Tongren HospitalCapital Medical UniversityBeijingChina
| | - Liming Mao
- Department of ImmunologySchool of MedicineNantong UniversityNantongChina
| | - Shengping Hou
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijing Tongren HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
43
|
Cebani L, Mvubu NE. Can We Exploit Inflammasomes for Host-Directed Therapy in the Fight against Mycobacterium tuberculosis Infection? Int J Mol Sci 2024; 25:8196. [PMID: 39125766 PMCID: PMC11311975 DOI: 10.3390/ijms25158196] [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: 06/18/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (M. tb), is a major global health issue, with around 10 million new cases annually. Advances in TB immunology have improved our understanding of host signaling pathways, leading to innovative therapeutic strategies. Inflammasomes, protein complexes organized by cytosolic pattern recognition receptors (PRRs), play a crucial role in the immune response to M. tb by activating caspase 1, which matures proinflammatory cytokines IL1β and IL18. While inflammation is necessary to fight infection, excessive or dysregulated inflammation can cause tissue damage, highlighting the need for precise inflammasome regulation. Drug-resistant TB strains have spurred research into adjunctive host-directed therapies (HDTs) that target inflammasome pathways to control inflammation. Canonical and non-canonical inflammasome pathways can trigger excessive inflammation, leading to immune system exhaustion and M. tb spread. Novel HDT interventions can leverage precision medicine by tailoring treatments to individual inflammasome responses. Studies show that medicinal plant derivatives like silybin, andrographolide, and micheliolide and small molecules such as OLT1177, INF39, CY-09, JJ002, Ac-YVAD-cmk, TAK-242, and MCC950 can modulate inflammasome activation. Molecular tools like gene silencing and knockouts may also be used for severe TB cases. This review explores these strategies as potential adjunctive HDTs in fighting TB.
Collapse
Affiliation(s)
| | - Nontobeko E. Mvubu
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa;
| |
Collapse
|
44
|
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
|
45
|
Jiang Q, Chen Y, Zheng S, Sui L, Yu D, Qing F, He W, Xiao Q, Guo T, Xu L, Liu Z, Liu Z. AIM2 enhances Candida albicans infection through promoting macrophage apoptosis via AKT signaling. Cell Mol Life Sci 2024; 81:280. [PMID: 38918243 PMCID: PMC11335202 DOI: 10.1007/s00018-024-05326-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024]
Abstract
Candida albicans is among the most prevalent invasive fungal pathogens for immunocompromised individuals and novel therapeutic approaches that involve immune response modulation are imperative. Absent in melanoma 2 (AIM2), a pattern recognition receptor for DNA sensing, is well recognized for its involvement in inflammasome formation and its crucial role in safeguarding the host against various pathogenic infections. However, the role of AIM2 in host defense against C. albicans infection remains uncertain. This study reveals that the gene expression of AIM2 is induced in human and mouse innate immune cells or tissues after C. albicans infection. Furthermore, compared to their wild-type (WT) counterparts, Aim2-/- mice surprisingly exhibit resistance to C. albicans infection, along with reduced inflammation in the kidneys post-infection. The resistance of Aim2-/- mice to C. albicans infection is not reliant on inflammasome or type I interferon production. Instead, Aim2-/- mice display lower levels of apoptosis in kidney tissues following infection than WT mice. The deficiency of AIM2 in macrophages, but not in dendritic cells, results in a phenocopy of the resistance observed in Aim2-/- mice against C. albican infection. The treatment of Clodronate Liposome, a reagent that depletes macrophages, also shows the critical role of macrophages in host defense against C. albican infection in Aim2-/- mice. Furthermore, the reduction in apoptosis is observed in Aim2-/- mouse macrophages following infection or treatment of DNA from C. albicans in comparison with controls. Additionally, higher levels of AKT activation are observed in Aim2-/- mice, and treatment with an AKT inhibitor reverses the host resistance to C. albicans infection. The findings collectively demonstrate that AIM2 exerts a negative regulatory effect on AKT activation and enhances macrophage apoptosis, ultimately compromising host defense against C. albicans infection. This suggests that AIM2 and AKT may represent promising therapeutic targets for the management of fungal infections.
Collapse
Affiliation(s)
- Qian Jiang
- School of Graduate, China Medical University, Shenyang, Liaoning, China
- School of Nursing, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yayun Chen
- School of Graduate, China Medical University, Shenyang, Liaoning, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Siping Zheng
- School of Graduate, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Lina Sui
- School of Graduate, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Dalang Yu
- School of Graduate, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Furong Qing
- School of Graduate, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Wenji He
- School of Graduate, China Medical University, Shenyang, Liaoning, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Qiuxiang Xiao
- School of Graduate, China Medical University, Shenyang, Liaoning, China
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Tianfu Guo
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Li Xu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
- Center for Scientific Research, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zhichun Liu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China.
| | - Zhiping Liu
- School of Graduate, China Medical University, Shenyang, Liaoning, China.
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China.
- Center for Scientific Research, Gannan Medical University, Ganzhou, Jiangxi, China.
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China.
| |
Collapse
|
46
|
Lin J, Wang J, Fang J, Li M, Xu S, Little PJ, Zhang D, Liu Z. The cytoplasmic sensor, the AIM2 inflammasome: A precise therapeutic target in vascular and metabolic diseases. Br J Pharmacol 2024; 181:1695-1719. [PMID: 38528718 DOI: 10.1111/bph.16355] [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: 06/14/2023] [Revised: 01/02/2024] [Accepted: 02/12/2024] [Indexed: 03/27/2024] Open
Abstract
Cardio-cerebrovascular diseases encompass pathological changes in the heart, brain and vascular system, which pose a great threat to health and well-being worldwide. Moreover, metabolic diseases contribute to and exacerbate the impact of vascular diseases. Inflammation is a complex process that protects against noxious stimuli but is also dysregulated in numerous so-called inflammatory diseases, one of which is atherosclerosis. Inflammation involves multiple organ systems and a complex cascade of molecular and cellular events. Numerous studies have shown that inflammation plays a vital role in cardio-cerebrovascular diseases and metabolic diseases. The absent in melanoma 2 (AIM2) inflammasome detects and is subsequently activated by double-stranded DNA in damaged cells and pathogens. With the assistance of the mature effector molecule caspase-1, the AIM2 inflammasome performs crucial biological functions that underpin its involvement in cardio-cerebrovascular diseases and related metabolic diseases: The production of interleukin-1 beta (IL-1β), interleukin-18 (IL-18) and N-terminal pore-forming Gasdermin D fragment (GSDMD-N) mediates a series of inflammatory responses and programmed cell death (pyroptosis and PANoptosis). Currently, several agents have been reported to inhibit the activity of the AIM2 inflammasome and have the potential to be evaluated for use in clinical settings. In this review, we systemically elucidate the assembly, biological functions, regulation and mechanisms of the AIM2 inflammasome in cardio-cerebrovascular diseases and related metabolic diseases and outline the inhibitory agents of the AIM2 inflammasome as potential therapeutic drugs.
Collapse
Affiliation(s)
- Jiuguo Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou, China
| | - Jiaojiao Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou, China
| | - Jian Fang
- Huadu District People's Hospital of Guangzhou, Guangzhou, China
| | - Meihang Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou, China
| | - Suowen Xu
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Peter J Little
- Pharmacy Australia Centre of Excellence, School of Pharmacy, University of Queensland, Woolloongabba, Queensland, Australia
| | - Dongmei Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou, China
| |
Collapse
|
47
|
Yu L, Gao F, Li Y, Su D, Han L, Li Y, Zhang X, Feng Z. Role of pattern recognition receptors in the development of MASLD and potential therapeutic applications. Biomed Pharmacother 2024; 175:116724. [PMID: 38761424 DOI: 10.1016/j.biopha.2024.116724] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 05/20/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has become one of the most prevalent liver diseases worldwide, and its occurrence is strongly associated with obesity, insulin resistance (IR), genetics, and metabolic stress. Ranging from simple fatty liver to metabolic dysfunction-associated steatohepatitis (MASH), even to severe complications such as liver fibrosis and advanced cirrhosis or hepatocellular carcinoma, the underlying mechanisms of MASLD progression are complex and involve multiple cellular mediators and related signaling pathways. Pattern recognition receptors (PRRs) from the innate immune system, including Toll-like receptors (TLRs), C-type lectin receptors (CLRs), NOD-like receptors (NLRs), RIG-like receptors (RLRs), and DNA receptors, have been demonstrated to potentially contribute to the pathogenesis for MASLD. Their signaling pathways can induce inflammation, mediate oxidative stress, and affect the gut microbiota balance, ultimately resulting in hepatic steatosis, inflammatory injury and fibrosis. Here we review the available literature regarding the involvement of PRR-associated signals in the pathogenic and clinical features of MASLD, in vitro and in animal models of MASLD. We also discuss the emerging targets from PRRs for drug developments that involved agent therapies intended to arrest or reverse disease progression, thus enabling the refinement of therapeutic targets that can accelerate drug development.
Collapse
Affiliation(s)
- Lili Yu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Feifei Gao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Yaoxin Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Dan Su
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Liping Han
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yueming Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Xuehan Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Zhiwei Feng
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China.
| |
Collapse
|
48
|
Karami A, Skorski T. DNA damage response genes as biomarkers of therapeutic outcomes in acute myeloid leukemia patients. Leukemia 2024; 38:1407-1410. [PMID: 38734788 PMCID: PMC11147752 DOI: 10.1038/s41375-024-02269-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024]
Affiliation(s)
- Adam Karami
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Tomasz Skorski
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
49
|
Zheng Z, Li K, Yang Z, Wang X, Shen C, Zhang Y, Lu H, Yin Z, Sha M, Ye J, Zhu L. Transcriptomic analysis reveals molecular characterization and immune landscape of PANoptosis-related genes in atherosclerosis. Inflamm Res 2024; 73:961-978. [PMID: 38587531 DOI: 10.1007/s00011-024-01877-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/01/2024] [Accepted: 03/27/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease characterized by abnormal lipid deposition in the arteries. Programmed cell death is involved in the inflammatory response of atherosclerosis, but PANoptosis, as a new form of programmed cell death, is still unclear in atherosclerosis. This study explored the key PANoptosis-related genes involved in atherosclerosis and their potential mechanisms through bioinformatics analysis. METHODS We evaluated differentially expressed genes (DEGs) and immune infiltration landscape in atherosclerosis using microarray datasets and bioinformatics analysis. By intersecting PANoptosis-related genes from the GeneCards database with DEGs, we obtained a set of PANoptosis-related genes in atherosclerosis (PANoDEGs). Functional enrichment analysis of PANoDEGs was performed and protein-protein interaction (PPI) network of PANoDEGs was established. The machine learning algorithms were used to identify the key PANoDEGs closely linked to atherosclerosis. Receiver operating characteristic (ROC) analysis was used to assess the diagnostic potency of key PANoDEGs. CIBERSORT was used to analyze the immune infiltration patterns in atherosclerosis, and the Spearman method was used to study the relationship between key PANoDEGs and immune infiltration abundance. The single gene enrichment analysis of key PANoDEGs was investigated by GSEA. The transcription factors and target miRNAs of key PANoDEGs were predicted by Cytoscape and online database, respectively. The expression of key PANoDEGs was validated through animal and cell experiments. RESULTS PANoDEGs in atherosclerosis were significantly enriched in apoptotic process, pyroptosis, necroptosis, cytosolic DNA-sensing pathway, NOD-like receptor signaling pathway, lipid and atherosclerosis. Four key PANoDEGs (ZBP1, SNHG6, DNM1L, and AIM2) were found to be closely related to atherosclerosis. The ROC curve analysis demonstrated that the key PANoDEGs had a strong diagnostic potential in distinguishing atherosclerotic samples from control samples. Immune cell infiltration analysis revealed that the proportion of initial B cells, plasma cells, CD4 memory resting T cells, and M1 macrophages was significantly higher in atherosclerotic tissues compared to normal tissues. Spearman analysis showed that key PANoDEGs showed strong correlations with immune cells such as T cells, macrophages, plasma cells, and mast cells. The regulatory networks of the four key PANoDEGs were established. The expression of key PANoDEGs was verified in further cell and animal experiments. CONCLUSIONS This study evaluated the expression changes of PANoptosis-related genes in atherosclerosis, providing a reference direction for the study of PANoptosis in atherosclerosis and offering potential new avenues for further understanding the pathogenesis and treatment strategies of atherosclerosis.
Collapse
Affiliation(s)
| | - Kaiyuan Li
- Dalian Medical University, Dalian, 116000, China
| | - Zhiyuan Yang
- Dalian Medical University, Dalian, 116000, China
| | - Xiaowen Wang
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cheng Shen
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yubin Zhang
- Dalian Medical University, Dalian, 116000, China
| | - Huimin Lu
- Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, 225399, China
| | - Zhifeng Yin
- Jiangsu Hanjiang Biotechnology Co., LTD, Taizhou, 225399, China
| | - Min Sha
- Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, 225399, China.
| | - Jun Ye
- Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, 225399, China.
| | - Li Zhu
- Dalian Medical University, Dalian, 116000, China.
- Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, 225399, China.
| |
Collapse
|
50
|
Valdés-López JF, Hernández-Sarmiento LJ, Tamayo-Molina YS, Velilla-Hernández PA, Rodenhuis-Zybert IA, Urcuqui-Inchima S. Interleukin 27, like interferons, activates JAK-STAT signaling and promotes pro-inflammatory and antiviral states that interfere with dengue and chikungunya viruses replication in human macrophages. Front Immunol 2024; 15:1385473. [PMID: 38720890 PMCID: PMC11076713 DOI: 10.3389/fimmu.2024.1385473] [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: 02/12/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
Interferons (IFNs) are a family of cytokines that activate the JAK-STAT signaling pathway to induce an antiviral state in cells. Interleukin 27 (IL-27) is a member of the IL-6 and/or IL-12 family that elicits both pro- and anti-inflammatory responses. Recent studies have reported that IL-27 also induces a robust antiviral response against diverse viruses, both in vitro and in vivo, suggesting that IFNs and IL-27 share many similarities at the functional level. However, it is still unknown how similar or different IFN- and IL-27-dependent signaling pathways are. To address this question, we conducted a comparative analysis of the transcriptomic profiles of human monocyte-derived macrophages (MDMs) exposed to IL-27 and those exposed to recombinant human IFN-α, IFN-γ, and IFN-λ. We utilized bioinformatics approaches to identify common differentially expressed genes between the different transcriptomes. To verify the accuracy of this approach, we used RT-qPCR, ELISA, flow cytometry, and microarrays data. We found that IFNs and IL-27 induce transcriptional changes in several genes, including those involved in JAK-STAT signaling, and induce shared pro-inflammatory and antiviral pathways in MDMs, leading to the common and unique expression of inflammatory factors and IFN-stimulated genes (ISGs)Importantly, the ability of IL-27 to induce those responses is independent of IFN induction and cellular lineage. Additionally, functional analysis demonstrated that like IFNs, IL-27-mediated response reduced chikungunya and dengue viruses replication in MDMs. In summary, IL-27 exhibits properties similar to those of all three types of human IFN, including the ability to stimulate a protective antiviral response. Given this similarity, we propose that IL-27 could be classified as a distinct type of IFN, possibly categorized as IFN-pi (IFN-π), the type V IFN (IFN-V).
Collapse
Affiliation(s)
- Juan Felipe Valdés-López
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | | | - Y. S. Tamayo-Molina
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | | | - Izabela A. Rodenhuis-Zybert
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| |
Collapse
|